Вы находитесь на странице: 1из 533

The Foundations of Virology

Discoverers and Discoveries


Inventors and Inventions
Developers and Technologies

Frederick A. Murphy
The Foundations of Virology
Discoverers and Discoveries
Inventors and Inventions
Developers and Technologies

Frederick A. Murphy

University of Texas Medical Branch


Galveston, Texas, USA

2013
The Foundations of Virology
Discoverers and Discoveries, Inventors and Inventions, Developers and Technologies

Copyright 2011 by Frederick A. Murphy

Key references pertaining to discovery, development and innovation are provided on each page, using the style of PubMed. A Further Reading list is
included at the end of the book.

The images contained in this book were obtained from (1) my own image collection, (2) image collections of colleagues, (3) public Internet sites, and
(4) private image archives and library collections (each with permission, cited with the image). It was found that sources cited on public Internet sites
were very often inaccurate, or secondary, tertiary, in daisy-chain fashion, never getting to the proper original source. Many of my images (mostly
electron micrographs) found on Internet sites suffer the same failings wrong attribution, inaccurate legends, misleading colorizing and other
manipulations, etc. If I receive objections to the inclusion of any images, I will remove them.

This book is meant for educational purposes only.

This book goes with a PowerPoint slide set (~900 slides) which is on a website at the University of Texas Medical Branch / Galveston. The link to the
slides is on the second page of the website: http://www.utmb.edu/virusimages/

This book was composed using Adobe InDesign CS5 (.indd files). In this program all image files are linked; therefore, the highest resolution original
image files (.tif, .jpg) are only available directly from me I would be please to share them, using email or if files are too large, Dropbox or CDs/
DVDs. The camera-ready products of InDesign files are converted to special Adobe Acrobat files (.pdf ), small file and press quality versions of
which are also available for online and hard-copy printing. This book was composed using the Adobe Warnock Pro typeface set.

Frederick A Murphy
University of Texas Medical Branch
Department of Pathology, Route 0609
301 University Boulevard
Galveston, TX 77555-0609 USA INFINITY PUBLISHING
(Email) famurphy@utmb.edu 1094 New Dehaven Street, Suite 100
West Conshohocken, PA 19428-2713
First Published April 2012 / Revision July 2013 Toll Free (877) BUY BOOK
Local Phone (610) 941-9999
Printed in the United States of America Fax (610) 941-9959
Info@buybooksontheweb.com
ISBN 978-0-7414-7365-3 / 0-7414-7365-8 www.buybooksontheweb.com
Dedication

This work is dedicated to my beloved wife, Irene.


Foreword because they have always been part of the context in which human viruses
are studied, and because it is the zoonotic and species-jumping viruses
from which most of the new, emerging and re-emerging human pathogens
If I have seen further it is by standing on the shoulders of giants. originate. Further, in the era of the founding of virology there was exceptional
Isaac Newton in a letter to Robert Hooke, 1676 crossover between human and veterinary virology, much more than is seen
todaythere is value in being reminded of this. (2) The book is not limited to
A great many highly creative scientists take it quite for granted that an the type viruses of families and genera; rather it includes as many as possible
interest in the history of science is a sign of failing powers. The history of of the important and interesting pathogens that are the focus of medical and
science does not often interest the scientist as science. veterinary virology and comparative virology research. (3) In some instances I
Peter Medawar, 1968 have included several entries for a given virus, for example some from the era
when the classification of an infectious agent as a virus was based solely upon
The foundation of the medical and veterinary virology/viral disease its ultrafilterability, and some from the modern era, when the virus is defined
sciences predates the concept of the specificity of disease causation and by more definitive methods. (4) The discovery/development of the great
is heavily dependent upon initial discoveries about bacteria and bacterial attenuated viruses used in vaccines is included, but in condensed forma
diseases. Upon a broad and venerable foundation, Louis Pasteur established comprehensive listing here would have to include the many independently
the microbiologic/virologic/infectious disease sciences, first in 1857 by derived vaccines and substrates, the subject for another book. (5) The
discovering the specificity of microbial fermentations (wine, beer, cheese), arboviruses and hemorrhagic fever viruses proved to be a special case because
then in 1865 by extending the concept to infectious diseases of silkworms, of their profusion, so a selective approach was taken in order to emphasize
and finally between 1877 and 1895 by extending the concept to human and those viruses causing major human and veterinary diseases and the initial
animal diseases. His early infectious disease work centered on septic war discoveries of viruses representing major groupsit will be necessary for
wounds; he then turned to anthrax and other bacterial diseases, and lastly to someone to produce a separate book to give due credit to all the deserving
the viral disease rabies. In each instance, he moved quickly from studies aimed discoverers, inventors and developers in this field. (6) The attempt to include
at discovering the causative agent to the development of specific intervention. inventors and their inventions, and developers and their technologies proved
In 1885, Pasteur gave the first rabies vaccine to a boy, Joseph Meister, who especially difficultthe crucial role of these in the advance of the virology/
had been bitten severely by a rabid dogthat day marked the opening of the viral disease sciences is acknowledged, but in many cases breakthroughs
modern era of infectious disease science aimed at disease prevention and derive to commercial corporations rather than individuals and credit is
control. Pasteur was joined by Robert Koch, who discovered the causative difficult to attribute. (7) The book may fail in not capturing all the excitement,
agents of tuberculosis and cholera and contributed much to the development the romance, the Eureka, behind each entryonly a full article, or in some
of laboratory methods in bacteriology. Koch also worked on several diseases cases a full book, on each discovery, invention and development would do
which others eventually showed were caused by viruses. As a result of the justice to the story behind each entry. (8) The book must be seen as a work-
work of Pasteur, Koch and others, the identification of the causative agents of in-progressit is meant to be used, to be modified, and changed to meet
many important human and domestic animal diseases proceeded at breakneck various purposessuch is the power of electronic publishing. Toward this
pace around the turn of the twentieth century. end, a PowerPoint slide set containing the images, but little text is on my
websitethe ease in updating the slides will facilitate the production of future
From the foundation laid by Pasteur, Koch and their colleagues, others editions of this book. (9) The book inevitably contains errorsthese are my
extended the breadth and depth of the infectious disease sciences in many responsibility. I will collect communications about such errors for correction
ways. There are many names to be remembered. This book is an attempt to of the next edition.
remember some of the discoverers and discoveries, inventors and inventions,
and developers and technologies that were seminal in the advance of medical
and veterinary virology. The content is completely arbitrary, based only on my
decision as to what to include and what to leave out. Several factors guided
this decision: (1) The book is not limited simply to human pathogenic viruses Frederick A. Murphy
per se; many important veterinary and zoonotic pathogens are included 2012
Many colleagues responded to my inquiries about this project, in every case with incredible enthusiasmit must
be that this subject, the founding of virology and the viral disease sciences, really touches a common chord. The
contributions of the following people are gratefully acknowledged:

Arstila, Pertti Gonzalez, Jean-Paul Mercurio, Jennifer Skehel, John


Barin, Francis Grafman, Eric Mettenleiter, Thomas Smith, Jean
Beaty, Barry Greenberg, Harry Mims, Cedric Smith, Ralph
Berger, Ronald Greifenstein, Charles Mocarski, Ed Smith, Roger
Berns, Kenneth Haddow, Andrew Monath, Thomas Sderqvist, Thomas
Bettinger, George Haire, Kevlin Morens, David Spamer, Earle
Biggs, Peter Hardy, Anne Morgan, Robin Staeheli, Peter
Bittle, James Heilman, Carole Moussatch, Nissin Stapleton, Darwin
Blair, Carol Hirsch, Martin Nathanson, Neal Steck, Marianne
Bloom, Marshall Holland, John Neumann-Haefelin, Dieter Studdert, Michael
Bollas, Kristin Horzinek, Marian Nimtz, Kelly Sudia, Daniel
Boschung, Urs Ivanova, Olga Norrby, Erling Talasek, J.D.
Boulton, Clare Jeggo, Martyn Okamoto, Hiroaki Taylhardat, Leonardo
Buchmeier, Michael Johnson, Karl Oldstone, Michael Teloh, Mary
Burk, Janet Joklik, Bill Olsen, Anders Terquem, Micheline
Burrell, Christopher Jozan, Martine Osterrieder, Klaus Tesh, Robert
Calisher, Charles Kaplan, Bruce Palmarini, Massimo Thiel, Heinz-Juergen
Carmichael, Leland Klenk, Hans-Dieter Palmenberg, Ann Timoney, Peter
, . Knowles, James Parrish, Colin Toma, Bernard
Coimbra, Terezinha L.M. Knowles, Wendy Peters, C.J. Torres, Alfonso
Compans, Richard Ksiazek, Thomas Philpott, Howard Traum, Jeffrey
Coto, Celia Kurata, Takeshi Popov, Vsevolod van der Groen, Guido
Coulson, Barbara Laemmli, Ulrich Preisler, Don van Regenmortel, Marc
Cox, Francis Larghi, Oscar Quiroz, Baudilio Vasilakis, Nikolaos
Degueurce, Christophe Lefkowitz, Elliot Rizzetto, Mario Villarreal, Luis
DeMartini, James Lipton, Howard Rose, Kenneth Wandeler, Alex
Dermody, Terence Luciw, Paul Roveland-Brenton, Blythe Weaver, Scott
Diers, Bailey MacFarlane, Ross Rowlands, David Webster, Leslie III
Estes, Mary Mackenzie, Jan Rupprecht, Charles Webster, Rob
Fagan, Kelvin Mackenzie, John Rybicki, Edward Wimmer, Eckard
Fenner, Frank MacLachlan, Jim Sabattini, Marta Witkowski, Jan
French, Gary Madeley, Richard Sawicki, Dorothea Woodall, John
Gaza, Judy Madin, Larry Schilgerius, Silvia Wooley, Dawn
Gelb, Jack Mahieux, Renaud Schmaljohn, Connie Wunner, William
Gelderblom, Hans Mahy, Brian Schreiber, Tiffany Monet Wyllie, Andrew
Gerin, John Mialot, Jean-Paul Shadrin, Olga Yamanouchi, Kazuya
Gibbs, Adrian Mcleod, William Shatkin, Aaron Yanagihara, Ric
Goldberg, Michael McMinn, Peter Sipe, Jean Zaki, Sherif
Table of Contents 1898 Henry Rose Carter discovery of yellow fever virus mosquito extrinsic incubation period
1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus)
67
68
1900 John McFadyean discovery of African horse sickness virus (the first orbivirus) 75
TITLE PAGE 1900 Hugo de Vries founding of genetics, rediscovery of Mendels work 76
DEDICATION 1901 John D. Rockefeller Sr. founding of the Rockefeller Institute for Medical Research 77
FOREWORD 1901 Eugenio Centanni, others discovery of fowl plague virus (the first orthomyxovirus) 79
ACKNOWLEDGMENTS 1902 Aladr Aujeszky discovery of pseudorabies virus (the first herpesvirus) 80
TABLE OF CONTENTS 1902 Amde Borrel discovery of sheeppox virus 81
1902 Maurice Nicolle, Mustafa Adil Bey discovery of rinderpest virus (the first morbillivirus) 82
DATE DISCOVERER/DEVELOPER/INVENTOR DISCOVERY/DEVELOPMENT/INVENTION 1902 James Spreull, Arnold Theiler discovery of bluetongue viruses 83
1903 Emil De Schweinitz, Marion Dorset classical swine fever virus (the first pestivirus) 84
~400BCE Hippocrates father of medicine, important epidemiologic observations 6 1903 Adelchi Negri discovery of the rabies inclusion bodythe Negri body 85
1224 Mesopotamian medicine rabies, the oldest documented disease of humans 7 1903 Paul Remlinger, Riffat-Bay, Alfonso di Vestea discovery of rabies virus (first rhabdovirus) 87
1546 Girolamo Fracastoro proposal that epidemic diseases are contagious, first germ theory 8 1903 Arne Tiselius development of electrophoresis 89
1595 Sacharias & Hans Jansen development of the first compound microscope 9 1904 Henri Valle, Henri Carr discovery of equine infectious anemia virus (the first retrovirus) 90
1665 Robert Hooke first microscopic observation of a microorganism, a mold, genus Mucor 10 1904 William Gorgas control of yellow fever, allowing construction of the Panama Canal 91
1668 Anton van Leeuwenhoek invention of (single lens) microscope, discovery of bacteria 11 1905 Henri Carr, Patrick Laidlaw, George Dunkin discovery of canine distemper virus 94
1718 Lady Mary Wortley Montagu introduction of variolation to western countries 12 1905 Adelchi Negri, Paul Remlinger, others proof that variola and vaccinia are viruses 95
1735 Carolus Linnaeus development of hierarchical system for classification of all organisms 13 1907 Ross Harrison development of first cell cultures (nerve fibers from frog embryo tissues) 96
1765 Lazzaro Spallanzani first experiments refuting the theory of spontaneous generation 14 1907 Percy Ashburn Charles Craig discovery of dengue viruses 97
1774 Benjamin Jesty vaccination against smallpox using material from cowpox lesions 15 1907 Giuseppe Ciuffo discovery of human papillomaviruses (the first papillomaviruses) 98
1789 Edward Jenner vaccination against smallpox using material from cowpox lesions 16 1908 Vilhelm Ellerman, Oluf Bang discovery of avian leukemia virus (the first leukemia virus) 99
1793 John Hunter, Pierre-Victor Galtier, Georg Zinke, others experimental pathology 19 1909 Karl Landsteiner, Erwin Popper discovery of polioviruses (the first enteroviruses) 100
1793 Benjamin Rush account of the great yellow fever epidemic in Philadelphia, 1793 21 1909 Constantin Levaditi, Karl Landsteiner poliovirus in non-nervous system tissues 101
1798 Founding of the U.S. Public Health Service, first as the Marine Hospital Service 23 1909 Robert Doerr, K. Franz, S. Taussig discovery of sandfly fever viruses (first phleboviruses) 102
1805 John Pintard, others founding of New York City Department of Health, first in the U.S. 24 1910 Thomas Hunt Morgan discovery of the nature and role of the chromosome 103
1830 John Dollond, Ernst Abbe, others advances in microscopes, achromatic lens, condenser 25 1911 John Anderson, Joseph Goldberger discovery of measles virus 104
1835 Charles Chevalier, others development of microtome, key to cellular biology/pathology 26 1911 Peyton Rous, Joseph Beard discovery of Rous sarcoma virus (first solid tumor virus) 106
1835 Theodor Schwann, Matthias Schleiden concept that organisms are composed of cells 27 1911 George de Hevesy, others development of radioisotopic labeling 107
1838 John Snow, John Graunt, William Farr, others founding of medical epidemiology 28 1912 Alexis Carrel, others development of many cell and tissue culture methods 108
1840 Friedrich Gustav Jakob Henle father of the germ theory, and the Henle-Koch Postulates 29 1913 Edna Steinhardt, Clara Israeli, Robert Lambert first cultivation of a virus in cell culture 111
1841 William Henderson discovery of the inclusion body of molluscum contagiosum virus 30 1914 Alfred Hess, Y Hiro, S Tasaka discovery of rubella virus (the only rubivirus) 113
1843 Ignaz Semmelweis, Oliver Wendell Holmes, Joseph Lister hygiene/disinfection 31 1916 Development of trypsinization for the dispersal of cells in culture 114
1857 Louis Pasteur father of microbiology, father of virology 33 1916 Frederick Twort, Flix dHerelle discovery of bacteriophages 115
1858 Rudolf Virchow father of cellular (microscopic) pathology and comparative pathology 35 1917 Robert Montgomery Nairobi sheep disease virus (first nairovirus, first tick-borne virus) 116
1859 Charles Darwin father of evolutionary science: common descent, natural selection 36 1918 Influenza pandemic, 40-100 million deaths worldwide 117
1863 Abraham Lincoln, others founding of the U.S. National Academy of Sciences 39 1918 Charles Nicolle, Charles Lebailly, Ren Dujarric de la Rivire discovery of influenza virus 118
1863 Casimir Davaine first association of a specific infectious organism and a specific disease 40 1918 Anton Breinl, John Cleland, Eric French discovery of Murray Valley encephalitis virus 120
1865 Gregor Mendel father of genetics 41 1919 Arnold Lwenstein discovery of herpes simplex virus 1 121
1868 Johan-Friedrich Miescher discovery and characterization of nucleic acids 42 1920 Gaston Ramon, others improvements in many early vaccine technologies 122
1874 William Osler father of modern scholarly medicine 43 1921 Robert Montgomery discovery of African swine fever virus (the only asfarvirus) 123
1876 Robert Koch along with Louis Pasteur, the founder of microbiology 44 1921 Hans Creutzfeldt, Alfons Jakob initial description of Creutzfeldt-Jakob disease 124
1877 Patrick Manson, Charles Laveran, Ronald Ross, Giovanni Grassi etiology of malaria 45 1923 Theodor Svedberg development of the ultracentrifuge 125
1879 Stanford Chaille father of hygiene and health education in the United States 46 1925 Florence Sabin the role of monocytes and other WBCs in viral/bacterial infections 126
1690 Yellow fever epidemics in the United States 47 1925 Helmut Ruska, Thomas Weller, others discovery of varicella-zoster virus 127
1881 Carlos Finlay initial experiments on the mode of transmission of yellow fever virus 48 1925 Henri Valle Otto Waldmann Herman Frenkel foot-and-mouth disease vaccines 128
1883 Elie Metchnikoff, Paul Ehrlich founding of immunology, hematology, chemotherapy 49 1926 Peter Olitsky, Jacob Traum, William Cotton discovery of vesicular stomatitis viruses 129
1884 Louis Pasteur, Emile Roux, Charles Chamberland, Louis Thuillier rabies vaccine 50 1926 Frederik Kraneveld, Thomas Doyle discovery of Newcastle disease virus (first paramyxovirus) 130
1884 Charles Chamberland development of porcelain ultrafilter, key to discovery of viruses 51 1927 Thomas Rivers publication of the paper that established virology as a distinct science 131
1886 Adolf Mayer the concept of transmissibility, earliest concept of an ultrafilterable virus 53 1928 Thomas Rivers publication of the first major virology book, Filterable Viruses 133
1887 John Buist, Edmund Cowdry elementary & inclusion bodies of vaccinia & variola viruses 54 1928 Rockefeller Foundation Virus Laboratory (RFVL) and its international research programs 134
1887 Joseph Kinyoun founding of the National Institutes of Health 56 1928 Fred Griffith transformation in bacteria, seminal to development of molecular genetics 135
1887 Louis Pasteur, colleagues founding of the Institut Pasteur 57 1928 Rebecca Lancefield, Edwin Lennette, others founding of viral disease diagnostics 136
1892 Hermann Biggs New York City Department of Health microbiology laboratory 58 1928 Adrian Stokes, Noel Hudson transmission of yellow fever virus to rhesus macaques 137
1892 George Sternberg discovery of virus neutralization (vaccinia virus) 59 1928 Constant Mathis, others transmission of yellow fever virus to rhesus macaques 138
1893 Theobald Smith, Cooper Curtice proof of arthropod transmission of disease, Texas fever 60 1928 Jean-Louis Verge, J. Christofferoni feline panleukopenia virus (the first parvovirus) 139
1892 Dmitry Ivanovsky discovery of tobacco mosaic virus 62 1929 Stefan Nicolau discovery of Borna disease virus (the first bornavirus) 140
1898 Martinus Beijerinck discovery of tobacco mosaic virus 63 1930 Robert Green, Newell Ziegler infectious canine hepatitis virus (the first adenovirus) 141
1898 Friedrich Loeffler, Paul Frosch discovery of foot-and-mouth disease virus (the first virus) 64 1930 Leslie Webster, Charles Armstrong, Max Theiler development of the mouse for virology 143
1898 Giuseppe Sanarelli discovery of myxoma virus (the first poxvirus) 66 1930 Karl Meyer, Beatrice Howitt western equine encephalitis virus (the first alphavirus) 145
1930 Richard Shope discovery of swine influenza virus 147 1945 Mikhail Chumakov, Ghislain Courtois, others Crimean-Congo hemorrhagic fever virus 212
1931 William Elford, Christopher Andrewes graded collodion membranes for virion size 148 1946 Joshua Lederberg, Edward Tatum discovery of genetic recombination in bacteria 213
1931 Alice Woodruff, Ernest Goodpasture embryonating hens eggs as host for viruses 149 1946 Max Delbrck discovery of genetic recombination in viruses (bacteriophage) 215
1931 Frank Macfarlane Burnet, Herald Cox embryonating hens eggs as host for viruses 150 1946 Kenneth Smithburn, Alexander Haddow, Alexander Mahaffy Bunyamwera virus 216
1931 Robert Daubney, John Hudson, Percy Garnham discovery of Rift Valley fever virus 151 1946 Lloyd Florio, Mabel Miller, Edward Mugrage Colorado tick fever virus (first coltivirus) 217
1932 John Bittner, Clarence Little Jackson Laboratory mouse mammary tumor virus 153 1947 Frank Macfarlane Burnet, Alfred Gottschalk, Ernst Klenk viral receptors on target cells 218
1932 Frederick Gay, Albert Sabin, Margaret Holden, Arthur Wright discovery of B virus 154 1947 Arnold Beckman development of the Spinco Model E analytical ultracentrifuge 219
1933 Wilson Smith, Christopher Andrewes, Patrick Laidlaw ferret model, etiology of influenza 155 1947 Mettler Instruments AG development of first single-pan precision analytical balance 220
1933 Richard Shope, Peyton Rous, Joseph Beard discovery of rabbit (Shope) papillomavirus 156 1948 Frank Fenner founding of viral pathogenesis research (Ectromelia virus in mice) 221
1933 Ernst Ruska, Max Knoll invention of the transmission electron microscope 157 1948 Gilbert Dalldorf, Grace Sickles discovery of Coxsackieviruses 222
1933 Ralph Muckenfuss, Leslie Webster, Charles Armstrong St. Louis encephalitis virus 159 1948 Thomas Rivers, Frank Horsfall, Igor Tamm book, Viral and Rickettsial Infections of Man 223
1933 Fred Soper, others discovery of the sylvan/jungle yellow fever cycle 161 1948 Gueh-djen Hsiung, Phillip Gardner, Pekka Halonen application of viral diagnostics 224
1933 William Dimmock, Elvis Doll equine abortion virus, equine rhinopneumonitis virus 162 1949 Edward Pickels development of the Spinco Model L preparative ultracentrifuge 225
1934 Claud Johnson, Ernest Goodpasture discovery of mumps virus 163 1949 John Enders, Thomas Weller, Frederick Robbins cell culture for polio, measles 226
1934 Arnold Beckman invention of the electronic pH meter 164 1949 Andr Lwoff, Louis Siminovitch, Niels Kjeldgaard lysogeny / induction (bacteriophage) 227
1934 Malcolm Merrill, Charles Lacaillade, Carl Ten Broeck eastern equine encephalitis virus 165 1950 Erwin Chargaff discovery in DNA that number of molecules of A+T equals that of G+C 228
1934 Michitomo Hayashi, Shiro Kasahara, others discovery of Japanese encephalitis virus 166 1950 Cedric Mims development of immunofluorescence in viral pathogenesis research 229
1935 Wendell Stanley purification and crystallization of tobacco mosaic virus 167 1951 George Gey HeLa cell line (human cervical adenocarcinoma) 230
1935 Max Theiler, Hugh Smith development of yellow fever vaccine 168 1951 Ludwik Gross, Charlotte Friend, others murine leukemia and lymphoma viruses 231
1935 Max Knoll, Manfred Ardenne, Vladimir Zworykin, Charles Oatley scanning microscope 169 1951 Esther Lederberg discovery of the bacteriophage 232
1936 Frederick Bawden, Norman Pirie tobacco mosaic virus is comprised of RNA/protein 170 1951 Linus Pauling, John Kendrew, Max Perutz structure of proteins (X-ray crystallography) 233
1936 Jean Cuill, Paul-Louis Chelle transmission of scrapie prion using ultrafiltered tissue 171 1952 Alfred Hershey, Martha Chase biological proof of DNA as the material of inheritance 234
1936 Charles Armstrong, Thomas Rivers, Erich Traub lymphocytic choriomeningitis virus 172 1952 Wendell Stanley establishment of Virus Laboratory at University of California Berkeley 235
1936 Peyton Rous, Joseph Beard induction of carcinomas in other species by papillomavirus 173 1952 Joshua Lederberg, Norton Zinder transduction: DNA transfer by bacteriophage 236
1936 Jacob Traum discovery of vesicular exanthema virus of swine (the first calicivirus) 174 1952 Karl Maramorosch discovery that some viruses can replicate in both plants and insects 237
1936 Theodosius Dobzhansky publication of Genetics and the Origin of Species 175 1952 Renato Dulbecco, Marguerite Vogt virus quantification by plaque assay 238
1937 Fred Beaudette, Charles Hudson avian infectious bronchitis virus (the first coronavirus) 176 1952 Seymour Cohen DNA of some bacteriophages contains 5-hydroxymethylcytosine 239
1937 Thomas Rivers criteria for proof of viral disease causation: Koch postulates revisited 177 1953 Wallace Rowe, Robert Huebner, others discovery of human adenoviruses 240
1937 Lev Zilber, Mikhail Chumakov, Elizabeth Levkovich tick-borne encephalitis virus 178 1953 James Watson, Francis Crick, Maurice Wilkins, Rosalind Franklin the structure of DNA 241
1937 Elizabeth Wollman, Eugne Wollman, Augustus Doermann the eclipse period 179 1953 James Murphy, Frederik Bang discovery of virus release by budding at the cell surface 243
1938 G. Krber, Lowell Reed, Hugo Muench methods for estimating fifty percent endpoints 180 1953 Nakao Ishida discovery of Sendai virus (murine parainfluenza virus 1) 244
1938 Vladimir Kubes, Francisco Rios discovery of Venezuelan equine encephalitis virus 181 1953 Rupert Billingham, Peter Medawar, Leslie Brent discovery of immunological tolerance 245
1938 Robert Doerr, Curt Hallauer the major virology book, Handbuch der Virusforschung 182 1953 Wilhelm Bernhard, Albert Dalton morphogenesis / classification of retroviruses 246
1938 Bodo von Borries, Ernst Ruska, Helmut Ruska first electron micrographs of viruses 183 1953 Albert Sabin discovery of orthoreoviruses (the first reoviruses) 247
1939 First international virology journal, Archiv fr die gesamte Virusforschung 184 1954 Renato Dulbecco, Marguerite Vogt concept of the latent period, eclipse period 248
1939 Charles Armstrong adaptation of poliovirus to cotton rats and mice 185 1954 Jonas Salk, Julius Youngner, Pierre Lpine, Thomas Francis inactivated polio vaccine 249
1939 Paul Mller discovery of insecticidal quality of DDT for control of vector-borne diseases 187 1954 Margaret Smith discovery of murine cytomegalovirus 250
1939 Emory Ellis, Max Delbrck one-step growth experiment (single burst experiment) 188 1954 Bjrn Sigurdsson concept of slow viruses (e.g., maedi-visna virus, scrapie prion) 251
1940 Kenneth Smithburn, Thomas Hughes, Alexander Burke, John Paul West Nile virus 189 1955 Gyula Taktsy invention of the microtiter system for virologic and serologic assays 252
1940 Ernst Mayr, others the conceptual basis of the modern evolutionary synthesis 190 1955 George Hirst, Peter Wildy, Lloyd Kozloff, Robert Wagner, others major virology journals 253
1940 Barbara McClintock development of the concept of transposable elements, transposons 191 1955 Richard Taylor, Herbert Hurlbut, Telford Work, others discovery of Sindbis virus 254
1940 Katherine Sanford, Wilton Earle development of the L cell line and L929 cloned cell line 192 1955 Heinz Fraenkel-Conrat, Robley Williams reconstitution of tobacco mosaic virus 255
1940 Keith Porter, others description of fine structure / biochemistry of cellular organelles 193 1955 Seymour Benzer, Paul Berg, Maxine Singer evolving definitions of the gene 256
1940 Presidential Medal of Freedom, National Medal of Science, Copley Medal of Royal Society 194 1955 Werner Schfer, Svonimir Dinter discovery that fowl plague virus is an influenza virus 257
1940 Albert Claude, Christian de Duve, George Palade structure / biochemistry of organelles 195 1956 J. Anthony Morris, Robert Chanock respiratory syncytial virus (the first pneumovirus) 258
1941 George Hirst influenza hemagglutinin, hemagglutination-inhibition, neuraminidase 196 1956 William Pelon, WH Price, William Mogabgab discovery of human rhinoviruses 259
1941 Norman Gregg discovery of congenital abnormalities caused by rubella virus infection 197 1956 Alfred Gierer, Heinz Fraenkel-Conrat infectivity of viral RNA (tobacco mosaic virus) 260
1941 William Hammon, William Reeves natural history of arboviruses in vector arthropods 198 1956 Stewart Madin, Charles York, Delbert McKercher infectious bovine rhinotracheitis virus 261
1941 Arnold Beckman development of the ultraviolet spectrophotometer (the Beckman DU) 199 1956 R.William Ross discovery of chikungunya virus 262
1941 Thomas Anderson discovery of the amazing variety of bacteriophage morphologies 200 1956 Robert Chanock discovery of human parainfluenza viruses 263
1942 Chen-Hsiang Huang development of the quantitative virus neutralization assay 201 1956 Margaret Smith, Thomas Weller, Wallace Rowe discovery of human cytomegalovirus 264
1942 Joseph Mountin, others founding of U.S. Centers for Disease Control and Prevention 202 1956 John Gorham, James Henson, Robert Leader, Frank Dixon Aleutian disease of mink virus 265
1942 Albert Coons development of immunofluorescence labeling methods 204 1957 Niels Jerne, David Talmage, Frank Macfarlane Burnet clonal selection in immunity 266
1943 U.S. Army, Camp Detrick military high-containment virology laboratories 205 1957 Marvin Minsky development of the confocal microscope 267
1944 Kenneth Smithburn, Alexander Haddow discovery of Semliki Forest virus 206 1957 Alick Isaacs, Jean Lindenmann discovery of interferons 268
1944 Oswald Avery, Colin MacLeod, Maclyn McCarty DNA as the material of inheritance 207 1957 Telford Work, F.R. Rodriguez, Pravin Bhatt discovery of Kyasanur Forest disease virus 269
1944 Erwin Schrdinger book, What is Life? motivating scientists toward molecular biology 208 1957 Elvis Doll, John Bryans, William McCollum equine arteritis virus (the first arterivirus) 270
1944 Fred MacCallum, Saul Krugman, others separation of hepatitis A and hepatitis B 209 1957 James Gillespie, K.M. Lee, others discovery of bovine virus diarrhea virus 271
1945 Max Delbrck, Salvador Luria, others founding of Phage Course at Cold Spring Harbor 210 1957 Matthew Meselson, Frank Stahl, Jerome Vinograd density gradient ultracentrifugation 272
1945 Thomas Francis, George Hirst, Fred Davenport, others influenza vaccines 211 1958 Matthew Meselson, Frank Stahl the semi-conservative mode of replication of DNA 273
1958 Sarah Stewart, Bernice Eddy discovery of murine polyoma virus (the first polyomavirus) 274 1966 Carleton Gajdusek, Clarence Gibbs, William Hadlow kuru prion in non-human primates 337
1958 Robert Kissling, Robert Goldwasser rabies immunofluorescence diagnostic method 275 1967 Rudolf Siegert, Werner Slenczka, Robert Kissling, Frederick Murphy Marburg virus 338
1958 Armando Parodi, Ignacio Pirosky, colleagues Junin virus (Argentine hemorrhagic fever) 276 1967 U.S. Communicable Disease Center civilian high-containment virology laboratory 339
1958 Denis Burkitt description of Burkitts lymphoma in African children 277 1967 Joseph Kates, Brian McAuslan viral DNA-dependent RNA polymerase (vaccinia virus) 340
1958 Gabriele Zu Rhein, Duard Walker progressive multifocal leukoencephalopathy, JC virus 278 1967 Peter Biggs, Anthony Churchhill, Ben Burmester, others Mareks disease virus 341
1959 James Gowans, Peter Medawar discovery of lymphocyte recirculation 279 1967 Jacob Maizel Jr., Ulrich Laemmli SDS polyacrylamide gel electrophoresis of proteins 342
1959 Albert Sabin, Herald Cox, Hilary Koprowski, others attenuated live-virus polio vaccine 280 1968 Aaron Shatkin, Jean Sipe discovery of dsRNA-dependent RNA polymerase (reovirus) 343
1959 Lawrence Kilham, others discovery of murine parvoviruses 281 1968 Werner Henle, Gertrude Henle Epstein-Barr virus and infectious mononucleosis 344
1959 Sydney Brenner Robert Horne invention of negative contrast electron microscopy 282 1968 Peter Wildy, Joseph Melnick, others the First International Congress for Virology 345
1959 Albrecht Kleinschmidt invention of electron microscopic visualization of viral DNA 284 1968 Werner Arber, Hamilton Smith, Daniel Nathans, others restriction endonucleases 348
1959 Gerald Edelman, Rodney Porter, Alfred Nisonoff structure of antibody molecules 285 1969 Robert Huebner, George Todaro development of the viral oncogene hypothesis 349
1959 Robert Sinsheimer that a virus may have a single stranded DNA genome (X174) 286 1969 Sonja Buckley, Jordi Casals-Ariet discovery of Lassa virus 350
1959 Rosalyn Yalow, Solomon Berson development of radioimmunoassays (RIAs) 287 1969 Charles Mebus, Marvin Twiehaus, others discovery of bovine rotavirus (first rotavirus) 351
1959 Arthur Kornberg, Severo Ochoa mechanisms in the replication of DNA and RNA 288 1970 Irvine Page, others Institute of Medicine of the U.S. National Academy of Sciences 252
1959 William Prusoff development of the first antiviral drug, 5-iodo-2-deoxyuridine (IUdR) 289 1970 Robert Shope, Frederick Murphy, others rabies-like viruses (the genus Lyssavirus) 353
1959 William Reeves, others founding of American Committee on Arthropod-Borne Viruses 290 1970 Frank Fenner, David White book, Medical Virology 354
1960 Walter Plowright development of attenuated-live virus rinderpest vaccine 291 1970 David Baltimore, Alice Huang RNA-dependent RNA polymerase in VSV 355
1960 Benjamin Sweet, Maurice Hilleman discovery of SV40 virus 292 1970 Howard Temin, David Baltimore, colleagues reverse transcriptase of retroviruses 356
1960 Vernon Riley, Kenneth Rowson discovery of lactate dehydrogenase-elevating virus 293 1970 Roland Robins, Joseph Witkowski, colleagues ribavirin, broad spectrum antiviral drug 357
1960 Wayne Thompson, Bernard Kalfayan, Ralph Anslow discovery of La Crosse virus 294 1970 Joseph Kates James Darnell Jr., others discovery of the 3 poly(A) tail on viral mRNAs 358
1960 Frank Dixon, others founding of immunopathology, viruses in immune complex diseases 295 1970 Walter Plowright, Robert Tesh, Douglas Watts, others transovarial transmission 359
1960 Cedric Mims, Neal Nathanson, others the modern era of viral pathogenesis research 296 1970 Pekka Halonen, others linkage of rapid virus diagnostics to clinical care 360
1960 Robert Merrifield solid phase synthesis technology (polypeptides, oligonucleotides) 297 1970 Seymour Kalter, Richard Heberling development of non-human primate virology 361
1961 Franois Jacob, Jacques Monod, Andr Lwoff genetic control of enzyme / virus synthesis 298 1970 Janet Butel, Keerti Shah controversy whether SV40 virus (vaccines) causes brain tumors 362
1961 Jacques Miller discovery of the role of the thymus in cellular immunity 300 1970 Gertrude Elion, George Hitchings, others rational drug design, acyclovir / her pes 363
1961 Michael Stoker, Ian Macpherson development of BHK-21 cell line 301 1971 Eva Engvall, Bauke van Weemen, colleagues enzyme-linked immunoassays 364
1961 Francis Crick, Sydney Brenner, others the triplet coding strategy of DNA 302 1971 David Baltimore system of virus categorization based on the replicative path to mRNA 365
1961 John Cairns discovery of the length of the circular DNA of the bacteriophage T2 303 1971 Theodore Diener discovery of viroids (infectious naked RNA molecules) 366
1961 U.S. Communicable Disease Center Morbidity and Mortality Weekly Report (MMWR) 304 1971 Sylvia Gardner, Anne Field, colleagues discovery of BK virus, kidney / bladder disease 367
1961 Marshall Nirenberg, Har Khorana, Robert Holley deciphering of the genetic code 305 1971 Donald Carey, Jin-Won Song, Robert Shope, Ric Yanagihara Thottapalayam virus 368
1961 Jordi Casals, Richard Taylor, Robert Shope, Charles Calisher arbovirus classification 306 1971 Bert Achong, Michael Epstein, Thomas Folks simian foamy virus infection in humans 369
1962 Yosihiro Yasumura, Yosio Kawakita establishment of Vero cell line 307 1971 George Baer, Franz Steck, Alex Wandeler oral vaccination of wildlife against rabies 370
1962 Lizbeth Kraft discovery of mouse hepatitis virus 308 1972 Albert Kapikian, colleagues discovery of Norwalk virus (the first human calicivirus) 371
1962 Cold Spring Harbor Symposium: Basic Mechanisms in Animal Virus Biology 309 1972 Paul Berg, Dale Jackson, Robert Symons development of recombinant-DNA technology 372
1962 Andr Lwoff, Robert Horne, Paul Tournier classification of viruses 310 1973 Stanley Cohen, Herbert Boyer application of recombinant DNA technology 373
1962 Aaron Klug, Donald Caspar the principles of the icosahedral structure of viruses 311 1973 Paul Berg, others Asilomar conferences: Biohazards in Biological Research 374
1962 John Trentin, colleagues the induction of tumors in hamsters by human adenoviruses 312 1973 Stephen Feinstone, Albert Kapikian, Robert Purcell discovery of hepatitis A virus 375
1962 Albert Cosgrove, Hiram Lasher infectious bursal disease virus (the first birnavirus) 313 1973 Ruth Bishop, Ian Holmes, Thomas Flewett, Albert Kapikian, others human rotaviruses 376
1962 Norman Anderson invention of rate-zonal ultracentrifugation 314 1973 Joseph Sambrook, others agarose gel electrophoresis of DNA 377
1962 Karl-Eduard Schneweis, Walter Dowdle, Andr Nahmias differentiation of HSV 1 and 2 315 1973 Ralph Steinman, Zanvil Cohn discovery of dendritic cells 378
1962 Robert Webster, Graeme Laver the link between avian and human influenza viruses 316 1973 Councilman Morgan, Ari Helenius, others variety and complexity of virus entry mechanisms 379
1963 Peter Gomatos, Igor Tamm discovery of double-stranded RNA in a virus (reovirus) 317 1973 Daniel Nathans, others restriction enzyme map of a viral genome (SV40 virus) 380
1962 Osamu Shimomura, Martin Chalfie, Roger Tsien green fluorescent protein 318 1974 Peter Doherty, Rolf Zinkernagel how immune system recognizes virus-infected cells 381
1963 John Enders, Thomas Peebles, Samuel Katz, Kevin McCarthy measles vaccine 319 1974 Georges Khler, Csar Milstein development of monoclonal antibodies 382
1963 Maurice Hilleman, others development, commercialization of pediatric viral vaccines 320 1974 Vigdis Torsvik, Ian Dundas, Allen Wais, others viruses of Archaea, odd morphologies 383
1963 Wilbur Downs, Charles Anderson, Leslie Spence discovery of Tacaribe virus 321 1974 Harald zur Hausen association of human papillomaviruses and cervical carcinoma 384
1963 Seiichi Matsumoto, Kaneatsu Miyamoto morphogenesis of rabies virus, Negri body 322 1974 Ilse Tischer, Hans Gelderblom discovery of porcine circovirus (the first circovirus) 385
1964 Michael Epstein, Bert Achong, Yvonne Barr Epstein-Barr virus and Burkitts lymphoma 323 1975 Yvonne Cossart, Anne Field, colleagues parvovirus B-19, fifth disease and aplastic crisis 386
1964 William Jarrett, colleagues discovery of feline leukemia virus 324 1975 C. Richard Madeley, colleagues discovery of human astrovirus 1 (the first astrovirus) 387
1964 Karl Johnson, Patricia Webb, others Machupo virus, Bolivian hemorrhagic fever 325 1975 Aaron Shatkin, Bernard Moss, colleagues discovery of 5 cap on messenger RNAs 388
1965 Michel Bouteille, John Sever, others etiology of subacute sclerosing panencephalitis 326 1975 Edwin Southern development of southern blotting 389
1965 Baruch Blumberg, Harvey Alter, others discovery of Australia antigen, hepatitis B virus 327 1976 Karl Johnson, Patricia Webb, Frederick Murphy, Guido Van der Groen Ebola virus 390
1965 David Tyrrell, June Almeida, others discovery of human coronaviruses 328 1976 J. Michael Bishop, Harold Varmus the cellular origin of retroviral oncogenes 392
1965 Wayne Atchison, Joseph Melnick, colleagues discovery of adeno-associated viruses 329 1976 Rudolf Rott, Christoph Scholtissek optimum gene constellation determines virulence 393
1965 Robin Valentine, Helio Pereira, Erling Norrby adenovirus structural elements 330 1976 Alfred Evans criteria for proof of viral disease causation: Henle-Koch postulates again 394
1965 Bernard Roizman seminal studies of the molecular biology of herpes simplex virus 331 1976 U.S. Government threat of swine flu epidemic, national emergency vaccination program 395
1965 Shinpei Kasakura, Kendall Smith, others discovery of the first cytokine, IL-2 332 1976 Walter Fiers, Frederick Sanger sequencing of viral genomes (phages MS2 and 174) 396
1965 Leonard Hayflick development of human diploid cell strains (WI-1 through WI-44) 333 1977 Phillip Sharp, Richard Roberts, colleagues RNA splicing and split genes (adenovirus) 397
1966 Mark Ptashne, Walter Gilbert identification of repressor genes (lac operon) 334 1977 D.A. Henderson, Isao Arita, Frank Fenner, many others global eradication of smallpox 398
1966 Peter Wildy, Frank Fenner, others International Committee on Taxonomy of Viruses 335 1977 Walter Gilbert, Frederick Sanger development of rapid DNA sequencing technology 399
1977 Mario Rizzetto, John Gerin, colleagues hepatitis delta virus (the only deltavirus) 400 1990 Nitza Frenkel discovery of human herpesvirus 7 (HHV7) 463
1977 Scott Halstead immune enhancement in dengue hemorrhagic fever / shock syndrome 401 1990 Cathrinus Terpstra, others porcine reproductive and respiratory syndrome virus 464
1977 Carl Woese, colleagues The Tree of Life, phylogeny based on mRNA gene sequences 402 1991 Tim Berners-Lee development of the World Wide Web (WWW) 465
1978 Ho Wang Lee, Pyung Woo Lee, Karl Johnson Hantaan virus, HFRS 403 1991 Craig Venter, Hamilton Smith (TIGR), others shotgun cloning/sequencing methods 466
1978 Leland Carmichael, Max Appel discovery / characterization of canine parvovirus 404 1991 Gregory Reyes, Michael Wigler, others novel techniques to find uncultivable viruses 467
1978 Charles Weissmann, colleagues first reverse genetics experiment (bacteriophage Q) 405 1991 Rosalba Salas, Robert Tesh, others Guanarito virus, Venezuelan hemorrhagic fever 468
1978 Anthony Waterson, Lise Wilkinson book, An Introduction to the History of Virology 406 1992 Jean Smith, Charles Rupprecht, others rabies virus genotyping, mapping of variants 469
1978 Steven Harrison, James Hogle, others structure of viruses (poliovirus, rhinovirus) 407 1992 Joshua Lederberg, Stephen Morse, others concept of New and Emerging Infectious Diseases 470
1978 Wolf Szmuness, Palmer Beasley, others HBV infection and hepatocellular carcinoma 408 1993 Stephen Morse, others Program for Monitoring Emerging Diseases (ProMED) 471
1979 Lynn Enquist, colleagues cloning viral DNA fragments into phage vector 409 1993 Stuart Nichol, C.J. Peters, Thomas Ksiazek, others Sin Nombre virus, HPS 472
1979 David Lane, Arnold Levine, others p53 tumor suppressor protein in SV40 infected cells 410 1994 Robert Webster, Alan Granoff, Brian Mahy, M. van Regenmortel Encyclopedia of Virology 473
1979 George Stark, others western blotting: transfer of proteins from gels, antibody detection 411 1994 Yuan Chang, Patrick Moore, colleagues herpesvirus 8, Kaposi sarcoma herpesvirus 474
1980 Robert Swanson, Herbert Boyer Genentech, first biotech company listed on NYSE 412 1994 Karl-Klaus Conzelmann, colleagues reverse genetics for negative-strand RNA viruses 475
1980 Bernard Poiesz, Robert Gallo, Mitsuaki Yoshida, David Golde, others HTLV 1 & 2 413 1994 Terezinha Lisieux Moraes Coimbra, others Sabi virus and Brazilian hemorrhagic fever 476
1980 Walter Dowdle, Frederick Murphy, James Hughes National Center for Infectious Diseases 414 1995 Keith Murray, Peter Hooper, Alex Hyatt, colleagues Hendra virus and reservoir host bats 477
1980 Elizabeth Williams, Stuart Young chronic wasting disease of deer / elk is a prion disease 415 1995 Richard Preston publication of the influential book, The Hot Zone 478
1980 Thomas Cech, Sidney Altman, colleagues catalytic properties of RNA, ribozyme concept 416 1996 Robert Will, James Ironside, John Collinge BSE prion, variant Creutzfeldt-Jakob disease 479
1981 MMWR - Pneumocystis carinii pneumonia in Los Angeles, Kaposi sarcoma in NY AIDS 417 1996 David Fredricks, David Relman sequenced-based criteria for causation of viral diseases 480
1981 Vincent Racaniello, David Baltimore, Eckard Wimmer infectious clone of poliovirus 418 1996 Martin Hirsch, David Ho, Thomas Merigan, others HAART therapy for AIDS 481
1981 Gerd Binnig, Heinrich Rohrer development of scanning tunneling electron microscope 419 1996 Jules Hoffmann, Bruce Beutler, others discovery of Toll genes / Toll-like receptors 482
1981 Don Wiley, John Skehel, Ian Wilson, others structure of influenza virus hemagglutinin 420 1996 Hans Kuypers, Gabriella Ugolini, others viruses as neuronal pathway tracers 483
1981 Robert Weinberg, Michael Wigler cloning of first human tumor-derived oncogene, H-ras 421 1996 National Center for Biotechnology Information development of PubMed 484
1981 Bill Joklik, Harry Ginsberg, others founding of the American Society for Virology 422 1997 Robert Webster, others highly pathogenic H5N1 influenza virus, lethal disease 485
1981 William Rutter, Pablo Valenzuela, colleagues first recombinant human vaccine, HBsAg 424 1997 Tsutomu Nishizawa, Hiroaki Okamoto, colleagues torque teno virus, anelloviruses 486
1982 Stanley Prusiner concept of the prion etiologic agents of spongiform encephalopathies 425 1997 Luminex Corporation, others development of high throughput viral diagnostics instruments 487
1982 Walter Goad, others development of GenBank 426 1998 Andrew Fire, Craig Mello RNA interference (RNAi, RNA silencing by dsRNA) 488
1982 Michael Oldstone, colleagues concept of viral damage to cellular homeostasis 427 1998 Larry Page, Sergey Brin development of Google 489
1982 Gerald Woode, Marian Horzinek, Graham Beards bovine, equine, human toroviruses 428 1998 Nancy Cox, Yoshihiro Kawaoka, Robert Webster molecular basis of influenza virulence 490
1983 Mikhail Balayan discovery of hepatitis E virus (the only hepevirus) 429 1999 Kaw Bing Chua, Kenneth Lam, William Bellini, T Ksiazek, colleagues discovery of Nipah virus 491
1983 Francoise Barr-Sinoussi, Luc Montagnier, Jean-Claude Chermann discovery of HIV1 430 1999 Deborah Asnis, Marcelle Layton, W. Ian Lipkin, Robert Lanciotti West Nile virus in U.S. 492
1983 Marvin Carruthers, Applied Biosystems, Inc. commercial DNA synthesis technology 431 2001 Albert Osterhaus, Bernadette van den Hoogen, colleagues human metapneumovirus 493
1983 Andrew Murray, Jack Szostack yeast artificial chromosome, cloning large DNA fragments 432 2001 U.S. Government anthrax bioterrorism events, large biodefense research programs 494
1983 Bernard Moss, Enzo Paoletti recombinant vaccinia virus as vector for viral vaccines 433 2001 Foot-and-mouth disease epidemic in U.K., destruction of large numbers of livestock 495
1985 Kary Mullis, colleagues, Cetus, Perkin-Elmer polymerase chain reaction (PCR) 434 2002 Eckard Wimmer, colleagues synthesis of poliovirus complementary DNA 496
1985 Julio Maiztegui, Julio Barrera Oro, George French Argentine hemorrhagic fever vaccine 435 2002 Joseph DeRisi, W. Ian Lipkin microarray technology for identification of viruses 497
1985 Bernard Fields, others publication of Fields Virology, now five editions 436 2002 Ian Frazer, Sanofi Pasteur, Merck, GlaxoSmithKline papillomavirus vaccine 498
1985 bioMrieux, Bio-Rad Genetic Systems, Abbott first FDA approved HIV antibody EIAs 437 2002 Erling Norrby papers and book on awarding of the Nobel Prizes pertinent to virology 499
1985 Norman Letvin, Ronald Desrosiers, colleagues simian immunodeficiency viruses (SIVs) 438 2003 Peter Walsh, Eric Leroy, others Ebola epidemics in endangered great apes 500
1985 Francis Barin, Phyllis Kanki, Max Essex, colleagues human immunodeficiency virus 2 439 2003 International Human Genome Project consensus sequence of the human genome 501
1985 Alec Jeffreys development of DNA fingerprinting, single nucleotide polymorphisms 440 2003 Carlo Urbani, Malik Peiris, Leo Poon, others SARS coronavirus and global epidemic 502
1985 Carole Richardson bovine spongiform encephalopathy in the United Kingdom 441 2003 Bernard La Scola, Didier Raoult, others discovery of mimivirus, the largest virus known 503
1985 Saul Kit, colleagues development of first recombinant live-virus vaccine (pseudorabies) 442 2003 Lawrence Livermore National Laboratory Autonomous Pathogen Detection System 504
1981 Development of modern viral vaccinology based upon innovative technologies 443 2005 Eric Leroy, Jonathan Towner, others reservoir hosts of Ebola / Marburg viruses are bats 505
1986 Niels Pedersen, Janet Yamamoto, colleagues discovery of feline immunodeficiency virus 444 2005 Jeffery Taubenberger, Terrence Tumpey, others 1918 influenza virus sequenced 506
1986 Leroy Hood, Lloyd Smith, Applied Biosystems automated DNA sequencing 445 2005 Renaud Mahieux, Antoine Gessain discovery of human T lymphotropic viruses 3 and 4 507
1987 Frank Fenner, others publication of book, Veterinary Virology (currently four editions) 446 2005 Takaji Wakita, Charles Rice, others in vitro replication of hepatitis C virus 508
1987 Marty St. Clair, Samuel Broder, Burroughs Wellcome/NCI first anti-HIV drug (AZT) 447 2005 Jonathan Rothberg, 454 Life Sciences (Roche) development of 454 pyrosequencing 509
1988 Mario Capecchi, Oliver Smithies, Martin Evans knockout, genetically manipulated mice 448 2005 Founding of the European Centre for Disease Prevention and Control (ECDC) 510
1988 Koichi Yamanishi, Takeshi Kurata, Philip Pellett, Carlos Lopez human herpesvirus 6B 449 2005 Tobias Allander, colleagues discovery of human bocavirus (parvovirus) 511
1988 Eckard Wimmer, Nahum Sonnenberg internal ribosomal entry sites in virus mRNA 450 2006 Laura Kahn, Bruce Kaplan, Thomas Monath One Health / One Medicine Initiative 512
1988 National Library of Medicine National Center for Biotechnology Information (NCBI) 451 2007 Massimo Pigliucci, Gerd Mller, others the Extended Evolutionary Synthesis 513
1988 H. Fred Clark, Albert Kapikian, Richard Ward development of human rotavirus vaccines 452 2007 Tobias Allander, David Wang, Yuan Chang, others human polyomaviruses KI, WU, MC 514
1988 Albert Osterhaus, colleagues phocine distemper virus, marine mammal morbilliviruses 453 2008 Bernard La Scola, Didier Raoult, others discovery of first virophage, Sputnik 515
1988 Manfred Eigen, others modern concepts of origin of viruses, virus evolution, quasispecies 454 2010 FAO Global Rinderpest Eradication Programme global eradication of rinderpest 516
1988 Thomas Brock discovery of Taq polymerase 457 2015 WHO Global Polio Eradication Initiative planned global eradication of poliomyelitis 517
1989 Michael Houghton, Qui-Lim Choo, George Kuo, Daniel Bradley hepatitis C virus 458 FURTHER READING
1989 Stephen Fodor, Patrick Brown, others development of microarray technology 459 SOME MAJOR IMAGE LIBRARIES
1989 Marc van Regenmortel definition of virus species 460 INDEX
1989 ivind Bergh, Curtis Suttle, Craig Venter, others high concentrations of viruses in ocean 461
1990 International Human Genome Sequencing Consortium Human Genome Project 462
Hippocrates of Kos II
(~460 BCE - ~370 BCE)
Hippocrates is considered the father of modern medicine, still influencing, 25 centuries after his time, various aspects of medical
and veterinary practice and biomedical research. His works (Corpus Hippocraticum, Hippocratic Collection) include references to
infectious diseases ranging from the nature of infection, hygiene, epidemiology, and the immune response, to detailed descriptions of
syndromes such as mumps, herpes, smallpox, hepatitis (jaundice), the common cold, pneumonia, tuberculosis, malaria and tetanus.
page 6

~400BCE Hippocrates father of medicine, important epidemiologic and clinical observations of many diseases
Man Bitten by Mad Dog
Illustration from an Arabic translation of the Materia
Medica, Baghdad, Iraq, 1224.
Freer Gallery of Art, Smithsonian Institution,
Washington, DC.

Rabies (derived from the Sanskrit, rabhas meaning to do


violence) was first described in the 23rd century BCE in the
Eshuma Code of Babylonit is the oldest documented disease
of humans.
Other ancient scholars in Mesopotamia, China, Greece, Rome,
and India described its clinical signs and symptoms. In the first
century CE, Celsus and Galen described treatment, including
excision of the bitten tissue and cauterization of the wound with
a hot iron.
In 1546 the Italian physician, Fracastoro, published The
Incurable Wound, a treatise describing clinical rabies in humans:
The patient can neither stand nor lie down, like a mad man he
flings himself hither and thither, tears his flesh with his hands,
and feels intolerable thirst. This is the most distressing symptom,
for he so shrinks from water and all liquids. He would rather die
than drink or be brought near to water. It is then he bites other
persons, foams at the mouth, his eyes look twisted, and finally he
becomes exhausted and painfully breathes his last.

[from Smithsonian Institution, used with permission]


Rabid dog attacking a calf. Relief from the ruin of a 4th
century Roman settlement at Neumagen-Dhron near Trier
page 7 (Rheinland-Pfalz), Germany [from Marian Horzinek]

1224 Mesopotamian medicine rabies, the oldest documented disease of humans and domestic animals
Girolamo Fracastoro (1478-1553)
(Hieronymus Fracastorius)
In 1546, Fracastoro proposed that epidemic diseases are caused by transferable
tiny particles or spores that can transmit infection by direct or indirect
contact or even without contact over long distances. I call fomites [from
the Latin fomes, meaning tinder] such things as clothes, linen, etc., which
although not themselves corrupt, can nevertheless foster the essential seeds
of the contagion and thus cause infection. His theory remained influential for
nearly three centuries, before being displaced by the germ theory of Pasteur
and Koch. page 8

1546 Girolamo Fracastoro proposal that epidemic diseases are contagious, leading to the germ theory
Sacharias Jansen was a Dutch spectacle-maker credited with inventing, or contributing advances
towards the invention of, the telescope. In 2008, the Netherlands commemorated the 400th
anniversary of the telescope, honoring Jansen as one of the two possible inventors, the other being
Hans Lippershey.
Jansen is credited with inventing the first compound microscope, with the help of his father, Hans,
in 1595. Jansens life was documented very well before WWII. Many of the archives were destroyed
by a bombardment during the Nazi invasion of the Netherlands. If there had not been earlier
studies, we would not know anything of Jansens life, since all primary files were lost in the fires
following the bombardment.
It must be said that early compound
(at least two lenses)microscopes
were only capable of magnifications
of about x20-30, less than van
Leeuwenhoeks first simple (single
lens) microscope. But, the quest to
see things smaller than visible to the
naked eye had begun.

Marker for the site of the Sacharias Jansen (Zacharias


Janse) house, Middleburg, The Netherlands

Jansens microscope consisted of three sliding tubes with lenses fixed in the ends. The
eyepiece lens was bi-convex and the objective lens was plano-convex, an advanced compound
Sacharias Jansen (~1580-~1632) design for the time. Focusing was achieved by sliding the draw tube in or out while observing
(Zacharias Janssen) the specimen. No early models of Jansen microscopes have survived, but there is this
page 9 candidate housed in the Middleburg Museum, which some historians attribute to Jansen.

1595 Sacharias & Hans Jansen development of the first compound microscope
Robert Hooke (1635-1703)
Robert Hooke is one of the most neglected important natural philosophers of all
time. He was the inventor of, among other things, the iris diaphragm in cameras, the
universal joint used in motor vehicles, the balance wheel in watches, and he was the
originator of the word cell in biology He was Surveyor of the City of London after
the Great Fire of 1666, and was also an architect, experimenter and astronomer.
And, he was one of the inventors of the compound microscope. [Sacharias and Hans
Jansen in Holland built the first compound microscope around 1595, but their work
was little appreciated outside Holland.] Hookes observations received widespread
acclaim, since they were published in his remarkable book, Micrographia, in 1665.
There are no incontrovertible images of Robert Hooke the formulaic image here is
from the Hooke memorial window, St. Helens Church, Bishopsgate, London, where
Hooke was buried (and which was destroyed in an IRA bombing). Hooke served
for many years as Curator of Experiments for the Colledge for the Promoting of
Physico-Mathematicall Experimentall Learning and then its successor, the Royal
Society. As such he resided at its site in Gresham College, Bishopsgate. After Hooke
died, Isaac Newton became president of the Royal Society, and it fell to him to move
to new headquarters. Among the many items to be relocated was Robert Hookes
portrait during the move the portrait simply disappeared.

Microscopic view of a hairy mould colony by


Robert Hooke, 1665.
Published in Micrographia.
This image was the first published depiction of a
microorganism. The reproductive structures (sporangia)
are characteristic of the genus Mucor. Sporangia in
different stages of maturation are identified by letters
A, B, C, D.
Robert Hookes
compound microscope, 1665,
and lighting system (a brine-filled glass globe
placed between the light source and the microscope
to focus the light). As published in Micrographia.
page 10

1665 Robert Hooke first microscopic observation of a microorganism, a mold, genus Mucor
Anton van Leeuwenhoek
(1632-1723)
van Leeuwenhoeks Discoveries van Leeuwenhoeks Microscopes
Using his simple microscopes, van Leeuwenhoek van Leeuwenhoek constructed hundreds of microscopes
discovered, protozoa, Giardia, spermatozoa, rotifers, he sent many to the Royal Society and left 247 completed
hydra and volvox. He made observations of blood cells, microscopes (only nine are known to have survived; one sold
the nucleus of fish erythrocytes, capillaries and lymphatics, in 2009 for $459,000) and 172 additional mounted lenses
but perhaps his greatest discovery was of bacteria. In 1674 actually, they were not microscopes but simple magnifying
he wrote: Last winter while being sickly and nearly unable lenses. To make a lens, he drew out a small soda lime glass
to taste, I examined the appearance of my tongue, which rod in a flame, creating a long glass whisker. By reinserting the
was very furred This led him to examine an ox tongue end of a whisker into the flame, he created a very small, high-
with his microscope, and he saw that there were very fine quality glass sphere. These spheres became the lenses of his
pointed projections (taste buds) which were composed of microscopes, with the smallest spheres providing the highest
very small globules (bacteria). Separately, he was curious magnifications. The lenses were of exceptional optical quality
why pepper and other spices (ginger, nutmeg and cloves) had such potent tastes. He examined pepper and had magnifying powers ranging from x50 to x275 (possibly
water, an infusion that had been sitting for three weeks he was astonished to see many very small up to x500). His microscopes consisted of a single spherical lens
organisms that he called animalcules (again, bacteria, typically 1-2m in diameter). He then looked for mounted between two metal plates as seen in the illustration.
animalcules in other places such as in plaque on his teeth: I have mixed it with clean rain water, in which The short (about 1 millimeter) focal lengths of the lenses would
there were no animalcules, and I almost always saw with great wonder that there were many very little have necessitated placing the eye almost in contact with the
animalcules, very prettily amoving. lens.
page 11

1668 Anton van Leeuwenhoek invention of the simple (single lens) microscope, discovery of bacteria
Variolation (also known as
Inoculation) was introduced to the
west by Lady Mary Wortley Montagu,
who witnessed it being practiced by
physicians in Constantinople, and
was greatly impressed: she had lost
a brother to smallpox and her own
famous beauty had been marred by a
bout with the disease. She was eager
to spare her children similar suffering,
and had them inoculated while in
Turkey. On her return to London,
she enthusiastically promoted the
procedure, but encountered a great
deal of resistance from the medical
establishment both because it was
an Oriental process and because
of her gender. However, the British
royal family had their own children
inoculated, furthering acceptance.
She wrote: I am going to tell you a
thing, that will make you wish yourself
here [Turkey]. The small-pox, so
fatal, and so general amongst us, is
here entirely harmless. There is a
set of old women, who make it their
business to perform the operation...;
they make parties for this purpose...;
the old woman comes with a nut-shell
full of the matter of the best sort of
small-pox. She opens that vein which
you offer to her and with a needle
puts into the vein as much matter as
can lie upon the head of her needle.
The children or young patients are in
perfect health to the eighth day. Then
the fever begins to seize them, and
they keep their beds for two days.... I
am well satisfied of the safety of this
experiment, since I intend to try it on
my dear little son. I am patriot enough
to take the pains to bring this useful
Lady Mary Wortley Montagu (1689-1762) invention into fashion in England...

page 12

1718 Lady Mary Wortley Montagu introduction of variolation to western countries


Carolus Linnaeus (1707-1778)

Carolus Linnaeus, is called the Father of Taxonomy, for his system for naming, ranking, and classifying organisms that is still in wide use today (with many changes). The first
edition of Systema Naturae was printed in the Netherlands in 1735. It was a twelve page work. By the time it reached its 10th edition (1758), the multivolume work included
4,400 species of animals and 7,700 species of plants. In it, the unwieldy names in use at the time were supplemented with concise and now familiar binomials, composed of
the generic name, followed by a specific epithet. Higher taxa were constructed and arranged in a simple manner based on shared characters genera were grouped into orders,
orders into classes, and classes into kingdoms. In 1939, Francis O. Holmes first tried to extend the Linnaean sytsem to the viruses: he divided the viruses into three groups
under one order, the Virales the Phaginae (bacteriophage), Phytophaginae (plant viruses) and Zoophaginae (animal viruses). He extended his notion as a supplement to the
1948 edition of Bergeys Manual of Determinative Bacteriology. However, for the most part his taxa were based on clinical signs and pathology in animal hosts and lesions
in infected plants, not on properties of the viruses themselves. Even in 1948 this was not considered appropriate. Then, as described later in this book, in 1962 Andr Lwoff,
Robert Horne and Paul Tournier developed the first comprehensive virus classification system based on virion properties except for its Latinized nomenclature, it is the basis
for the current system managed by the International Committee on Taxonomy of Viruses.
Holmes FO, Proposal for extension of the binomial system of nomenclature to include viruses. Phytopathology 1939;29:431-436
page 13

1735 Carolus Linnaeus development of the hierarchical system for the classification of all organisms
Spallanzanis great experiment
refuting spontaneous generation
The debate over spontaneous generation continued for centuries. In 1745, John Needham,
an English clergyman, did what he considered the definitive experiment. Everyone knew
that boiling killed microorganisms, so he boiled chicken broth, put it into a flask, sealed it,
and waited - sure enough, microorganisms grew. Needham claimed victory for spontaneous
generation.
The Italian priest, Lazzaro Spallanzani, was not convinced, and he suggested that perhaps the
microorganisms had entered the broth from the air after the broth was boiled, but before it was
sealed. To test his theory, he placed the chicken broth in a flask, sealed the flask, drew off the
air to create a partial vacuum, then boiled the broth. No microorganisms grew. Proponents of
spontaneous generation argued that Spallanzani had only proven that spontaneous generation
could not occur without air.
The theory of spontaneous generation was finally laid to rest in 1859 by Louis Pasteur. The
French Academy of Sciences sponsored a contest for the best experiment either proving or
Lazzaro Spallanzani (1729-1799) disproving spontaneous generation. Pasteurs winning experiment was a variation of the
methods of Needham and Spallanzani. He boiled meat broth in a flask, heated the neck of the
flask in a flame until it became pliable, and bent it into the shape of an S. Air could enter the
flask, but airborne microorganisms could not - they would settle by gravity in the neck. As
Pasteur had expected, no microorganisms grew. When Pasteur tilted the flask so that the broth
reached the lowest point in the neck, where any airborne particles would have settled, the broth
rapidly became cloudy with microbes. Pasteur had both refuted the theory of spontaneous
generation and convincingly demonstrated that microbes are everywhere even in the air.
page 14

1765 Lazzaro Spallanzani first experiments refuting the theory of spontaneous generation
In recent years, there has been a growing recognition that Benjamin Jesty was one of the first to
vaccinate against smallpox. In fact, the use of smallpox and cowpox materials was rather widely
known among country physicians in 18th century England. When smallpox was present in Jestys
locality in 1774, he was determined to protect his family. Jesty used lesion material from cattle
suffering from cowpox, transferring the material with a small lancet to the arms of his wife and
two sons. The vaccinees remained free of smallpox, although they were exposed on numerous
occasions in later life. However, this does not diminish Edward Jenners accomplishments. It was
Jenners relentless promotion and devoted research of vaccination that led the way to the control
of smallpox globally.

Benjamin Jesty (1737-1816)


page 15

1774 Benjamin Jesty vaccination against smallpox using material from cowpox lesions
Edward Jenner Vaccinating James Phipps, 1796
painting by Gaston Melingue, 1879, Academie Nationale de Medecine, Paris
Edward Jenner (1749-1823)
In Jenners time, smallpox was greatly feared, as one-third of those who contracted the disease died, and those
who survived were often badly disfigured. Noting the common observation that milkmaids did not generally
get smallpox, Jenner theorized that contact with the lesions of cowpox on the teats of infected cows protected
them from smallpox. In 1796, Jenner tested his hypothesis by inoculating James Phipps, an eight year old boy
(the son of Jenners gardener), with material from cowpox lesions on the hand of Sarah Nelmes, a milkmaid
who had been infected with cowpox from a cow named Blossom. This produced a fever and some uneasiness
in James Phipps, but no great illness. Later, he injected the boy several times with material from smallpox
lesions no disease followed and he went on to test his method successfully on a series of 23 subjects.
[There is no mention whether Jenner knew that smallpox and cowpox material had been used for some years
to prevent smallpox.] Jenners unique contribution was not that he inoculated a few persons with cowpox Edward Jenner
virus, but that he then proved they were immune to smallpox. The medical establishment, cautious as ever, An Inquiry into the Causes and
considered his findings for some time before accepting them, but in time vaccination became universal, even Effects of th e Variol Vaccin
leading to the eradication of smallpox globally the first human disease to be eradicated globally. (London: Sampson Low, 1798)
page 16

1796 Edward Jenner application of cowpox virus for vaccination against smallpox
Edward Jenner vaccinating his son, who is held by his wife, while the maid prepares for her vaccination
Water-colored engraving by Jean Claude Manigaud (1825-?) from a painting by Edouard Jean Conrad Hamman (1819-1888). Wellcome Images.

page 17

1796 Edward Jenner application of cowpox virus for vaccination against smallpox
The Cow-Pock-or-the Wonderful Effects of the New Inoculation!-vide. the Publications of ye Anti-Vaccine Society.
Print (hand colored engraving) published in 1802 by H. Humphrey, St. Jamess Street, London. The British satirist James Gillray (1756-1815)
caricatured a vaccination scene at the Smallpox and Inoculation Hospital at St. Pancras, London, showing Edward Jenner vaccinating a frightened
young women, and cows emerging from different parts of peoples bodies. The cartoon was inspired by the controversy over inoculating against the
dread disease, smallpox. The inoculation agent, cowpox vaccine, was rumored to have the ability to sprout cow-like appendages. A serene Edward
Jenner stands amid the crowd. A boy next to Jenner holds a container labeled VACCINE POCK hot from ye COW; papers in the boys pocket are
labeled Benefits of the Vaccine. The tub on the desk next to Jenner is labeled OPENING MIXTURE. The painting on the wall depicts worshippers
of the Golden Calf. page 18

1796 Edward Jenner application of cowpox virus for vaccination against smallpox
In 1793, John Hunter (1754-1809) and
Samuel Argent Bardsley (1764-1851) each
published papers suggesting experiments to
determine the mode of rabies transmission,
including the inoculation of dogs and other
species with saliva from rabid dogs and even
from an hydrophobic human patient. The
inoculated animals were to be observed,
recording progress of the disease, and of the
length of time beyond which excision of the
tissue around the inoculation site could no
longer prevent development of the disease.

In 1804, Georg Gottfried Zinke (1771-1813),


carried out all the experiments suggested by
Hunter except for using saliva from a human
patient he successfully produced rabies in
healthy dogs, cats and rabbits by inoculating
saliva from rabid dogs. The experiment with
saliva from a human case was carried out in
1821 by Franois Magendie (1783-1855). At
the same time William Youatt (1776-1847)
showed that the rabbit was a particularly
useful experimental animal.

In 1879, Pierre-Victor Galtier (1846-1908),


Pierre-Victor Galtier professor at the Ecole Vtrinaire de Lyon,
reintroduced the rabbit into rabies research
(1846-1908) he transmitted rabies to rabbits and from
rabbit to rabbit. Pasteur was aware of the
work of Galtier before he started his work on
rabies in 1880 he had visited Lyon, where
Jean-Baptiste Auguste Chauveau (1827-1917)
John Hunter (1754-1809) had introduced him to Galtiers publications.
Galtier received many honors for his work
[There were two nearly contemporary John and was nominated for a Nobel Prize in 1908,
Hunters, the other the famous surgeon and but he died a few months before the Nobel
anatomist (1728-1793), who was a mentor and Commissions decision.
colleague of the John Hunter cited here.]
Wilkinson L. The development of the virus
concept as reflected in corpora of studies
on individual pathogens. 4. Rabies two
millennia of ideas and conjecture on the
aetiology of a virus disease. Medical History
Plaque honoring Galtier, Langogne, France 1977;21:15-31.
page 19

1793 John Hunter, Pierre-Victor Galtier, Georg Zinke, others beginnings of experimental pathology
In 1804, Georg Gottfried Zinke (1771-1813) first transmitted rabies from a rabid dog to a normal one, and from
a dog to a rabbit and a hen, by injection of saliva. This proved that the disease was infectious.
By 1826, Franz Christian Karl Krugelstein (1779-1864) wrote a full account of rabies, with a bibliography of
300 entries. But the nature of species susceptibility was unclear until Pierre-Victor Galtier demonstrated the
transmission from dog to rabbit to rabbit, in series. He then immunized sheep by inoculating rabid saliva
intravenously. This did not produce the disease, but interestingly, protected the animals from the effects of a
subsequent inoculation. His work aroused the interest of Louis Pasteur who with Charles Chamberland, Emile
Roux and Louis Thuillier wrote the first of their papers in 1881, heralding the beginning of Pasteurs studies on
rabies. It first lodges and multiplies in the spinal cord and brain.
Zinke GG. Neue ansichten der hundswuth, ihrer ursachen und folgen, nebst einer sichern behandlungsart der von tollen
thieren gebissenen menschen. Jena: CE Gabler; 1804.

Krugelstein FCK. Die geschichte der hundswuth und der wasserscheu und deren behandlung. Gotha: In der Henningsschen
Buchhandlung; 1826.

Galtier P-V. tudes sur la rage. Ann Med Vet. 1879;28:627-639.

Galtier V. Les injections de virus rabique dans le torrent circulatoire ne provoquent pas lclosion de la rage et semblant
confrer limmunit. La rage peut tre transmise par lingestion de la matire rabique. C R Acad Sci (Paris) 1881;93:284-285.

Wilkinson L. The development of the virus concept as reflected in corpora of studies on individual pathogens. 4. Rabies
two millennia of ideas and conjecture on the aetiology of a virus disease. Medical History 1977;21:15-31.

Emile Roux trephining a rabbits


skull, for rabies research, while
Louis Pasteur observes

page 20

1793 John Hunter, Pierre-Victor Galtier, Georg Zinke, others beginnings of experimental pathology
Philadelphias yellow fever epidemic of 1793 was among the largest in U.S. history,
with nearly 5,000 deaths (10% of the population). As the number of deaths began
to climb, 20,000 citizens fled to the countryside, including President George
Washington, Thomas Jefferson, and other members of the fledgling federal
government. Another severe epidemic occurred in 1798 by this time, the
advantages of flight were clear all but 7,000 residents left the city.
Benjamin Rush believed that yellow fever was caused by unsanitary conditions,
especially those of the docks, sewage system, and rotting vegetables such as rotting
coffee on the Arch Street wharf. He concluded that the illness was not transmitted
from human to human but by putrid exhalations in the atmosphere. He also
recognized that weather played a part. Though many people wanted to blame newly
arrived Saint Domingue revolution refugees, Rush instead blamed the sanitary
conditions of the city and implored residents to clean up the city so as to not entrail
the disease upon future generations.
page 21

1793 Benjamin Rush account of the great yellow fever epidemic in Philadelphia, 1793
Benjamin Rush (1746-1813), at age 37
by Charles Willson Peale, 1783
[Winterthur Museum, used with permission]

page 22

1793 Benjamin Rush account of the great yellow fever epidemic in Philadelphia, 1793
Establishment of the U.S. Public Health Service [first as the
Marine Hospital Service (1798-1902), then the Public Health
and Marine Hospital Service (1902-1912), and finally as the
Public Health Service (1912-present)]

page 23

1798 Founding of the U.S. Public Health Service, first as the Marine Hospital Service
John Pintard (1759-1844)

At The Gates: Our Safety Depends


Upon Official Vigilance
Harpers Weekly, Sept 5, 1885. The specters of cholera,
yellow fever, and smallpox recoil in fear as their way
through the Port of New York is blocked by a barrier on
which is written quarantine and by an angel holding a
sword and shield on which is written cleanliness. The kind of assisted emigrant we can not afford to admit.
Stephen Smith (1823-1922) Puck, 1883
In 1793, when a yellow fever epidemic killed 5,000 people in Philadelphia, doctors in New York organized a committee to prevent ships from Philadelphia from entering
New York harbor. These quarantine efforts may have helped keep the disease away that year but in 1795, 1799, and 1803, yellow fever felled thousands in New York. These
epidemics lent weight to the view that sanitary measures, and not just quarantine, were needed to curb the disease. The citys Common Council determined that it needed
greater authority over local sanitation, so in 1805 it passed an ordinance to create the first New York City Board of Health. John Pintard (1759-1844) began collecting mortality
statistics for New York in 1802, and two years later, the city appointed him its first City Health Inspector to keep official mortality statistics. In the 1805 City Inspectors Report,
he wrote that the public health system was designed to furnish data for reflection and calculation, in order that the awful malady of yellow fever could one day become
more controllable and less mortal. That year, there had been 600 reported cases and 262 deaths from yellow fever. This Board of Health eventually failed and had to be re-
established in 1866. This was led by Stephen Smith (1823-1922); again, it was the first such public health agency in the U.S. Smith later founded the American Public Health
Association. page 24

1805, 1866 John Pintard, others founding of the New York City Department of Health, the first in the U.S.
John Dollond Giovanni Battista Amici Joseph Jackson Lister Ernst Karl Abbe
(1706-1761) (1786-1863) (1786-1869) (1840-1905)
(the father of Joseph Lister)

Early microscopes had severely limited resolution caused by spherical and chromatic aberration the understanding of cells and cellular
pathology could not have occurred without improvements. Such improvements started in the 1730s with Chester Moore Hall, George
Bass, John Dollond and in the early 1800s with Giovanni Battista Amici and Vincent and Charles Chevalier who dealt with chromatic
aberration. The solution was to use a doublet lens, composed of two lenses made from glasses with different refractive indices so that
red and blue light waves were focused at the same point. In 1830, Joseph Jackson Lister solved the problem of spherical aberration (light
bends at different angles depending on where it hits the lens) by placing lenses at precise distances from each other. In 1866 Carl Zeiss
recruited Ernst Abbe, who laid out the mathematical framework of what would become the modern computational optics approach, and
who invented a condenser that controlled the diameter of the beam of incoming light and focused it on the specimen. Combined, these
developments allowed the manufacture of microscopes that could magnify 500 times with a very high quality image.

Lister type microscope


page 25
~1830

1830> John Dollond, Ernst Abbe, others advances in microscopes, achromatic lenses, Abbe condenser, light sources
Charles Chevalier Wilhelm His Rudolph Jung Charles Sedgwick Minot
(1804-1859) (1831-1904) (1845-1900) (1852-1914)
The first microtome was devised around 1790;
it consisted of a wooden device with a berth to
accommodate the specimen and a flat surface to slide
a razor against the specimen surface. The term
microtome was given to these instruments by Charles
Chevalier, who along with Johannes Purkinje (1787-
1869), perfected the instrument around 1835. Finally,
starting in 1865, precision mechanical devices were
developed and used by Wilhelm His, Charles Sedgwick
Minot, Adam Pfeiffer, and others. They consisted of a
metal stage holding a paraffin or collodion tissue block
and a mechanical swing holding the blade in precise
alignment. Rudolph Jung, a precision engineer and the
founder of Leitz, together with the pathologist Rudolf
Thoma (1847-1923) developed the first commercially
produced microtome. Carl Zeiss (1816-1888) was also
making microtomes by the 1870s. The famous Cambridge
rocking arm microtome, designed by Horace Darwin
(1851-1928; son of Charles) in 1885, was produced until
recently.
page 26

1835 Charles Chevalier, others development of the microtome, key to the founding of cellular biology/pathology
Theodor Schwann (1810-1882) Matthias Schleiden (1804-1881)
In 1838, Theodor Schwann and Matthias Schleiden were enjoying after-dinner coffee and talking about their studies on cells. When Schwann heard Schleiden describe
plant cells with nuclei, he was struck by their similarity to cells he had observed in animal tissues. The two scientists went immediately to Schwanns lab to look at his slides.
Schwann published his book on animal and plant cells the next year, a treatise devoid of acknowledgments of anyone elses contribution, including Schleidens. He made three
conclusions: (1) The cell is the unit of structure, physiology, and organization in living things. (2) The cell retains a dual existence as a distinct entity and a building block in the
construction of organisms. (3) Cells form by free-cell formation, similar to the formation of crystals (spontaneous generation) [Not true! The correct interpretation of cell
formation by division was finally enunciated in Rudolph Virchows 1858 epigram: Omnis Cellula e Cellula (every cell originates from another existing cell like it).]
page 27

1835 Theodor Schwann, Matthias Schleiden, others development of concept that organisms are composed of cells
John Snow (1813-1858)
John Graunt, William Farr, Louis-Ren Villerm, Peter Ludwig Panum, Mitchell Pruden, Hermann Biggs, and others are recognized for their
varied contributions to the founding of medical epidemiology, but it is John Snow who is most often given the title Father of Epidemiology.
This title stems from his two classic studies of the London cholera epidemic of 1853-1855. Snows first study concerned the Broad Street pump
outbreak of 1854, which killed more than 600 people in the Soho region of London. He used skilled reasoning, interviews with inhabitants,
graphs, and the now famous spot map locating cases to demonstrate the impact of contaminated water coming from the Broad Street pump.
His second study compared London neighborhoods receiving water from two different water companies. One company relied on inlets on the
upper River Thames, away from urban pollution, and the other company relied on inlets in the heart of London, where sewage contamination
was apparent. Snow compared the effect of contaminated water in two nearly equivalent populations, and suggested intervention strategies
to control the epidemic. His ideas and observations, including innovative disease maps, were published in his book, On the Mode of
Communication of Cholera (1849 and 1855). [The causative agent, Vibrio cholerae, was initially discovered in 1854 by Filippo Pacini, but his
discovery was not widely known until Robert Koch rediscovered it thirty years later.]
The Broad Street Pump
page 28

1838 John Snow, John Graunt, William Farr, others founding of medical epidemiology
Friedrich Gustav Jakob Henle, anatomist,
pathologist, and eminent scholar, was
an important figure in the development
of modern medicine. For example, he is
credited with the discovery of the loop
of Henle in the kidney. Henle developed
the concepts of contagium vivum and
contagium animatum [Von den Miasmen
und Kontagien (On Miasma and
Contagia)] in 1840, following the ideas of
Girolamo Fracastoro and Agostino Bassi
(who discovered that the muscardine
disease of silkworms was caused by a
living, very small, parasitic organism, a
fungus that would be named Beauveria
bassiana in his honor, and who in 1844
extended his findings to notions that
microscopic organisms also caused
human diseases). Henle thus laid the
groundwork for our understanding that
microorganisms are the cause of many
diseases. He did not find any bacteria
as etiologic agents this was achieved
by his student Robert Koch and others
including Louis Pasteur. Henle and
Koch, together with Friedrich Loeffler,
developed the fundamental rules for
proving the etiologic association between
an infectious agent and a specific disease:
the Henle-Loeffler-Koch Postulates.
Friedrich Gustav Jakob Henle
(1809-1885)
Sculpture of The Loop of Henle
Honoring Henle at
der Universitt Gttingen
page 29

1840 Friedrich Gustav Jakob Henle father of the germ theory, development of the Henle-Loeffler-Koch Postulates
Molluscum contagiosum
Skin lesion filled with poxvirus
thin section electron microscopy

Molluscum contagiosum lesion,


skin biopsy
H&E

In 1841, William Henderson (1810-1872), a pathologist at the Royal Infirmary of Edinburgh, wrote in the Edinburgh Medical & Surgical Journal about molluscum
contagiosum, which to him appeared to be among the rarest of cutaneous diseases. Within a transverse section of the wall of a lesion, he described and illustrated the
microscopic appearance of globular bodies as follows:
the wall consists of parallel cells projecting toward the interior cavity from an investing tegument or cyst of much delicacy. Within their parallel cells are exhibited small
globular bodies in considerable abundance, seen the most distinctly toward the inner extremities of the parallel cells. It is of these globular bodies that the atheromatous
matter which can be squeezed from the tubercles chiefly consists. The globules vary in size and contain nucleoli, some of which appear also in the fluid in a separate state.
These globular bodies are from the 1400th to the 3000th part of an inch in diameter.
In that same year, 1841, Dr. Robert Paterson (1814-1889), a physician of the Leith Dispensary, also found, microscopically, globular bodies within lesions of molluscum
contagiosum. This was the first description of virus-infected cells, a forerunner of the science of viral pathology.
Henderson W. Notice of the molluscum contagiosum. Edinburgh Medical & Surgical Journal 1841;56:213-218 and 279-288.
page 30

1841 William Henderson discovery of the inclusion body of molluscum contagiosum virus
Ignaz Philipp Semmelweis Oliver Wendell Holmes, Sr. Joseph Lister
(1818-1865) (1809-1894) (1827-1912)
In 1843, Oliver Wendell Holmes, Sr. investigated the circumstances surrounding puerperal (or childbed) fever and concluded that it was transmitted from patient to patient by
doctors, nurses and midwives, via their hands and clothing. He advocated the washing of hands with strong soap and chlorinated lime solution. His essay on the subject took
a strong line against opinions then prevailing (Holmes OW. The contagiousness of puerperal fever. New England Quarterly Journal of Medicine and Surgery 1842-1843;1:503-
530). Ignaz Philipp Semmelweis independently discovered at about the same time that the incidence of puerperal fever in obstetrical clinics could be drastically cut by the use
of hand washing with chlorinated lime solution. Puerperal fever was common in mid-19th-century hospitals, with mortality rates of 10-35%. Semmelweis worked in the Vienna
General Hospitals First Obstetrical Clinic, where doctors wards had three times the mortality rates of midwives wards. He published a book, The Etiology, Concept and
Prophylaxis of Childbed Fever, in which controlled quantitative studies of the value of disinfection were included. Joseph Lister was concerned about frequently fatal wound
infections that followed surgical procedures, and, in search of solutions to this problem, he studied the work of European bacteriologists, notably Louis Pasteur. Lister thought
that bacteria caused postoperative infections, and although the connection between bacteria and infection had not been confirmed at that time, he understood that bacteria
could be killed by antiseptics. He came up with the idea of using carbolic acid for preoperative cleansing of his and his assistants hands. He also sprayed carbolic acid in the
air in the operating room and on the surgical incision site. His dramatic beneficial results were reported in The Lancet in 1867 (Lister J. Illustrations of the antiseptic system of
treatment in surgery. The Lancet 1867;2:668), and soon transformed the practice of surgery into a relatively safe procedure for many conditions including the management of
childbirth.
page 31

1843 Ignaz Semmelweis, Oliver Wendell Holmes, Joseph Lister development of methods of hygiene/disinfection
Lister spraying carbolic acid over a surgical incision site:
While he was a professor of surgery at the University of
Glasgow, Lister became aware of the work of Louis Pasteur,
showing that microbial growth could occur under anaerobic
conditions. Pasteur suggested three methods to eliminate the
microorganisms responsible for gangrene: filtration, exposure
to heat, or exposure to chemical solutions it was the latter
that interested Lister. He confirmed Pasteurs conclusions with
his own experiments, which led him to develop antiseptic
techniques for wounds. Carbolic acid (phenol) had been in
use as a means of deodorizing sewage, so Lister tested it,
using spraying devices for surgical incision sites, and soaking
dressings with it. He found that the carbolic acid solution
remarkably reduced the incidence of gangrene. He published
a series of articles on the Antiseptic Principle of the Practice
of Surgery in 1867, immediately influencing medical/surgical
practice everywhere.

Ignaz Philipp Semmelweis: In the 1840s, puerperal or childbirth fever, a bacterial


infection occurring after childbirth, killed up to 30% of women who gave birth in
hospitals. Women who gave birth at home were relatively unaffected. Semmelweis, an
assistant professor on the maternity ward of the Vienna General Hospital observed that
women examined by student doctors who had not washed their hands had very high death
rates. When a colleague who had received a scalpel cut died of infection, he concluded
that puerperal fever was septic and contagious. He ordered students to wash their hands
with chlorinated lime before examining patients; as a result, the maternal death rate was
reduced from 12% to 1% in 2 years. Shortly afterward, as a professor of obstetrics at the
University of Pest Hospital, he enforced antiseptic practices and reduced the death rate
from puerperal fever to 0.85%.
page 32

1843 Ignaz Semmelweis, Oliver Wendell Holmes, Joseph Lister development of methods of hygiene/disinfection
Louis Pasteur
(1822-1895)
Louis Pasteur established the microbiologic/virologic/infectious
disease sciences, first in 1857 by discovering the specificity of microbial
fermentations (wine, beer, cheese), then in 1865 by extending the
concept to infectious diseases of silkworms, and finally between 1877
and 1895 by extending the concept to animal and human infectious
diseases. His early infectious disease work centered on septic war
wounds; he then turned to anthrax and other bacterial diseases,
and lastly to rabies. In each instance, he moved quickly from studies
aimed at discovering the causative agent to the development of
specific intervention. Clearly, Pasteur deserves his title of Father of the
Microbiologic/Virologic/Infectious Disease Sciences.
page 33

1857> Louis Pasteur father of microbiology, father of virology, father of the infectious disease sciences
Jubil de Pasteur a la Sorbonne 27 December 1892 [Painting by Jean Andr Rixens (1846-1924)] Pasteurs seventieth birthday was the occasion of a national
holiday. The celebration was held in the great hall of the Sorbonne. Pasteur entered on the arm of the President of the Republic, Sadi Carnot. Both were wearing the grand cor-
don of the Lgion dHonneur. Pasteur was greeted by an immense ovation; he was too weak to speak to the delegates who had gathered from all over the world, so his address
was read by his son: Gentlemen, you bring me the greatest happiness that can be experienced by a man whose invincible belief is that science and peace will triumph over igno-
rance and war. Have faith that in the long run the future will belong not to the conquerors but to the saviors of mankind. The painting shows Joseph Lister coming forward
to greet his old friend. He said: It is my great privilege to convey to you, tributes, thanks and respect from all involved in medicine and surgery; it is true to say that, of all people
in the world today, medical sciences owe you the most. For centuries, infectious diseases have been shrouded, as it were under a dark curtain. In discovering the microbial origin of
disease you have raised that dark curtain! page 34

1857> Louis Pasteur father of microbiology, father of virology, father of the infectious disease sciences
Die Cellularpathologie (1858)
English translation (1860)
Virchow R. Die cellularpathologie in
ihrer begrundung auf physiologische und
pathologische gewebelehre. Berlin: August
Hirschwald; 1858.
Virchow RLK. Cellular pathology. London:
John Churchill; 1860.

Rudolf Ludwig Karl Virchow (1821-1902)


Virchow is credited with many important advances in medical science: he was the first to accept the work of Robert Remak who showed that the origin of cells was the division
of preexisting cells (Virchows 1858 epigram: Omnis Cellula e Cellula (every cell originates from another existing cell like it.). Virchow founded the fields of cellular pathology
and comparative pathology, always urging the use of the microscope (think microscopically). Virchow astutely observed the interdependence of animal and human diseases
and noted that each relies on a common pool of medical and scientific knowledge this led him to coin the term zoonosis to describe diseases transmitted from animals to
humans. He stated, Between animal and human medicine there is no dividing line nor should there be. The object is different but the experience obtained constitutes the basis
of all medicine.
page 35

1858> Rudolf Virchow father of cellular (microscopic) pathology and comparative pathology
HMS Beagle in Sydney Harbor
On the voyage of HMS Beagle (1831 to 1836) to carry out surveys to produce Admiralty
navigation charts of the coastline of South America and Australia, Charles Darwin, then
22 years of age, as guest naturalist, spent as much time as possible exploring on land; this
experience provided the basis for ideas which he developed into his theory of evolution by
natural selection. HMS Beagle painting by Ron Scobie.
Darwin: In October 1838, that is, fifteen months after I had begun my systematic enquiry, I happened
to read for amusement Malthus on Population, and being well prepared to appreciate the struggle
for existence which everywhere goes on from long-continued observation of the habits of animals and
plants, it at once struck me that under these circumstances favourable variations would tend to be
preserved, and unfavourable ones to be destroyed. The result of this would be the formation of new
species. Here, then, I had at last got a theory by which to work...
Darwins book was half completed when, in 1858, he received a paper from Alfred Russell
Wallace (1823-1918) describing natural selection. Shocked that he had been forestalled,
Darwin sent it on to Charles Lyell, suggesting that it be sent to any journal that Wallace chose.
Lyell and Joseph Hooker decided on a joint presentation at the Linnean Society: there was
Charles Robert Darwin (1809-1882) little immediate attention to the presentation. Darwin then struggled for thirteen months to
Watercolor, painted by produce his big book, and Lyell arranged to have it published. On the Origin of Species proved
George Richmond in the late 1830s unexpectedly popular, with the entire stock of 1,250 copies subscribed when it went on sale in
[Darwin Museum at Down House, Downe, used with permission] 1859. In his book, Darwin set out one long argument providing his bases for what has been
called the single best idea anyone has ever had.
page 36

1859 Charles Darwin father of evolutionary science, concepts of common descent, gradualism, natural selection
Charles Robert Darwin (1809-1882)
Cropped version of a portrait from 1881 by John Collier (1850-1934), painted in 1883.
Darwins son, Erasmus, stated: The picture is a replica of the one in the rooms in the
In mid-July 1837 Darwin started his B
Linnaean Society and was made by Collier after the original. I took some trouble about it
notebook on Transmutation of Species, and
and as a likeness it is an improvement on the original.
on page 36 wrote, I think. This was his first
[National Portrait Gallery, London, used with permission]
evolutionary tree.
page 37

1859 Charles Darwin father of evolutionary science, concepts of common descent, gradualism, natural selection
When Charles Darwins On the Origin of Species went to press in 1859, viruses had One Long Argument: Charles Darwin and the Genesis of Modern Evolutionary
yet to be discovered it would be another 40 years before their discovery, and a Thought. Ernst Mayr, Harvard University Press, 1993:
century before virology research would provide a clear understanding of their genetic
make-up, how they replicate and how they cause disease. From our contemporary Ernst Mayr was a force in defining and elaborating Darwinism. One of his criticisms
perspective, perhaps the most unexpected finding has been the discovery of the of scholarly naysayers was that they did not deal with each of Darwins ideas in logical
degree to which viruses have been an evolutionary force, as pathogens affecting fashion, but rather confused one for another. Mayrs way of focusing arguments was
host (human and animal) evolution and as agents of horizontal transfer of genetic to dissect Darwins conceptual framework of evolution into five parts, only the whole
information (e.g., via mobile elements, transposons) and as such major contributors to worthy of being called Darwins Theory:
the variation upon which natural selection works. 1. Evolution as such. Living organisms [and viruses] are not constant or recently
created or perpetually cycling, but rather are forever changing.
The microbial world offers the most direct evidence of natural selection. The mass 2. Common descent. All groups of organisms, including animals, plants, and
of evidence of extraordinary biological diversity of the microbes and the viruses, microorganisms, are descended from a common ancestor, ultimately going back
at molecular, morphologic and phenotypic levels, has continued to increase, to a single origin of life on earth.
exponentially. Viruses have proven to be wonderful subjects for studying the forces 3. Multiplication of species. The origin of the enormous organic diversity seen on
and outcomes of genetic variation: competition, selection bottleneck events, etc. earth stems from the multiplication of species, either by splitting of species into
Their very rapid rate of mutation has been the substrate for amazing discoveries in daughter species or by budding, that is, by the establishment of geographically
genetics, genomics, functional genomics, etc. In some ways, this understanding of isolated founder populations that evolve into new species.
the mechanisms of variation has not been equaled by our understanding of the forces 4. Gradualism. Evolutionary change takes place through the gradual change of
driving natural selection per se we tend to oversimplify this, and evolutionary populations and not by the sudden (saltational) production of new species.
biologists tend to ignore haploid organisms (bacteria) and viruses which replicate 5. Natural selection. Evolutionary change comes about through the abundant
clonally. Many discoveries described later in this volume illustrate great progress, production of genetic variation in every generation. Few variants survive; those
and yet more mystery and more of a sense of complexity, in arriving at the point of that do are better adapted by virtue of their combination of inherited characters,
satisfying our understanding of virus ecology and viral disease pathogenesis and and these variants give rise to succeeding generations.
pathophysiology.

A typical caricature Hugo Rheinholds


of Darwin, Affe mit Schdel
The Hornet, (Ape with Skull),
a satirical magazine, 1893
1871

page 38

1859 Charles Darwin father of evolutionary science, concepts of common descent, gradualism, natural selection
President Abraham Lincoln Signs the Charter of the U.S. National Academy of Sciences
Artist: Albert Herter. This painting, hanging in the U.S. National Academy of Sciences Board Room, depicts President Lincoln signing the charter, on March 3rd,
1863. In his apocryphal scene the Academy founders look on. Founding Members (left to right): Benjamin Pierce, Alexander Dallas Bache (first president of the
Academy), Joseph Henry, Louis Agassiz, President Lincoln, Senator Henry Wilson, Admiral Charles Henry Davis, and Benjamin Apthorp Gould
[from U.S. National Academy of Sciences, used with permission]

The National Academy of Sciences was created to investigate, examine, experiment, and report upon any subject of science or art whenever called upon to do so by any
department of the government. The demands of the Civil War (1861-1865) were conducive to the formation of a scientific consulting body, especially to advise on new
weapons. A huriedly written bill for the incorporation of the National Academy of Sciences and naming 50 charter members was signed by President Lincoln the same day
that it was passed by the Congress. Although hailed as a great step forward in government recognition of the role of science in American civilization, the National Academy
of Sciences did not solve the problems facing a nation in its Civil War, nor did it centralize American scientific efforts. However, over the years it developed into the most
influential body of its kind and extended its reach into all aspects of science and society.
page 39

1863 Abraham Lincoln, others founding of the U.S. National Academy of Sciences
Pierre-Franois-Olive Rayer
(1793-1867)

Casimir Davaine (1812-1882)

In 1850, Casimir Davaine, along with French dermatologist Pierre Franois Olive Rayer (1793-1867),
discovered a certain microorganism in the blood of diseased and dying sheep. In the diseased blood,
Rayer and Davaine observed the bacillus which is now known as Bacillus anthracis, the causative
bacterium of anthrax. Soon afterwards, Rayer published an essay on anthrax, which contained the first
description of Bacillus anthracis.

In 1863, Davaine demonstrated that the anthrax bacillus could be directly transmitted from one animal
to another. He was able to identify the causative organism but was unaware of its true etiology. Later,
Robert Koch did considerable research on Bacillus anthracis, and discovered its ability to produce
resting spores that could stay alive in the soil for a long period of time and serve as a continuing
source of infection.
page 40

1863 Casimir Davaine first association of a specific infectious organism with a specific disease
Mendels first experiment

Gregor Mendel was an Augustinian priest, who gained posthumous fame as the founder
of the science of genetics for his study of the inheritance of certain traits in pea plants.
Mendel showed that the inheritance of these traits follows particular laws, which were later
named after him. The significance of Mendels work was not recognized until the turn of the 20th
century, when the his laws were independently rediscovered.
Mendel conducted his study in his monasterys two hectare garden; between 1856 and 1863
Mendel cultivated and tested some 29,000 pea plants. His experiments led him to make two
generalizations, the Law of Segregation and the Law of Independent Assortment, which later
became known as Mendels Laws of Inheritance.
Mendel did read his paper, Experiments on Plant Hybridization, at two meetings in Brnn, Gregor Johann Mendel (1822-1884)
Moravia in 1865 and published his work in 1866 in the Proceedings of the Natural History
Society of Brnn, but it had little impact and was cited only three times during the next thirty-
five years. Mendels work was re-discovered in 1900 (see Hugo de Vries ).
page 41

1865 Gregor Mendel father of genetics


Ludwig Karl Martin Leonhard
Albrecht Kossel (1853-1927)

Johan-Friedrich Miescher isolated various


phosphate-rich chemicals, which he called nuclein
(now nucleic acids), from the nuclei of white blood
cells in 1869 at Felix Hoppe-Seylers laboratory
at the University of Tbingen, Germany, thereby
paving the way for the identification of DNA as
the carrier of inheritance. The significance of the
discovery, first published in 1871, was not at first
apparent. Miescher and his students researched
much of the basic chemistry of the nucleic acids but
Johan-Friedrich Miescher (1844-1895) their function remained unknown. The importance
of Mieschers discovery was not apparent until
Albrecht Kossel carried out research on the
chemical structure of nuclein (i.e., DNA).

page 42

1868 Johan-Friedrich Miescher discovery and characterization of nucleic acids


Physician
Pathologist
Comparative Pathologist
Clinician
Teacher
Diagnostician
Bibliophile
Historian
Classicist
Essayist
Author

The Renaissance Man

William Osler (1849-1919)

Osler is credited with coining the term One Medicine to encompass the relationship
between human and veterinary medicine. Oslers philosophy and his most famous
work, The Principles and Practice of Medicine, did much to change the teaching and
practice of medicine from its historic didactic base to a clinical base, where students
learned from seeing and talking to patients. He established the first medical residency
program, and is credited with developing the concept of rounds. His approach changed
the teaching and practice of veterinary medicine in similar fashion.

Osler conducting Grand Rounds,


Johns Hopkins School of Medicine, 1900
page 43

1874 William Osler father of modern scholarly medicine


Koch in Africa, ~1904, seeking the cause of rinderpest

Robert Koch is credited with the first incontrovertible proof that a microorganism
can cause disease (Bacillus anthracisanthrax). After Casimir Davaine showed the
direct transmission of the anthrax bacillus between cows, Koch studied anthrax more
closely. He invented methods to purify the bacillus from blood and grow pure cultures.
He found that, while the anthrax bacillus could not survive outside a host for long,
it produces endospores that survive for many years. He developed bacterial staining
techniques, bacterial liquid and solid growth media (and agar plates thanks to the
advice of Angelina and Walther Hesse), and he developed the Petri dish, named after
its inventor, his assistant Julius Richard Petri. With these techniques, he was able to
discover the bacterium causing tuberculosis (Mycobacterium tuberculosis) in 1882.

With Friedrich Gustav Jakob Henle and Friedrich Loeffler, in 1884, he developed the
Henle-Loeffler-Koch Postulates (Kochs Postulates) outlining the criteria required to
establish a causal relationship between a microbe and a disease:
1. The microorganism must be found in abundance in all organisms suffering from
the disease, but should not be found in healthy animals. 2. The microorganism must
be isolated from a diseased organism and grown in pure culture. 3. The cultured
microorganism should cause disease when introduced into a healthy organism. 4. The
Heinrich Hermann Robert Koch (1843-1910) microorganism must be reisolated from the inoculated, diseased experimental host and
identified as being identical to the original specific causative agent.
page 44

1876 Robert Koch along with Louis Pasteur, the founders of microbiology
Patrick Manson Charles Louis Laveran Ronald Ross Giovanni Battista Grassi
(1844-1922) (1845-1922) (1857-1932) (1854-1925)

In 1877-1879, Patrick Manson (1844-1922) demonstrated that the mosquito was the host of the filarial worm Wuchereria bancrofti, the etiologic agent of filariasis; he
suggested that the infectious agent that causes malaria is also spread by a mosquito.
In 1880, Charles Louis Alphonse Laveran (1845-1922), a French army doctor working in the military hospital in Constantine, Algeria described parasites inside red blood cells
of people suffering from malaria; he proposed that malaria was caused by this protozoan, the first time a protozoan was identified as causing disease.
In 1898, Ronald Ross (1857-1932), working in India, proved that avian malaria is transmitted by mosquitoes.
In 1898, Giovanni Battista Grassi (1854-1925) described the entire developmental cycle of the plasmodium in the body of Anopheles mosquitoes and proved that human
malaria is transmitted by Anopheles mosquitoes.

All of these discoveries may have been influential in guiding the work of Walter Reed and his colleagues in their discovery of the first human virus, yellow fever virus, and its
transmission cycle involving Aedes aegypti mosquitoes. However, after the Yellow Fever Commission had made its discoveries in 1900, Walter Reed cited only four influences
that had guided the research: (1) the work of Carlos Finlay on the yellow fever vector mosquito, Aedes aegypti, (2) the work of Henry Rose Carter providing evidence of a
mosquito extrinsic incubation period, (3) the work of Theobald Smith and his colleagues proving that the protozoan agent of Texas fever (Babesia bigemina) was transmitted
by an arthropod (the tick, Boophilus annulatus), and (4) the work of Friedrich Loeffler and Paul Frosch showing that the agent of foot-and-mouth disease was an ultrafilterable
virus. It has been said that the failure of the early malaria research to have influenced the work of the Yellow Fever Commission in Havana was due to the slowness of
communication of scientific findings at the time, the proof that human malaria is transmitted by Anopheles mosquitoes having only come in 1898.

page 45

1877> Patrick Manson, Charles Laveran, Ronald Ross, Giovanni Grassi discovery of the etiologic agent of malaria
Stanford Emerson Chaille, known as
the Father of Hygiene and Health
Education in America. Chaille gained
fame as the head of the U.S. Havana
Yellow Fever Commission of 1879,
organized to study the disease following
the 1878 epidemic in New Orleans.

A national figure, Chaille was


spokesman for the establishment of
community sewerage and drainage
systems, street paving, pure water
supplies and mosquito control. He was
also instrumental in the establishment
of the National Board of Health, the
forerunner of the U.S. Department of
Health and Human Services.

Stanford Emerson Chaille (1830-1911)

Yellow fever dragging down Florida, 1888


Artist Matt Morgans drawing in
Frank Leslies Illustrated Newspaper
There were more than 400 deaths in Jacksonville in 1888
leading to widespread panic

Thanksgiving Mass At Father Matthews Yellow Fever Camp,


near Memphis, during the epidemic of 1878
page 46

1879 Stanford Chaille father of hygiene and health education in the United States
Some of the Most Devastating Yellow Fever Epidemics
in the United States
YEAR AREA
1690 New York
1693 Boston
1699 Charleston & Philadelphia
1702 New York
1706 Charleston
1732 Charleston & New York
1734 Virginia
1741 Virginia
1762 Philadelphia
1793 Philadelphia (~5,000 deaths)
1794 Philadelphia
1798 Philadelphia
1803 New York
1841 Nationwide
1847 New Orleans
1850 Nationwide
1852 New Orleans (~8,000 deaths)
1855 Nationwide
1878 New Orleans
1886 Jacksonville
1888 Florida
1905 New Orleans

During the yellow fever epidemic of 1878, fear and anxiety pervaded the country, in both the North and the South. Yellow fever evoked sympathy from many northerners,
and as they donated funds and prayed for southerners, the roots of post-Civil War national reconciliation were established. In the midst of their benevolence, northerners
consciously effaced memories of southerners as belligerents seeking to destroy the Union southerners now became stricken, honorable people in need. At the same time
southerners began to see northerners as fellow citizens. In effect, yellow fever helped open the way for southerners to accept national unity.
The 1878 epidemic began in New Orleans in May, likely with the arrival of a sailor infected in Havana. The disease spread in New Orleans during June, and the number of
victims began to multiply in frightening fashion from July onward. Cities were hardest hit: Memphis (5,000 of 40,000 residents died), New Orleans (>4,600 deaths) the disease
then struck large parts of Mississippi, Louisiana, Tennessee, Kentucky, Georgia, Ohio, and Missouri.
Local relief societies recognized that they could not handle the crisis on their own and pleaded for assistance. Southern requests for aid supplied northern newspapers and
magazines an opportunity not only to encourage the North to show compassion for the South, but also to describe southerners as full-fledged citizens of the nation. Leslies
Illustrated Newspaper maintained that sympathy must be made tangible with direct relief: These horrors should at least inspire a genuine sympathy with the suffering, and
induce on the part of all of us generous contributions . Donation boxes in public places served as physical proof of northern generosity to southerners as well as other
northerners. Leslies Illustrated Newspaper and Harpers Weekly regularly carried images of these donation boxes, allowing people to see themselves and others giving.
page 47

1690-1905 Yellow fever epidemics in the United States


Aedes aegypti
Finlay hypothesized that the common house mosquito (then
Stegomyia fasciata, now Aedes aegypti) transmitted yellow
fever by directly injecting the blood from an infected person
into a susceptible person. He did not appreciate the need for
an extrinsic incubation period in the mosquito. In retrospect,
at most only 1 of his 104 experiments from 1881-1898
demonstrated mosquito transmission of yellow fever. Many
thought that Finlay had disproved his own hypothesis. However,
upon their arrival in Havana in 1900, Walter Reed and his
colleagues were greatly influenced by Carlos Finlays hypothesis
and he helped them initiate their studies, especially by providing
mosquitoes for the transmission experiments.
Carlos Juan Finlay (1833-1915)

page 48

1881 Carlos Finlay initial experiments on the mode of transmission of yellow fever virus
Ilya Ilyich Mechnikov Paul Ehrlich Jules Bordet Emil Adolf von Behring
(Elie Metchnikoff) (1854-1915) (1870-1961) (1854-1917)
(1845-1916)

The founding of the science of immunology was celebrated by the Nobel Prize award to Elie Metchnikoff and Paul Ehrlich in 1908. Metchnikoff discovered phagocytosis by
macrophages and microphages as a critical host-defense mechanism and thus is considered the father of cellular innate immunity. Ehrlich described the side-chain theory of
antibody formation and the mechanisms of how antibodies neutralize toxins and induce bacterial lysis and thus is considered one of the fathers of humoral adaptive immunity.
Both Ehrlich and Metchnikoff provided a paradigm shift by developing concepts of how the host can deal with invading pathogens to overcome infectious diseases. Their ideas
evolved, however, on the shoulders of Pasteur and Koch, who had prompted another paradigm shift shortly before. Pasteur provided the ultimate disproof of spontaneous
generation by demonstrating absence of microbial growth after heat inactivation. Pasteurs conclusion that fermentation and putrefaction were the work of microorganisms
and that these processes occur not only in organic material but also in the living body laid the foundation for the germ theory of infectious disease. Koch clarified the
microbial etiology of anthrax, sepsis and tuberculosis. Bordets discovery that the bacteriolytic effect of acquired specific antibody is significantly dependent in vivo on the
presence of innate serum components which he termed alexine (which are now known as the component molecules of the complement system). This mechanism became
the basis for complement-fixation testing methods that enabled the development of serological tests for syphilis and many other diseases. von Behring developed the first
immunotherapeutic system, the highly effective serum therapy used to treat diphtheria. He also worked on the development of tetanus antitoxin with Emile Roux. The work of
these pioneers was quickly applied to the emerging science of virology in the first years of the 20th century.

page 49

1883> Elie Metchnikoff, Paul Ehrlich, others founding of immunology, hematology and chemotherapy
In 1881 and 1882, Louis Pasteur, Charles Chamberland,
Emile Roux and Louis Thuillier began their research toward
developing a rabies vaccine. They modified Pierre-Victor
Galtiers technique by inoculating nervous tissue from a rabid
dog directly into the brain of the next dog via trephination. By
successive passages in dogs, they obtained a virus of maximum
virulence and with a fixed incubation period of about 10
days. They estimated the degree of attenuation of brain tissue
from each passage. They then passaged the infectious agent
serially in rabbits. This final attenuation procedure consisted
of suspending the spinal cord of a rabid rabbit in a flask, in
a warm dry atmosphere, to achieve slow desiccation. They
succeeded in producing attenuated viruses of different
strengths, that is a standardized range of viruses, the weakest
of which could be used to prepare the first dose of a vaccine.
Inoculating dogs with a sequence of spinal cords of increasing
virulence rendered the recipient resistant to inoculation with
fully virulent virus.

Louis Pasteur (1822-1895)


Painting byAlbert Edelfeldt, 1885
[from Institut Pasteur, used with permission]
Pierre-Paul-Emile Roux (1853-1933)
page 50

1884 Louis Pasteur, Emile Roux, Charles Chamberland, Louis Thuillier development of rabies vaccine
Charles Chamberland (1851-1908)
Theodor Albrecht Edwin Klebs (1834-1913) and Ernst Tiegel, in 1871, found that the causative agent of anthrax would not pass through a filter made of unfired clay it
was a non-filterable bacterium. As early as 1876, Louis Pasteur, in collaboration with the French physicist Jules Joubert, used a plaster-of-Paris filter connected to a vacuum
pump to confirm Krebs diacovery. The use of ultrafilters to retain and concentrate bacteria became standard practice after 1884, when Charles Chamberland and Emile
Roux, experimenting with a broken clay pipe purchased from Chamberlands tobacconist, developed unglazed porcelain ultrafilters (candles, bougies de Chamberland) that
retained bacteria. Chamberland filters were produced in 13 types: L1 to L13; L1 filters having the coarsest pore size, L13 the finest.
Chamberland C. Sur un filtre donnant de leau physiologiquement pure. C. R. Acad. Sci. Paris 1884;99:247-248.
page 51

1884 Charles Chamberland development of the porcelain ultrafilter, key to the discovery of the viruses
From 1860 onward, Louis Pasteur made known his germ theory, but
skeptics, especially Henri Charlton Bastian, argued against it because
in some cases germs (bacteria) seemed to appear in sealed vessels after
heating to 100C, Pasteurs standard inactivating temperature. Charles
Chamberland, working in Pasteurs laboratory, showed that some germs
(bacterial spores) could withstand a temperature of 110-120C. In the
course of this work, betweem 1879 and 1884 Chamberland invented the
autoclave (lautoclave Chamberland), expanding upon a precursor known
as the steam digester created by Denis Papin in 1679. A Parisian firm
marketed Chamberlands Autoclave in 1884 and by the turn of the century Emile Roux using one of Charles Chamberlands new autoclaves
steam-jacketed autoclaves from which steam could be evacuated at the end [from Institut Pasteur, used with permission]
of the heating cycle were in use widely.

Pasteur L. Memoire sur les corpuscles organises qui existent dans latmosphere. Examen de la doctrine des generations spontanees.
Annales des Sciences Naturelles, 4th series. 1861;16:5-98.
Pasteur L, Joubert JF, Chamberland C. La thorie des germes et ses application la mdecine et la chirurgie. C R Acad Sci Hebd
Seances Acad Sci D 1878;86:1037-1043.
Chamberland C. Sur un filtre donnant de leau physiologiquement pure. C R Acad Sci Paris 1884;99:247-248.
page 52

1884 Charles Chamberland invention of the autoclave


Adolf Eduard Mayer (1843-1942) Tobacco mosaic disease, Mayers work, 1866
Adolf Mayer was a German agricultural chemist and director of the Agricultural Experiment Station at Wageningen in the Netherlands. He is credited as the first person to
transmit tobacco mosaic virus by using juice extracts from diseased plants as the inoculum to infect other plants. Mayer published a paper in 1886 describing tobacco mosaic
disease in detail. He performed chemical analyses of healthy and diseased leaves to see if a difference in nutrition could explain the disease. He investigated temperature,
light, fertilization, and looked for fungi or animal parasites. He tried to follow Kochs Postulates and was able to culture organisms from his extracts, but none of these would
reproduce the disease. He was left with the conclusion that the infectious agent was likely some sort of microorganism. Although Mayer came to the wrong conclusion about
his finding, his work laid the groundwork for the experiments of Dmitry Ivanovsky and Martinus Beijerinck, who showed that the etiologic agent of tobacco mosaic disease
would pass through an ultrafilter, that is, it is an ultrafilterable virus.
Mayer, A. Concerning the mosaic disease of tobacco. Die Landwirtschaftliche Versuchsstationen 1886;32:451-467. Translation published in English as Phytopathological
Classics Number 7 (1942). American Phytopathological Society Press. St. Paul, MN.
page 53

1886 Adolf Mayer the concept of transmissibility and the earliest concept of an ultrafilterable virus
John Brown Buist (1846-1915) was a Scottish pathologist and medical practitioner who devoted much of his professional life to
vaccination. For a time he was also medical superintendent of the smallpox hospital at Barrow-in-Furness in Scotland. In the book
he published in 1887 (Vaccinia and Variola: a Study of their Life-History), Buist described and illustrated minute bodies which
he observed in films of lymph from vaccinia and smallpox vesicles after prolonged staining with the aniline dye gentian violet.
He regarded these bodies as the infectious agents. Although he believed the tiny bodies he saw were spores, he was nonetheless
the first person to see (and photograph) a virus. Although nearly all viruses are invisible under the light microscope, vaccinia and
variola virions are just big enough to be visible when heavily stained or observed by dark-field light microscopy. Vaccinia and
variola virions are brick-shaped, about 200x200x250nm in size.

Variola (smallpox) virus


Negative contrast electron microscopy
page 54

1887-1922 John Buist, Edmund Cowdry discovery of elementary & inclusion bodies of vaccinia & variola viruses
Edmund Vincent Cowdry (1888-1975)
[from Rockefeller Archives, used with permission]
Edmund Cowdry was the founder of viral disease cytology and the person who initiated
our appreciation of inclusion bodies in cells infected by many viruses. But, his career at the
Rockefeller Institute and then Washington University School of Medicine was much more
diverse for example, here is a listing of the books he wrote over his long career: General
Cytology (1924), Special Cytology, Form and Functions of the Cell in Health and Diseases
(1928>) , Human Biology and Racial Welfare (1930), A Textbook of Histology (1934>),
Problems of Ageing (1939>), Microscopic Technique in Biology and Medicine (1943), Laboratory
The variety of poxvirus inclusion bodies
Cowdry E. The supravital staining of vaccine bodies.
Technique in Biology and Medicine (with Victor Emmel, 1964), The Care of the Geriatric
Journal of Experimental Medicine 1922;36:667-684.
Patient (1968>), Etiology and Prevention of Cancer in Man (1968), and Cancer Cells (1955).
page 55

1887-1922 John Buist, Edmund Cowdry discovery of elementary & inclusion bodies of vaccinia & variola viruses
Joseph J. Kinyoun (1860-1919)

The National Institutes of Health (NIH) was founded in 1887, within the Marine Hospital Service (MHS), the predecessor agency to the U.S. Public Health Service (PHS). The
MHS had been established in 1798 to provide for the medical care of merchant seamen. A clerk in the Treasury Department collected twenty cents per month from the wages
of each seaman to cover costs at a series of contract hospitals. In the 1880s, the MHS was charged by Congress with examining passengers on arriving ships for clinical signs
of infectious diseases, especially for cholera and yellow fever, in order to prevent epidemics. In the 1870s and 1880s scientists in Europe (Pasteur, Koch) presented compelling
evidence that microscopic organisms were the causes of several infectious diseases. In 1884, for example, Koch described a comma-shaped bacterium as the cause of cholera.
Officials of the MHS followed these developments with great interest. In 1887, they authorized Joseph J. Kinyoun, a young MHS physician trained in the new bacteriological
methods, to set up a one-room laboratory in the Marine Hospital on Staten Island, New York. Kinyoun called this facility a Laboratory of Hygiene in imitation of German
facilities and to indicate that the laboratorys purpose was to serve the publics health. Within a few months, Kinyoun had identified the cholera bacillus in suspicious cases
and used his microscope to demonstrate it to his colleagues as confirmation of their clinical diagnoses. In 1902, the Hygienic Laboratory was established in federal law as
the center for medical research within the federal government. The MHS was renamed the Public Health and Marine Hospital Service (PH-MHS), and the pathological and
bacteriological work as the Division of Pathology and Bacteriology was recognized by creating three new components that represented the most fruitful areas for research at
that time: the Divisions of Chemistry, Pharmacology, and Zoology. In 1912, another reorganization shortened the name of the PH-MHS to the Public Health Service (PHS or
USPHS). The National Institute of Allergy and Infectious Diseases (NIAID) was established at a unit of the NIH in 1955.
page 56

1887 Joseph Kinyoun founding of the National Institutes of Health


Sculpture of Jean-Baptiste Jupille
on the grounds of the Institut Pasteur
The Institut Pasteur was founded in 1887 by Louis Pasteur and his colleagues, whose experiments
led to the founding of the science of microbiology and virology. Pasteur discovered the principle of
food sterilization, which in the case of milk came to be known as pasteurization. His discoveries led
to the universal practice of surgical antisepsis. He also developed vaccination concepts and methods
to control several bacterial diseases and the viral disease rabies. As soon as the Institut was created,
Pasteur established five departments, each with a distinguished director: Emile Duclaux (general
microbiology research), Charles Chamberland (research applied to hygiene), Ilya Ilyich Mechnikov
(microbiology and immunology research), Jacques-Joseph Grancher (rabies research), and Emile Roux
(microbiology and virology research). One year after the inauguration of the Institut Pasteur, Roux set
up the first course of microbiology ever taught, then entitled Cours de Microbie Technique (course in
microbiological research techniques).
page 57

1887 Louis Pasteur and colleagues founding of the Institut Pasteur


Hermann Michael Biggs (1859-1923) Hester Street, 1900, a time of cholera and smallpox outbreaks
Hermann Michael Biggs was the founder of public health laboratory sciences in the United States. He was born in the village of Trumansburg, N. Y. and attended Cornell
University and Bellevue Hospital Medical College. During his internship at Bellevue, he developed an interest in histopathology and the new science of bacteriology. In 1884
he visited Robert Kochs laboratory in Germany and in 1885 he visited Louis Pasteurs laboratory in France. When he returned from his first European trip, he became a senior
member of the newly established Carnegie Laboratory at Bellevue, devoted to bacteriology. This was the first bacteriological laboratory in the United States to apply this
science directly to public health. When a division of bacteriology was created within the New York City Department of Health in 1892 in response to the threat of cholera,
Biggs was appointed chief inspector and director of the laboratory. Biggs became the leading advocate for the use of the diagnostic laboratory; his leadership led to the
establishment of similar laboratories elsewhere: in Massachusetts in 1894, in Philadelphia in 1895, et al. In 1901 he was appointed general medical officer of New York City and
at the same time he was appointed to the board of directors of the newly formed Rockefeller Institute for Medical Research. In 1914 he became the commissioner of health for
the state of New York. page 58

1892 Hermann Biggs founding of the New York City Department of Health microbiology laboratory
Mechanisms of Virus Neutralization by Antibody from: Occupancy and Mechanism in
Antibody-Mediated Neutralization of Animal Viruses. P. J. Klasse and Q. J. Sattentau.
Journal of General Virology, 2002.
a. Ab binding to a proportion of the receptor-interactive structures on the virion may block virus attachment to
the surface of target cells. In the schematic example shown, the Abs are bound to protein spikes on an enveloped
virus, which is thereby prevented from making contact with either of the two cell-surface receptors that it uses for
attachment and entry.
b. Ab inhibition of the interactions between the viral envelope protein and the cell-surface receptors is shown to occur
after the virion has attached by binding via the receptors. For example, one receptor may serve as an attachment
point as well as a trigger of conformational changes that allow interactions with a co-receptor, which in turn
mediate later events such as membrane fusion. Ab interference with any of these necessary links in a chain of events
that lead to entry would constitute a neutralization mechanism.
c. In order to infect, some viruses require internalization by endocytosis and the lowering of the pH in the endosome
as a trigger of conformational changes in viral proteins. Abs that have not blocked virus attachment but allowed
internalization are shown. Their block of requisite interactions between viral and cell-membrane proteins would
George Miller Sternberg (1838-1915) delay or prevent the penetration of the viral core into the target-cell cytoplasm. The virion may thus ultimately be
destroyed through lysosomal degradation. Such effects of Ab would constitute a mechanism of virus neutralization.
In addition, Ab-mediated derouting of viruses that preferentially enter directly via the cell surface to a less
Sternberg GM. Wissenschaftliche untersuchungen ber das spezifische permissive endosomal compartment may abrogate infectivity.
infcktionsagens der blattern und die erzeugung knstlicher immunitt d. The intercalation of Ab in the fusion interface between the cell membrane and the envelope of a virus may block
fusion at the cell surface, as illustrated, or in an endosome.
gegen diese krankheit. Centralbl f Bakteriol Abt. I, 1896;19:857-869.
e. It has been conjectured that even a very low occupancy of antibody on the virion can cause global or internal
Klasse PJ, Sattentau QJ. Occupancy and mechanism in antibody-mediated changes by transmitting a signal across the viral envelope or outer layer. These hypothetical changes would allow
the viral core to enter the cytoplasm but compromise further replicative steps.
neutralization of animal viruses. J Gen Virol. 2002;83:2091-2108.
g. The neutralization of naked viruses could potentially differ from that of enveloped viruses. Although naked viruses
Klasse PJ, Sattentau QJ. Mechanisms of virus neutralization by antibody. have been shown in several instances to be neutralized by a block of viral attachment to target cells, as in (a),
conformational effects on the whole virion by Ab binding have also been registered as pI (isoelectric pH) shifts.
Curr Top Microbiol Immunol. 2001;260:87-108.
The possibility that the Ab-virion complex enters the cytoplasm and that the Ab blocks further replicative events is
shown.
page 59

1892 George Sternberg discovery of virus neutralization (vaccinia virus)


Theobald Smith (1859-1934)
Cooper Curtice (1856-1939)
Fred L. Kilborne (1858-1936)
Daniel E. Salmon (1850-1914)

The first proof of arthropod


transmission of a disease:
The disease: Texas fever.
The infectious agent: Babesia bigemina.
The tick vector: Boophilus annulatus.
Boophilus annulatus tick
As acknowledged by Walter Reed in
1901, this discovery was a key to the
discovery of the first human virus,
yellow fever virus, and its transmission
by mosquitoes.

Theobald Smith was born in Albany,


New York in 1859. He graduated from
Cornell University in 1881 and
received an MD from Albany Medical
College in 1883. From 1884-1895
he was director of the pathology
laboratory of the Bureau of Animal
Industry, USDA, where he investigated
infectious diseases of animals. From
1895 to 1915 he served as Professor
of Comparative Pathology, Harvard
Medical School. In 1915 he became
director of the Department of Animal
Pathology at the Rockefeller Institute
for Medical Research in Princeton.
He was president of the American
Associated of Pathologists and
Bacteriologists, the American Society
of Tropical Medicine, the Congress of
American Physicians and Surgeons,
and the International Society Against
Tuberculosis. He was a Fellow of the
AAAS. He received twelve honorary
degrees and eleven honorary medals,
Babesia bigemina bovine blood film
among which was the Copley Medal
of the Royal Society.
Theobald Smith
page 60

1893 Theobald Smith, Cooper Curtice, others first proof of arthropod transmission of a disease, Texas Fever
Experimental fields from 1889. Scale: inch =
33 feet. a dwelling house; b station laboratory; c
house stable; d cow stable; e breeding pens; f tool
house; g shed in field. Field Vfield of healthy
Texas Longhorns heavily infested with ticks. Field
IVfield of healthy North Carolina cattle without
ticks. Experimental fields I-III: On experimental
field I, Texas longhorns grazed together with
North Carolina cattle. North Carolina cattle
Cooper Curtice became infested with ticks, and subsequently
Texas fever field investigation, collecting ticks from a dead cow, 1893 they developed Texas cattle fever and died. On
experimental field II, Texas longhorns without
This photo served as the frontispiece through many editions of the book, Hagen and Bruners Infectious Diseases ticks (ticks were removed manually by Smith and
of Domestic Animals, which was the authors textbook at the College of Veterinary Medicine, Cornell University. Kilbourne) grazed together with healthy North
Carolina cattle. North Carolina cattle remained
healthy. Next, healthy North Carolina cattle
Smith T, Kilbourne FL. Investigations into the nature, causation, and prevention of Texas or southern cattle fever. US were transferred to the heavily tick-infested
Dept of Agriculture, Bureau of Animal Industry, 8th and 9th Ann Repts, Washington, DC. 1893:177-304. experimental field I where the cattle became
[There are infamous stories of how the authorship of this work was manipulated by Daniel Salmon (1850-1914), the infested with ticks, developed Texas cattle fever,
director of the Bureau of Animal Industry, USDA, favoring Frederick Kilborne and downplaying the work of Cooper and died. As a last step, Smith transferred grass and
Curtice. Nevertheless, the experiments in cattle proving the transmission of the disease by ticks are considered a classic, ticks from experimental field I to experimental field
soundly designed, with all appropriate controls, and with clear conclusions.] III. Subsequently, healthy North Carolina cattle
became tick-infested and died of Texas cattle fever.
page 61

1893 Theobald Smith, Cooper Curtice, others first proof of arthropod transmission of a disease, Texas Fever
Ivanovskys experiments involved passing sap from diseased tobacco plants through
Chamberland porcelain ultrafilters, which were considered to be the ultimate test for bacteria, as
bacteria would be retained in the filters but soluble toxins would not. He was surprised when the
infectious moiety passed through the filters and suspected that the filters was defective. Further
testing of the filters convinced him that they were not defective: According to the opinions
prevalent today, it seems to me that the latter is to be explained most simply by the assumption
of a toxin secreted by the bacteria present, which is dissolved in the filtered sap. Besides this there
is another equally acceptable explanation possible, namely, that the bacteria of the tobacco
plant penetrated through the pores of the Chamberland filter-candles, even though before every
experiment I checked the filter used in the usual manner and convinced myself of the absence of
fine leaks and openings. He presented his findings to the Academy of Science in St. Petersburg in
1892. Ivanovsky published a more detailed paper in 1903, which included a detailed description
of the disease, including microscopic observations on the two types of inclusions found in cells of
infected tissues and extensive unsuccessful efforts to culture the agent. In the end he still came to
the conclusion that the causal agent was an uncultivable bacterium.
Iwanowski D. Concerning the mosaic disease of the tobacco plant. St Petersburg Academy Imp
Sci Bul. 1892;35:67-70. Translation published in English as Phytopathological Classics Number 7.
American Phytopathological Society Press, St. Paul, Minnesota; 1942.
Iwanowski D. ber die mosaikkrankheit der tabakspflanze. Zeit Pflanzenkr Pflanzenpathol
Pflanzenschutz. 1903;13:1-41.

Dmitry Ivanovsky (1864-1920)


(or Dmitrii Iwanowski)

page 62

1892 Dmitry Ivanovsky discovery of tobacco mosaic virus


Beijerinck Laboratory, Delft

Beijerinck repeated the ultrafiltration


experiments of his predecessors (although
he was apparently unaware of the work
of Ivanovsky) and concluded that what
passed through porcelain ultrafilters
remained infectious but did not contain
detectable microorganisms. He concluded
that it was a contagium vivum fluidum,
that is a contagious living fluid. He found
that from the sap of a diseased tobacco
plant ...an infinite number of healthy
plants may be inoculated and infected.
He concluded that, unlike a toxin, the
infectious agent reproduces in diseased
plants. He conducted diffusion experiments
in which he allowed the infectious agent to
penetrate agar he again concluded that
the infectious agent was soluble. He also
observed that the infectious agent in sap
extract remained viable after drying, but
Martinus Willem Beijerinck (1851-1931) was inactivated by heating to 90C.

Beijerinck MJ. Concerning a contagium vivum fluidum as cause of the spot disease of tobacco leaves. Verhandelingen der Koninkyke Akademie Wettenschapppen te
Amsterdam 1898;65:3-21. Translation published in English as Phytopathological Classics Number 7. American Phytopathological Society Press. St. Paul, Minnesota; 1942.
page 63

1898 Martinus Beijerinck discovery of tobacco mosaic virus


Friedrich A. J. Loeffler (1852-1915) Paul Frosch (1860-1928)

Loeffler F, Frosch P. Berichte der kommission zur erforschung der maul-und klauenseuche bei dem institut fur
infektionskrankheiten. Part I. 1898;23:371-391. In Milestones in Microbiology: 1556 to 1940, translated and edited by
Thomas D. Brock, ASM Press; 1998.

Friedrich-Loeffler-Institute Bundesforschungsinstitut fr Tiergesundheit


(German Federal Research Institute for Animal Health)
on the Isle of Riems is named in Loefflers honor
page 64

1898 Friedrich Loeffler and Paul Frosch discovery of foot-and-mouth disease virus (the first virus: see text)
Who discovered the first virus?
There are many, many articles arguing the priority of the discovery of the first virus this is a place
for strongly held opinions

Dmitry K. Lvov: Centenary of Virology, in Concepts in Virology: From Ivanovsky to the Present
[Mahy BWJ (Ed.)]. Brian Taylor & Francis, Inc.; 1993. It was Ivanovsky

A. van Kammen: Beijerincks Contribution to the Virus Concept. In 100 Years of Virology The
Birth and Growth of a Discipline. [Calisher CH, Horzinek MC (Eds.)]. Archives of Virology, Suppl.
15; 1999. It was Beijerinck

Marc H. V. van Regenmortel: The Nature and Classification of Viruses, in Topley & Wilsons
Microbiology & Microbial Infections [Mahy BWJ, ter Meulen V (Eds.)]. Hodder Arnold; 2005.
It was Loeffler and Frosch...

An Opinion on the Priority of the Work of Ivanovsky, Beijerinck and Loeffler and Frosch by
Marc van Regenmortel, 2006 abridged:
...Although all historical accounts of the beginnings of virology refer to the work of Ivanovsky,
Beijerinck, and Loeffler and Frosch (working with Koch), there is disagreement among authors
about who should be credited with the discovery that viruses were a new type of infectious agent.
This debate concerns the question of what is a scientific discovery.
Although Ivanovsky was clearly the first one to show that the agent causing tobacco mosaic disease
passed through a bacteria-retaining filter, all his publications show that he did not grasp the
significance of his observation. He believed that the filter he used might have had fine cracks and
that small spores of a microbe might have passed through the filter.
Beijerinck on the other hand, realized he was dealing with something different from a microbe but
he thought that the virus was an infectious liquid and not a particle.
Only Loeffler and Frosch correctly concluded that the virus causing foot-and-mouth disease was a
small particle that passed through a Chamberland filter, but was stopped by a finer-grained Kitasato
filter.
The debate about who should be considered the founder of virology may be settled only if it is
accepted that, in order to make a discovery, it is not sufficient to make a novel observation (i.e.,
Friedrich Loeffler Robert Koch the filterability of an infectious agent), but that it is also necessary to interpret the observation
(1852-1915) (1843-1910) correctly.
Good science does not consist only in making new observations but it requires also unbiased,
imaginative thinking which enables the scientist to arrive at the correct interpretation of his
experimental findings.
Loeffler and Froschs interpretation of their filtration experiments came the closest to the modern
concept of a virus and so they should be acknowledged as the founders of virology.
page 65

1898 Friedrich Loeffler and Paul Frosch discovery of foot-and-mouth disease virus (the first virus: see text)
European rabbit (Oryctolagus cuniculus)
lesion of myxomatosis above eye

Myxoma virus
Negative contrast
electron Microscopy

Myxoma virus was discovered quite serendipitously by Giuseppe Sanarelli of the University of Siena
when he was invited by the government of Uruguay to establish a Hygiene Institute in Montevideo.
In setting up laboratories, he procured domestic European rabbits (Oryctolagus cuniculus). Soon
after, his rabbit colony suffered an outbreak of a highly infectious, fatal disease. Sanarelli named the
disease myxomatosis and showed that it was caused by a ultrafilterable virus. In a series of classical
studies, Henrique de Beaurepaire Arago showed that Sylvilogus brasiliensis, which occurs widely in
South America, is the natural reservoir of myxoma virus and that the virus is transmitted by biting
mosquitoes. The systemic infection characteristic of myxomatosis does not occur in Sylvilogus
brasiliensis; instead the virus produces skin tumors which remain localized to the injection site and
Giuseppe Sanarelli (1864-1940) eventually heal. These lesions serve as the source of the virus that is transmitted mechanically by
mosquitoes.
Sanarelli G. Das myxomatogene virus. Beitrag zum stadium der krankheitserrege ausser halb des
sichtbaren. Zentralbl Bakteriol Abt 1. 1898;23:865-873.
Arago HB. Myxoma dos coelhos. Mem Inst Oswaldo Cruz 1927;20:225-247.
page 66

1898 Giuseppe Sanarelli discovery of myxoma virus (the first poxvirus)


Henry Rose Carter, a physician in the Marine Hospital
Corps (forerunner of the U.S. Public Health Service)
spent 20 years in Louisiana where he observed the strange
but uniform timing of yellow fever outbreaks on ships
arriving from Central and South American ports. He
noted that within a day of leaving such ports, yellow fever
cases occurred, but then it was two weeks before the
next cases appeared. In 1898, he was called to two small
towns, Orwood and Taylor Mississippi, to investigate a
yellow fever outbreak. There he put his observations to the
Henry Rose Carter (1852-1925) test, meticulously documenting exposures and cases and
recording the timing of every case. He concluded that the
infectious agent leaving the patient must undergo some
change in the environment before it is capable of infecting
another man. He thought this period was about 2 weeks,
Carter HR. A note on the spread of yellow fever in houses. Extrinsic and he called it the period of extrinsic incubation.
incubation. Medical Record 1901;59:933-937.
Henry Rose Carter collecting mosquito
larvae in the field, North Carolina
page 67

1898 Henry Rose Carter discovery of the yellow fever virus mosquito extrinsic incubation period
Conquerors of
Yellow Fever
by Dean Cornwell
from the series Pioneers
of American Medicine

An allegorical depiction of
one of the great moments
in medical history. With
Walter Reed (in white
uniform) and Carlos
Finlay (with white hair)
looking on, Jesse Lazear,
who died a month later,
is shown applying an
infected mosquito to the
arm of James Carroll. The
painting includes Aristides
Agramonte (behind
Lazear), Leonard Wood (in
brown helmet), Jefferson
Kean (in white helmet) and
several of the volunteers
who subsequently were
infected in the same way.
Carroll became infected as
a result of this experiment
in the summer of 1900
this was the first known
experimental transmission
of yellow fever from a
mosquito.

page 68

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
The history of the discovery of
yellow fever virus and its mosquito
transmission cycle is wonderfully
preserved in the Philip S.
Hench Walter Reed Yellow Fever
Collection at the University of
Virginia Health Sciences Library.
Walter Reed (1851-1902)

In 1900, the U.S. Yellow Fever Commission, led by Walter Reed, major U.S. Army Medical Corps, conducted experiments at stations just outside Havana, Cuba, that proved
that yellow fever is transmitted by mosquitoes, rather than by direct contact or fomites. He and the other members of the Commission, James Carroll, Aristides Agramonte,
and Jesse Lazear, proved that Aedes aegypti is the vector of yellow fever virus, and James Carroll conducted experiments that proved that the etiologic agent of yellow fever is
a virus the first human virus. The Commission operated as part of the American occupation force in Cuba after the Spanish-American War of 1898. During the war and the
following occupation yellow fever (and malaria, and dysentery) accounted for most deaths and disabilities. The Commissions discoveries focused efforts on the eradication
of the mosquito within six months yellow fever had been eliminated from Havana and within 10 years from Cuba. Although Reed received much of the credit for the
discoveries, he credited Carlos Finlay with the discovery of the yellow fever vector, the key to how the disease might be controlled. He also credited the insight provided by
Henry Rose Carters evidence of a mosquito extrinsic incubation period, by Theobald Smith and his colleagues proof that the agent of Texas fever was transmitted by an
arthropod, and Friedrich Loeffler and Paul Froschs proof that the agent of foot-and-mouth disease was a virus.
page 69

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
American Association for the Advancement of Science Resolution:
Death of Dr. James Carroll from Yellow Fever Experimentation

Whereas, The late Major James Carroll, M.D., U.S. Army, was the first
to submit voluntarily to the bite of an infected stegomyia, and from
the bite of this mosquito, suffered a severe attack of yellow fever, the
effects of which led to his ultimate death, and
Whereas, This was the first experimentally produced case of yellow
fever leading to the present knowledge of this disease, which has
practically enabled its complete control, therefore, be it
Resolved, That the American Association for the Advancement of
Science now in session in Chicago, Illinois recommends to the
Senate and House of Representatives of the United States of America
the passage of a bill securing to Mrs. Jennie Carroll, widow of the
late Major James Carroll of the Yellow Fever Commission, of the
United States Army, a special pension for the support of herself and
her seven children.
[Adopted by the AAAS Council, December 30, 1907.]

James Carroll (1854-1907)


From: The Scientific Work and Discoveries of the late
Major Walter Reed, Surgeon in the Army of the United States.
Prepared by Major Jefferson Randolph Kean.
U.S. Senate Document No. 118, 57th Congress, 2nd Session.
Washington: Government Printing Office; 1903.
page 70

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
Discovery of the First Human Virus
The original work of the Yellow Fever Commission was concluded in February 1901, but James Carroll was
allowed to return to Cuba in August of that year. Camp Lazear had been closed, so he worked at Las Animas
Hospital. To determine if the agent of yellow fever was ultrafilterable, he obtained some infected mosquitoes from
Juan Guitras and produced new cases. In August through October 1901, he produced six cases of yellow fever. A
Spaniard, Pablo Ruiz Castilo, was bitten by four mosquitoes on September 16 and became severely ill three days
later. Another Spaniard, Jacinto Mendez Alvarez, was bitten on October 9 by eight mosquitoes and developed a
mild illness.
On October 15, Paul Hamann, an American soldier, was injected with serum from Alvarez that had been passed
through a Berkefeld ultrafilter. Four days later, he developed yellow fever. Albert Wall Covington, another soldier,
was injected with the same ultrafiltered serum and became ill four days later. John R. Bullard, a civilian, was
given a similar injection. Four days later, he had a modest elevation in temperature and pulse, accompanied by a
headache and pain between his shoulders (not considered a certain case of yellow fever).
One week later, Bullard was injected again with ultrafiltered serum, this time from blood drawn from Paul
Hamann; he developed clinical signs of yellow fever just twenty-four hours later. Carroll was at a loss to explain
Bullards case: if he became infected from the first injection, the incubation period was nine days, three days
longer than any other case. If it was from the second, the incubation period was one day, less than half as long
as any other case. Carroll was inclined to believe that Bullards case was from the second injection, but he was
unsure.
It is certain that Carroll proved that the agent of yellow fever is an ultrafilterable agent and not a toxin in the
blood of yellow fever patients. In fact, James Carroll discovered the first human virus and demonstrated the
first experimental transmission of a viral disease from one human to another.
Shortly after this, Carroll received orders forbidding him from doing further human research this was partly
due to the public outcry over the death of the American nurse Clara Maass, and partly due to the fact that the
mosquito-control campaign had by this time practically eliminated the disease from Havana.

From: Pierce JR, Writer J. Yellow Jack,


How Yellow Fever Ravaged America
and Walter Reed Discovered its Deadly
Secrets. Hoboken, NJ: Wiley; 2005.

James Carroll (1854-1907)


Las Animas Hospital
Yellow fever ward
Havana, Cuba, 1901
page 71

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
Jesse William Lazear (1866-1900) Aristides Agramonte y Simoni
The Washington Observer (1868-1931)
September 29, 1900

Aristides Agramonte y Simoni was a physician, pathologist and bacteriologist with expertise in tropical medicine. In 1898 he was appointed Acting Assistant Surgeon, Medical
Corps, U.S. Army and was sent to Cuba. Although he had no recollection of having had yellow fever as a child, he was thought to be immune because he had been born in
Cuba. While a member of the U.S. Army Yellow Fever Commission he autopsied numerous victims of yellow fever and helped eliminate the widely held notion that the disease
was caused by a common bacterium, Sanarellis Bacillus icteroides.
Jesse William Lazear was a physician and bacteriologist trained at Johns Hopkins, Columbia and the Institut Pasteur; in 1900 he reported for duty as an Assistant Surgeon,
Medical Corps, U.S. Army at Columbia Barracks (Quemados), Cuba, where he joined the Yellow Fever Commission. Three months later, without telling his colleagues, he
allowed himself to be bitten by yellow fever-infected mosquitoes and died. Although Lazear never admitted to experimenting on himself, when Walter Reed reviewed his
notebook he evidently found entries strongly suggesting Lazears infection was not accidental, as officially reported. Unfortunately, the notebook, so crucial to the preparation
of the Commissions initial paper, The Etiology of Yellow Fever A Preliminary Note, vanished from Reeds Washington office after his death in 1902.
page 72

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
Camp Lazear, Quemados, Cuba, 1900
Left: Infected Mosquito Building Center: Infected Clothing Building

The Yellow Fever Human Subjects Experimentation, Cuba, 1900-1901


The experimental work of the U.S. Army Yellow Fever Commission took place at a site at Quemados near
Havana, later named Camp Lazear. Here, Walter Reed designed two small wood-frame buildings, each 14
by 20 feet one conspicuously clean and well ventilated, the other filthy and fetid, all to test whether yellow
fever transmission involved contact and fomites or mosquitoes.
Three trials of twenty days each, involving seven men, were conducted in Building One, called the Infected
Clothing Building this was to determine whether contaminated bedding and clothing (bed linens, blankets,
nightshirts etc., recently worn and soiled by patients in the wards of Columbia Barracks Hospital and Las
Animas Hospital in Havana) could be involved in the transmission of yellow fever no cases occurred.
Building Two, called the Infected Mosquito Building, contained a principal room, divided into two sections
by a floor-to-ceiling wire mesh screen, so configured to keep infected mosquitoes inside one section only. A
separate room attached to the building held the laboratory where mosquitoes were raised and stored. Four
trials were conducted, each requiring two groups of volunteers, one to be exposed and another to serve as
controls. Loaded mosquitoes, that is mosquitoes obtained from rooms inhabited by known yellow fever
patients, were released into one screened section where volunteers remained for about twenty minutes,
enough time to receive several mosquito bites. These volunteers then exited to a quarantine tent outside.
The controls spent the night in the section of the room free of mosquitoes they returned to sleep in the
room for as many as eighteen more nights. As Reed stated, the absence of yellow fever in the controls
showed, that the essential factor in the infection of a building with yellow fever is the presence therein of
[infected] mosquitoes, and nothing more. Taken together, all the experiments led to 22 cases of yellow fever:
14 by mosquito bite, 6 by blood injection, and 2 by injection of ultrafiltered serum. There were no deaths in
the main series of experiments, but there were three in experiments carried out later by William Gorgas and
Juan Guitras one was the American nurse, Clara Maass.
page 73

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
Hospital Corps Detachment, Camp Columbia, Havana, 1900 Clara Louise Maass (1876-1901)
Most of the volunteers for the yellow fever experiments came from The only woman volunteer.
this unit. Human subjects were needed to test the mosquito theory After experimental mosquito exposure she
because, at the time, no one knew of any animals susceptible to the developed severe yellow fever and died.
virus. The four members of the Army Yellow Fever Commission Her death in 1901 led to a public outcry
agreed to experiment on themselves before requesting volunteers. that ended human experimentation.
Agramonte was exempted since it was thought that he was immune,
having been infected in childhood. When Reed departed for Clara Maass was the first nurse honored
Washington to complete a typhoid report, only Carroll and Lazear with an American stamp (issued in 1976).
were left to share the risk with their volunteers. Several Spanish
immigrants also participated in the experiments as volunteers.
page 74

1900 Walter Reed, colleagues discovery of yellow fever virus (the first human virus) and its transmission cycle
John McFadyean, Principal, Royal
Veterinary College, London (1892-
1927). The teaching staff, 1895.
John McFadyean, center front row

John McFadyean, considered the father of veterinary research, was trained in Edinburgh
as a veterinarian and as a physician. Early on he became interested in bacteriology and
pathology. In 1888 he originated the Journal of Comparative Pathology and Therapeutics,
which he edited for many years. In 1892, he was appointed principal and professor of
bacteriology and pathology at the Royal Veterinary College, London, and for the next 35
years he had a profound effect on the development of veterinary science. In 1900, working
John McFadyean (or MFadyean) (1853-1941) in London with blood from infected horses in Africa brought in sealed tubes by a returning
English veterinarian, McFadyean found that diluted blood passed through Berkefeld and
MacFadyean J. African horse-sickness. J Comp Path Ther. 1900;13:1-30. Chamberland F and B ultrafilters remained infectious he confirmed his results in many
experiments and proved the efficacy of the filters by mixing the blood to be tested with
MacFadyean J. A further contribution to the pathology of African horse- known bacteria which the filters retained as usual. At the time he thought that the agent was
sickness. J Comp Path Ther. 1901;14:103-118. nothing more than a very small bacterium, but by 1908, in view of the inability to grow the
MacFadyean J. The ultravisible viruses. J Comp Path Ther. 1908;21:58-68, 168- agent on artificial media, he now described the agent as ultravisible an obligatory parasite.
175, 232-242. In 1901, McFadyeans ultrafiltration results were confirmed by Arnold Theiler in South Africa
page 75 and Edmund Nocard in France.

1900 John McFadyean discovery of African horse sickness viruses (the first orbiviruses)
Hugo Marie de Vries (1848-1935) Oenothera lamarckiana (Evening Primrose)
drawn by Hugo de Vries
Hugo de Vries was a Dutch botanist who conceived the concept of genes, rediscovering the laws of heredity in the 1890s, while unaware of Gregor Mendels work thirty years
earlier. de Vries developed the laws of dominance and recessiveness, segregation, and independent assortment of inherited characters to explain the 3:1 ratio of phenotypes in
the second generation of his cross-breeding of evening primrose plants. In the late 1890s, de Vries became aware of Mendels obscure paper of 1866 and altered some of his
terminology to match. de Vries Mutation Theory (1900-1903) was one of the chief contenders to explain how evolution works, leading, for example, Thomas Hunt Morgan to
study mutations in the fruit fly this, in turn, stimulated Theodosius Dobzhansky and others to restate their definition of Darwinism in the modern evolutionary synthesis in
the 1930s and 1940s.
page 76

1900 Hugo de Vries founding of genetics, rediscovery of Mendels work


Rockefeller Institute, 1904 John D. Rockefeller Sr. (1839-1937)
and John D. Rockefeller Jr. (1874-1960)
The origins of the Rockefeller Institute for Medical Research lie, in part, in personal tragedy. After John D. Rockefeller Srs grandson died from scarlet fever in January 1901,
he formalized plans to establish the research center he had been discussing for three years with his adviser Frederick T. Gates and his son John D. Rockefeller Jr. At the time
of the institutes founding, infectious diseases such as tuberculosis, diphtheria, typhoid fever and yellow fever were considered the greatest threats to human health. New
research centers in Europe, such as the Institut Pasteur, were successfully applying laboratory science to increase understanding of these and other diseases. Following their
lead, the Rockefeller Institute became the first biomedical research center in the United States. From the beginning, Rockefeller researchers made important contributions to
understanding, preventing and treating disease, and within a few years their impact was felt globally, especially in the emerging discipline of virology.
page 77

1901 John D. Rockefeller Sr. founding of the Rockefeller Institute for Medical Research
Rockefeller Institute for Medical Research
First Board of Directors, Left to right:
Theobald Smith
Hermann Biggs
Simon Flexner (director)
William Welch (chair)
Theophil Prudden
Luther Holt
Christian Herter
Photo ~1909

Simon Flexner (1863-1946)


First director of the Rockefeller Institute
[from Rockefeller Archives, used with permission]

page 78

1901 John D. Rockefeller Sr. founding of the Rockefeller Institute for Medical Research
In 1901, three papers were published, all reporting
successful filtration experiments with the agent of fowl
plague. Two of the papers were by Italians, published
as reports to academies of medicine by Centanni and
Savonuzzi, and Maggiora and Valenti. The third paper
was by the Austrians Lode and Gruber. This almost
simultaneous appearance of three very detailed papers
reflects the rapidly growing interest in general in the
nature of the filterable viruses. Maggiora and Valenti
concluded that the agent behaved as a true virus, which
multiplied in the infected host, and not as a mere toxic
substance. Lode and Gruber considered three distinct
possibilities for the nature of the virus their notion
started out rather as a combination of the concepts of
Loeffler and Frosch and Beijerinck. Soon Lode concluded
from further ultrafiltration experiments using graded
Berkefeld filters that he was dealing with ultrasmall
bacteria, in keeping with the original concept of Loeffler
and Frosch.
Centannis comments were brief and objective: In view
of the incomplete methods currently at our disposal,
we must of course defer the question of whether the
reproduction of this virus involves living organisms or
complex chemical molecules, or even elements belonging
in some transitional area between the two.
During the next ten years, every attempt to isolate
and study unknown pathogens included attempts at
ultrafiltration. Review articles began to appear, recording
the rapidly growing number of known filterable agents.
When Emile Roux published the first review article in
1903 he included a total of nine known viruses. When
his colleague Paul Remlinger reviewed the situation three
years later the number had doubled to eighteen.
More than fifty years later the virus of fowl plague was
Eugenio Centanni (1863-1942) shown to be an avian influenza A virus. In 1983 avian influenza virus
From: Wilkinson L. The development of the virus concept (H5N2) (fowl plague virus)
Centanni E, Savonuzzi E. La peste aviaria I & II. Communicazione fatta as reflected in corpora of studies on individual pathogens. appeared in Pennsylvania, killing
allAccademia delle Scienze mediche e naturali di ferrara, 1901. 17 million chickens, and costing
1. Beginnings at the turn of the century. Medical History
Maggiora A, Valenti GL. Su una epizoozia di tifo essudativo dei gallinacei. 1974;18:211-221. more than $65M. The same kind
Accad Med di Modena, 20 June 1901. of mutation to virulence occurred
Lode A, Gruber F. Bakteriologische studien fiber die aetiologie einer in the commercial broiler industry
epidemische erkrankung der hhner in tirol. Zentbl. Bakt Parasitkde Abt I. of Mexico in the 1990s and in Asia
1901;30:593-604.
from 1995 onward.
page 79

1901 Eugenio Centanni, others discovery of fowl plague virus (avian influenza virus, the first orthomyxovirus)
Aladr Aujeszky (1869-1933)

Aujeszky A. ber eine neue infektionskrankheit bei


haustieren. Zbl Bakt 1 Orig. 1902;32:353-357.

Pseudorabies (or Aujeszkys disease, and in cattle mad itch) is a disease of swine that is endemic in most
parts of the world. It is caused by pseudorabies virus (PRV), which is also called porcine herpesvirus 1
or suid herpesvirus 1. It is considered the most economically important viral disease of swine in areas
where classical swine fever has been eradicated. Aladr Aujeszky, a Hungarian physician and veterinarian,
recovered the virus from dogs, cats, and cattle as well as swine, and discovered that it was transmissible to
rabbits and guinea pigs. He proved it was a virus by ultrafiltration through graded Berkefeld ultrafilters.
page 80

1902 Aladr Aujeszky discovery of pseudorabies virus (the first herpesvirus)


A graphic explanation of the
Andy Warhol-like artistic
image of poxviruses: within the
typical complex brick-shaped
virion of all orthopoxviruses,
lies a dumbell-shaped core
containing the viral DNA.
Some idea of the complexity of
the poxvirus virion is indicated
by the fact that it contains more
than 200 proteins.
Amde Borrel (1867-1936)
[from Institut Pasteur, used with permission] Sheeppox virus, along with
goatpox and lumpy skin disease
Borrel A. Epitheliosee infecteuses et epitheliomas (infectious epithelioses and viruses, is responsible for
epitheliomas). Annales de LInstitut Pasteur 1903;17:81-91. economically significant diseases
of domestic ruminants in many
Borrel A. tude exprimentale de las clavele. Annales de LInstitut Pasteur areas of Africa and Asia.
1903;18:123-137.
page 81

1902 Amde Borrel discovery of sheeppox virus


Maurice Nicolle, brother of Nobel laureate Charles Nicolle, a staff member of the Institut Pasteur, Paris, went to
Constantinople (Istanbul) in 1893 at the request of the Sultan of the Ottoman Empire, Abdulhamid II. There, he
founded the Imperial Institute of Bacteriology (for human and animal diseases) and became its first director. This was
the third of the 31 Pasteurean institutes founded around the world.
Mustafa Adil Bey was a Turkish veterinarian, a graduate of the Ecole Nationale Vtrinaire dAlfort, who had been
trained by Edmond Isidore tienne Nocard. He was the founder of the Turkish Veterinary Bacteriology Institute.
Nicolle and Adil Bey worked on rinderpest from 1899 onward: they started mass production of bovine antiserum, they
showed varying breed resistance, they defined the incubation period of the infection, etc.
In 1902 they published their ultrafiltration studies, showing that the disease rinderpest was caused by a virus. Nicolle
and Adil Bey also demonstrated the ultrafilterability of vaccinia virus, five year before Negri and others, but their
publication was quite obscure.
Nicolle M, Adil Bey M. Etudes sur la peste bovine, (troisime mmoire). Experiences sur la filtration du virus. Ann Inst
Pasteur 1902;16:56-64.
Nicolle M, Adil Bey M. Sur la nature du virus vaccinal. Comptes Rendus de lAcademie des Sciences. Serie III, Sciences
de la Vie 1906;143:1196.

Rinderpest, South Africa, ~1900


Maurice Nicolle (1862-1932)

Mustafa Adil Bey


(1871-1904)
page 82

1902 Maurice Nicolle and Mustafa Adil Bey discovery of rinderpest virus (the first morbillivirus)
Arnold Theiler (1867-1936) Onderstepoort Veterinary Research Institute
founded in 1908, Arnold Theilers statue in front
Bluetongue was first described in South Africa, where it had likely been endemic in wild
ruminants from antiquity. It was recognized when imported Merino sheep were found
to be highly susceptible to infection with fatal consequences. The first detailed studies
were done by James S. H. Spreull (1874-1948) in 1902-1905, when he was a beginning
staff member of the Veterinary Services of the Union of South Africa (founded in 1896;
Arnold Theiler, first Director), Grahamstown, South Africa. The separate Government
Veterinary Laboratory at Onderstepoort (now the ARC-Onderstepoort Veterinary
Institute), was commissioned in 1908; Spreull became an early staff member. and
Theiler its founding director. Spreull: Cause of the Disease [bluetongue]: Repeated
microscopic examinations have revealed no organism in this disease but as the blood
conveys the infection readily upon inoculation into a clean animal we can conclude that
the cause is ultra visible and this is confirmed by the fact that when virulent blood or its
serum only are passed through the Berkefeld filter the filtrate is found to be capable of
reproducing the disease (Robertson and Theiler) [William Robertson (1872-1918)]. Mr.
Dixon [Rowland W. Dixon (-1948)] and I performed this experiment in 1901 with
pipetted [ultrafiltered?] serum the results being exactly similar Bluetongue virus
Model: outer capsid layer (blue/red),
Spreull J. Malarial catarrhal fever (bluetongue) of sheep in South Africa. J Comp Pathol Ther. 1905;18:321-337.
inner capsid (grey), one of the ten
Theiler A. Bluetongue in sheep. Annual Report of the Director of Agriculture, Transvaal, 1904-1905, 110-121. segments of its double-stranded RNA
page 83 genome (aqua). Tom Goddard, UCSF

1902 James Spreull, Arnold Theiler discovery of bluetongue viruses


Classical swine fever virus
negative contrast electron microscopy
[Micrograph from Frank Weiland]

Emil Alexander de Schweinitz Marion Dorset


(1864-1904) (1872-1935)
De Schweinitz EA, Dorset M. New facts concerning the etiology of hog cholera [now called classical swine fever].
Bureau of Animal Industry, USDA, 20th Annual Report, 1903.
The first practical preventive measure used widely to prevent a livestock viral disease was anti-hog-cholera serum
injected together with virulent virus. This invention of Marion Dorset (with D. McBryde and W.B. Niles) was
successfully tested in 1907 and immediately commercialized.

Spleen from infected pig, showing diffuse, acute


necrosis of a lymphoid follicle surrounded by
red pulp hemorrhage, the typical lesion of
classical swine fever (hog cholera)
page 84

1903 Emil De Schweinitz, Marion Dorset discovery of classical swine fever virus (the first pestivirus)
Adelchi Negri (1876-1912) Negri bodies, brainstem and Purkinje cell, human, Giemsa stain
In 1890, just after Pasteurs studies on rabies, the famous Camillo Golgi, professor of pathology at the University of Pavia, investigated the neuropathology of rabies, without
obtaining significant findings. Adelchi Negri, an assistant pathologist in Golgis laboratory, continued these studies. Using Mann stain, he observed, first in rabbits and then
in rabid dogs, characteristic inclusions in nerve cells of the spinal ganglia and brain. This discovery was presented in 1903 at a meeting of the Societ Medico-Chirurgica of
Pavia. Others immediately found the same inclusions in human rabies cases. Negri was convinced that the inclusions were specific features of rabies, and he believed he had
identified the etiologic agent responsible for the disease as a parasite. The report by Negri in 1903 immediately stimulated a scientific debate. Within a few months Alfonso
di Vestea in Naples and Paul Remlinger and Riffat Bey in Constantinople showed that the etiologic agent of rabies is an ultrafilterable virus. However, Negri tried until 1909
to demonstrate that the intraneuronal inclusions named after him corresponded to different steps of the developmental cycle of a protozoan. Despite his incorrect etiological
hypothesis, Negri had made a discovery of great practical importance his finding provided a breakthrough in the rapid diagnosis of rabies and the detection of Negri bodies
was immediately adopted by all the anti-rabies institutes of the world.
page 85

1903 Adelchi Negri discovery of the rabies inclusion bodythe Negri body
Rabies inclusions (Negri bodies);
remarkable camera lucida
drawings by Adelchi Negri
included in his original paper
(1903).

(a) Cerebral cortex of rabbit subdurally infected


with street virus: death occurred after 19 days.

(b) Ammons Horn, and

(c) Cerebellum, of dogs 19 days after experimental


subdural inoculation with street virus.

(d) Spinal ganglion of rabbit infected in the eye with


street virus: death occurred after 17 days.

Fixation in Zenkers solution. Manns staining.

Reproduced from Negris paper:


Negri A. Contributo allo studio delleziologia della
rabia. Bollettino della Societa Medico-Chirurgica,
Pavia 1903;2:88-115.

page 86

1903 Adelchi Negri discovery of the rabies inclusion bodythe Negri body
Paul Ambroise Remlinger (1871-1964)
[from Institut Pasteur, used with permission]

Remlinger PA, Riffat Bey.


Le virus rabique traverse
la bougie Berkefeld. CR Rabies virus, negative contrast and thin section electron microscopy,
Soc Biol. 1903;55:730-731. purified virus and brain of hamster infected with street virus (colorized)
The Sultan of the Ottoman State sent a mission to Paris in 1886 to learn how to prepare Pasteurs
Remlinger PA, Le passage rabies vaccine. Upon its return to Constantinople (now Istanbul), a Rabies Vaccine Laboratory (Rabies
du virus rabique traverse Hospital) was established. In 1900, Paul Remlinger, a military physician, who had been at the Institut
les filtres. Ann Inst Pasteur in Tunis, and had previously trained at the Institut Pasteur, Paris, was appointed director of
Pasteur 1903;17:834-849. the Laboratory, where in succeeding years he carried out many remarkable studies on rabies. In 1902,
Remlinger, Rifat Bey, a Turkish veterinarian who had studied at the Institut Pasteur, Paris, and Hamdi
Efendi, a laboratory assistant, performed a key experiment: they mixed a fixed rabies virus brain
homogenate with fowl cholera bacteria (Pasteurella multocida), put the mixture through a Berkefeld
V ultrafilter, and inoculated the filtrate intracerebrally into rabbits. While the absence of Pasteurella in
the inoculated animals confirmed the success of the ultrafiltration, their later death from rabies made
clear that the etiologic agent of rabies was ultrafilterable. They repeated the experiment using both
Rifat Bey fixed and street viruses, and graded Berkefeld and Chamberland filters. Remlinger later served for
page 87 many years as a senior staff member of the Institut Pasteur, Paris.

1903 Paul Remlinger, Riffat Bey, Alfonso di Vestea discovery of rabies virus (the first rhabdovirus)
Alfonso di Vestea (1854-1938)

Rabies virions at margin of a Negri body, hamster brain,


thin section electron microscopy

Remlinger P. Le passage du virus rabique a travers les filtres. Ann Inst Pasteur
1903;17:834-849.

Remlinger PA, Riffat Bey. Le virus rabique traverse la bougie Berkefeld. C R de


la Socit de Biologie. 1903;55:730-731.

Di Vestea A. Sul trovato della filtrabilit del virus della rabbia Annali di igiene
Time series: development of Negri bodies sperimentale. 2005;15:147.
neuroblastoma cells, at 4, 8 and 24 hours post-infection
page 88

1903 Paul Remlinger, Riffat-Bay, Alfonso di Vestea discovery of rabies virus (the first rhabdovirus)
Tiselius original liquid electrophoresis technology

Preparative and analytical methods developed by separation scientists have played an important role
in the history of virology. This started in 1937 with the earliest liquid electrophoresis technique of
Tiselius; however, using liquid electrophoresis to separate molecules based on their charge without
stabilizing media has very poor resolution since molecules (or even virions) that are not stabilized
in a supporting medium, such as a gel, diffuse quickly after initially being separated. Early on, the
Rockefeller Foundations financial assistance brought Tiselius electrophoretic apparatus to selective
American research institutions this was seminal in the birth of molecular biology (and molecular
virology). Immediately, Tiselius understood the early shortcomings of liquid electrophoresis and
had ideas for their resolution: It seems practical to distinguish between boundary electrophoresis
Arne Wilhelm Kaurin Tiselius (1902-1971) as performed in the common moving boundary apparatus, and zone electrophoresis where the
migration of more or less completely separated zones is studied, usually by application of some
immobilizing medium to prevent convection. All the electrophoretic technologies that we know
today followed first paper electrophoresis, then starch block electrophoresis, but eventually many
gel electrophoresis methods.
page 89

1903 Arne Tiselius development of electrophoresis


In their 1904 publication, Henri Carr and
Henri Valle presented remarkable findings
from their experimental studies of equine
infectious anemia done at Alfort. They
showed that the disease was transmissible by
inoculation of blood, that the etiological agent
could be serially passaged in horses, and that
the singular disease in nature appeared in
three quite different forms, acute, subacute and
chronic. This was in an era when many experts
did not think that a single etiologic agent
could cause different clinical syndromes. Most
remarkable was their proof of the nature of the
etiologic agent (from a translation, abridged):
We have shown that the anemia virus in the
horse belongs to the category of microbes,
the so-called invisible microbes, those that
can pass through filters of diatomaceous earth
or porcelain, which hold back the microbes
large enough to be seen under the microscope.
New experiments have shown that regardless
of the time during the infection when blood
is taken, it contains the virus which does
not lose its infectivity even after the fifth
dilution [? five-fold dilution or 10-5 dilution]
Henri Valle (1870-1957) Henri Carr (1868-1938) before ultrafiltration, and the results are the
same whether using the diatomaceous earth
Berkefeld candles or porcelain Chamberland F
and B candles. These candles, however, retain
a fairly large amount of virus so the incubation
period of the disease produced by inoculation
of filtrates is always longer.... Finally, we have
shown, importantly, that horses that seem
absolutely cured of the chronic form of the
disease retain their infectivity. The inoculation
of blood from one of these horses into a horse
in excellent condition caused disease in its
acute form with death in 27 days. There is
reason to fear that the presence of silent virus
carriers in infected regions is a very big
obstacle to the extinction of the disease by any
prophylactic measures.
Equine infectious anemia virus
thin section electron microscopy Ecole Nationale Vtrinaire DAlfort, ~1900 Carr H, Valle H. Sur lanmie infectieuse du
cheval. CR Acad Sci. 1904;139:331-334.
page 90

1904 Henri Valle, Henri Carr discovery of equine infectious anemia virus (the first retrovirus, the first lentivirus)
Yellow Fever Control / Elimination in Panama
1880: French company headed by Ferdinand de Lesseps breaks ground on
a sea-level canal in Panama.
1881: First construction crews arrive; first workers die of yellow fever.
1884: Work force of 19,000; it is estimated that 12,000 workers die during
the construction of the Panama Railway, and over 22,000 during the
French effort to build a canal.
1888: French abandon sea level canal and begin work on a lock canal.
1889: French canal company dissolved and work on canal stops; new
company formed and makes first excavations in Culebra Cut.
1898: New companys capital gone, offer to sell rights to the United States.
Gorgas in Panama, 1905
1898: At the end of the Spanish-American War, William Gorgas is
appointed Chief Sanitary Officer in Havana, and after the discoveries by
the Yellow Fever Commission he leads the campaign to eradicate yellow
fever from Havana, succeeding by 1902.
1902: President Theodore Roosevelt tries to buy rights to dig canal from
Colombia, but is refused.
1903: Roosevelt backs Panamanian uprising against Colombia by
stationing warships offshore. Panama declares independence and United
States signs a treaty with Panama allowing it to build canal. The treaty also
creates Canal Zone, a sovereign part of United States.
Teddy Roosevelt at Pedro
Miguel Lock, 1906 1904: United States buys French companys rights and properties, begins
construction; maximum workforce, 45,000 by 1911.
1904: Gorgas team arrives in Panama within a month all contract
malaria Gorgas urgent requests for action to eliminate mosquitoes are
ignored by John Findley Wallace, the Chief Engineer in charge of building
William Crawford Gorgas (1854-1920) the canal.
1905 (March): Yellow fever outbreak causes panic in Panama, most
American canal workers flee and work is virtually halted.
1905 (July): John Stevens arrives as new Chief Engineer and Gorgas public
health efforts are given top priority. As in Cuba, these efforts include the
Fumigation of buildings, draining of ponds and swamps, fumigation of buildings, mosquito netting of
Panama, 1906 beds, and public water systems to replace open standing water vessels.
1905 (December): Yellow fever eliminated from Panama.
1914: Official opening of Panama Canal S.S. Ancon makes first official
passage.

Draining swamp
Panama, 1911
page 91

1904 William Gorgas control of yellow fever, allowing construction of the Panama Canal
1904 1905 page 92

1904 William Gorgas control of yellow fever, allowing construction of the Panama Canal
Spraying
oil in
ditches,
Panama Canal
1906

Mosquito control gangs, Panama Canal, 1906

page 93
SS Ancon, first passage through the Panama Canal, 1914
1904 William Gorgas control of yellow fever, allowing construction of the Panama Canal
Le Docteur Henri Carr

Carr H. Sur la maladie des jeunes chiens. C R Acad Sci


(Paris) 1905;140:689-690 and 1489-1491.
Dunkin GW, Laidlaw PP. Studies in dog-distemper. I.
Dog-distemper in the ferret. II. Experimental
distemper in the dog. III. The nature of the virus. J Comp
Pathol. 1926;39:201-212, 213-221, 222-230.
[George W. Dunkin, MRCVS, was a staff member of the
National Institute of Medical Research, Mill Hill, and
later director of the Agricultural Research Council of
Henri Carr (1868-1938) Patrick Playfair Laidlaw the UK and President of the Royal Society of Medicine,
(1881-1940) Section on Comparative Medicine. No photo was found.]
Phylogenetic studies have indicated that measles virus and rinderpest virus diverged during
the 11-12th centuries. The divergence of canine distemper virus seems to have occurred more
recently, with the first case described in 1905 by Henri Carr at Alfort, who then showed that
the infectious agent was ultrafilterable. It is now known that the virus affects all populations
of the domestic dog and importantly many species of wildlife. The virus contributed to the
near-extinction of the black-footed ferret and to the extinction of the Tasmanian tiger and
one species of African wild dog. The initial major canine distemper virus research program
was developed at the National Institute for Medical Research-Mill Hill (NIMR)(and later at
the Baker Institute for Animal Health at Cornell University). Rick Carver and John Skehel, in
their Mill Hill Essay, described that canine distemper research was central to the mission of
the NIMR from its founding in 1913. Contributions from dog owners through an appeal from
The Field magazine, allowed pathologist Patrick Laidlaw and veterinarian George Dunkin to
confirm Carrs work and by 1929 to develop the first protective vaccine. One key to the Mill
Hill success was the realization that dogs used in experiments had to be bred and maintained in
strict isolation to avoid introduced infection and that unique facilities were required to achieve
this Mill Hill was the site of the first high containment research animal facility. The NIMR
interest in canine distemper was also based on its similarity to human diseases such as measles Canine distemper virus
and influenza, just at the time of great measles outbreaks and the 1918 influenza pandemic. dog, trachea, intracytoplasmic inclusions
page 94

1905 Henri Carr, Patrick Laidlaw, George Dunkin discovery of canine distemper virus
Proof that Variola and Vaccinia are
Viruses:
Finding the initial proof of the viral nature of variola
and vaccinia, by passage through Chamberland
or Berkefeld filters, was difficult there are
contradictory, incomplete citations, many without
formal references, in the older literature. By ~1912,
many statements were being made that make it
seem as if the proof was common knowledge and
did not need referencing. Nothing was found in
newer literature to resolve this. In any case, quite a
few different investigators are mentioned by various
authors in the older literature:

Negri A. Esperienze sulla filtrazione del virus


vaccinico. Nora prima. Gazzetta Medica Italiana,
Anno LVI 1905;13-46.
Adelchi Negri Paul Remlinger Negri A. ber filtration des vaccinevirus. Zeitschrift
(1876-1912) (1871-1964) fr Hygiene und Infektionskrankheiten 1906;54:327-
346.
Remlinger P, Nouri-Osman. Le virus vaccinal
traverse la bougiede Berkefeld V. C R Societe de
Biologie, Paris, 1905, and, Sur le passage du
virus vaccinal travers la bougiede Berkefeld V. C R
Societe de Biologie, Paris, 1905.
von Prowazek S. Untersuchungen ber das wesen
des vaccineerregers. Deutsche Med Wochenschr S.
1905;19:752.
Siegel J. Untersuchungen ber die tiologie der
pocken und der maul-und-klauenseuche. Abhandl d
knigl preu. Akademie d Wissenschaften 1, 1905.
Rouget J. Contribution ltude du virus vaccinal. C
R Acad Sci Hebd Seances Acad Sci. Nr. 21, 1905.
Nicolle M, Adil Bey M. Sur la nature du virus
vaccinal. C R lAcad des Sci. 1906;143: 1196.
And others, cited but without specific references:
Maurice Nicolle Stanislaus von Prowazek Carini, Volpino, Paschen, Casagrandi, Galli, Valerio.
(1862-1932) (1875-1915)
Variola virus
chorioallantoic membrane of embryonating
egg, thin section electron microscopy
page 95

1905 Adelchi Negri, Paul Remlinger, Stanislaus von Prowazek, others proof that variola and vaccinia are viruses
Diagram by P. R. White to illustrate the history of tissue
culture and relationships between workers in the field
Ross Granville Harrison (1870-1959) From: Witkowski JA. Alexis Carrel and the mysticism of tissue culture.
Medical History 1979;23:279-296.
Using expert surgical and aseptic technique developed from successfully operating on tiny frog embryos, Ross Harrison explanted embryonic frog neural tube fragments into
a drop of fresh frog lymph on a sterile coverslip. Once the lymph clotted, he inverted the coverslip over the well in a glass depression slide creating a hanging drop culture, a
technique previously used by microbiologists for studying bacteria. He then watched the development of frog nerve fibers from the neurons in the explanted tissue. In doing
so, he solved several basic cell culture problems: medium, culture vessel, visualization and culture contamination (which he did encounter). Because they were relatively short-
term cultures he was not faced with the problems of feeding or subculturing. As he stated: The immediate object of the experiments was to obtain a method by which the end
of a growing nerve could be brought under direct observation while alive, in order that a correct conception might be had regarding what takes place as the nerve fiber extends
during embryonic development from the nerve center out to the periphery. In 2004, Haig Keshishian stated: Ross Harrisons [1910 paper] is likely the most important paper
ever published in the Journal of Experimental Zoology.... In a single stroke Harrison invented the method of tissue culture, and then used it to prove the neuron doctrine.
(Journal of Experimental Zoology 2004;301a:201-203.)
Harrison R. Observations on the living developing nerve fiber. Anat Rec. 1907;1:116-128. Read before the Society for Experimental Biology and Medicine, New York, 1907.
Harrison R. The outgrowth of the nerve fiber as a mode of protoplasmic movement. J Exp Zool. 1910;9:787-846. page 96

1907 Ross Harrison development of the first cell cultures (nerve fibers from frog embryo tissues)
Aedes aegypti
Ashburn and Craig worked with only one dengue virus, later
identified as DEN-2 virus. They passed diluted blood from febrile
patients through Berkefeld ultrafilters and injected filtrates into
healthy volunteers. In 1943, Hotta and Kimura in Japan isolated
DEN-1 virus. A few months later, Sabin and Schlesinger, working
in Hawaii and New Guinea, isolated DEN-1 and DEN-2 viruses.
DEN-3 and DEN-4 were isolated in the Philippines in 1960 by
William Hammon and his colleagues.
Percy Moreau Ashburn Charles Franklin Craig Ashburn PM, Craig CF. Experimental investigations regarding the
(1872-1940) (1872-1950) etiology of dengue fever. J Infect Dis. 1907;4:440-475.

page 97

1907 Percy Ashburn Charles Craig discovery of dengue viruses


Papillomavirus, negative contrast and thin section electron microscopy
Giuseppe Ciuffo, from the Fourth Italian Congress of Pathology, Paris, 1906: With a series of experiments...
I sought to clarify the problem of the etiology, pathogenicity and contagiousness of cutaneous or mucosal
papillomatous forms, such as the common wart and condyloma acuminatum, which are assuredly infectious. My
principal aim is to be able to find which is the specific microscopic germ or invisible virus that is responsible for
these lesions and what are its characteristics, its mode of transmission from man to man or to animals and finally
to determine whether a comparative clinical, anatomical and bacteriological examination could result in the
identification of the responsible agent. He proceeded to produce a wart on his own hand by the inoculation of a
wart extract which had been ultrafiltered through a Berkefeld N candle!
Ciuffo G. Innesto positivo con filtrado di verrucae volgare. Ital Mal Venereol. 1907;48:12-15. page 98

1907 Giuseppe Ciuffo discovery of human papillomaviruses (the first papillomaviruses)


IMMATURE

MATURE

Vilhelm Ellerman (1871-1924) Oluf Bang (1881-1937)


Vilhelm Ellerman and Oluf Bang, in 1908, demonstrated that an extract ultrafiltered through a
Berkefeld filter could transmit leukemia among chickens. However, the scientific community largely
ignored this finding, as leukemia was not recognized as a neoplastic disease until after 1930. Peyton
Rous work two years later, although not ignored, was met with some indifference. The prevailing
attitude was that tumors that could only grow in chickens must bear little resemblance to human
cancers (which were thought to be noninfectious in origin) and were unlikely to provide any useful
information. Rous was eventually vindicated and his discovery earned him a Nobel Prize in 1966 (he
noted the work of Ellerman and Bang in his Nobel lecture).
Ellerman V, Bang O. Experimentelle leukemia im huhnem. Zentralbl Bakteriol Parasitenkd
Infektionskr Hyg. Abt. I (Orig.). 1908;46:595-609.

page 99

1908 Vilhelm Ellerman, Oluf Bang discovery of avian leukemia viruses (the first leukemia virus)
Several investigators had, early
on, attempted to isolate an
etiologic bacterium from tissues
from poliomyelitis victims, but
all bacteriological findings were
negative; likewise, several attempts
to transmit the disease to the
usual laboratory animals, such
as rabbits, guinea pigs, and mice,
failed. In 1908, Karl Landsteiner
and Erwin Popper (1879-1955)
injected intraperitoneally a
suspension of spinal cord from a
9-year-old boy who had died of
poliomyelitis into two nonhuman
primates, a Hamadryas baboon
(Papio hamadryas) and a rhesus
macaque (Macaca mulatta). The
inoculated material, which was
bacteriologically sterile, yielded
negative results when injected into First electron micrograph of poliovirus , shadow-cast technique,
rabbits, guinea pigs, and mice. The 1953, RCA Laboratories, details unknown, from Sarnoff Library
two nonhuman primates, however,
exhibited lesions in the spinal cord,
medulla, pons, and brain stem that
were indistinguishable from those
in cases of human poliomyelitis.
The macaque developed complete
flaccid paralysis of both legs.
Landsteiner and Popper were
unable to passage the agent, but
this was achieved independently
in 1909 by others. In 1909,
Landsteiner and Constantin
Levaditi of the Institut Pasteur
successfully passed a suspension
Karl Landsteiner (1868-1943) of spinal cord material from a
poliomyelitis case through a
Berkefeld type V ultrafilter.
Landsteiner K, Popper E. Uebertragung der poliomyelitis acuta auf affen. Z Immunittsforsch.
1909;2:377-390.
Landsteiner K, Levaditi C. La transmission de la paralysie infantile aux singes. C R Soc Biol.
1909;67:592-594.
Poliovirus 1, negative contrast electron microscopy
of purified virus, Joseph Esposito, ~1975 page 100

1909 Karl Landsteiner, Erwin Popper discovery of polioviruses (the first enteroviruses)
Constantin Levaditi (1874-1953)
With Karl Landsteiner, Levaditi also discovered the presence of poliovirus
in tissues other than those of the nervous system. He isolated poliovirus in
tissue explants. He authored the first monograph dedicated to the disease, La
Poliomylite aigu pidmique (1913).

Karl Landsteiner (1868-1943)

In 1900 Landsteiner discovered that mixing the blood of two people


usually causes the erythrocytes to agglutinate, and in 1901 he found
that this effect was due to contact of the erythrocytes of one person
with the serum of the other. As a result he succeeded in identifying
the three major human blood groups A, B and O. Landsteiner
also found out that blood transfusion between persons with the
same blood group did not lead to agglutination and destruction of
erythrocytes, whereas this occurred between persons of different
blood groups. Based on his findings, in 1907 the first successful
blood transfusion was performed by Reuben Ottenberg at Mount
Sinai Hospital in New York. In 1930 Landsteiner was awarded
the Nobel Prize in Physiology or Medicine in recognition of these
achievements.

Two of many styles of computer modeling of the poliovirus virion surface


page 101

1909 Constantin Levaditi, Karl Landsteiner discovery of poliovirus in non-nervous system tissues
Global distribution of sandfly (phlebotomus) fever

Robert Doerr (1871-1952)


From the Transactions of the Society of Tropical Medicine and Hygiene, London, 1913 (abridged): In the year 1908, Doerr [Robert Doerr, K. Franz, S. Taussig] announced the
results of his experiments on the infectivity of the blood of the Dalmatian summer fever, and on the mode of the transmission of the virus by the phlebotomus [midge]. Since
then the closely related ailments in Malta [and Italy and Crete] have been investigated in a similar manner. It has been found that : (1) The subcutaneous injection of blood
or serum withdrawn during the first 24 hours of the patients illness caused fever 19 times. (2) Inoculation with the filtrate obtained by passing the diluted blood through a
porcelain candle, which retained the micrococcus melitensis, excited the disease 14 times. (3) Feeding experiments with infected sandflies were successful on 21 occasions.
This evidence is sufficient to show that the fever is specific, and that it is caused by a filter-passing virus, which circulates in the blood during the first day of the illness, and that
it is conveyed by the phlebotomus [midge]. During World War II, sandfly fever affected large numbers of U.S. Army personnel serving in Mediterranean operations. Study of
patients led to a clinical description, confirmation of the vector (Phlebotomus papatasii) and the distinction of the two viruses, namely sandfly fever Sicilian virus and sandfly
fever Naples virus.
Doerr R, Franz K, Taussig S. Das pappatacifieber. Leipzig und Wien: Franz Deuticke; 1909. page 102

1909 Robert Doerr, K. Franz, S. Taussig discovery of sandfly (phlebotomus) fever viruses (the first phleboviruses)
The Fly Room, Columbia University
Thomas Hunt Morgan had become interested in species variation, and in 1911,
he established the Fly Room at Columbia University to determine how a species
changed over time. For the next 17 years, in this 16x23 ft. room, described by many as
cramped, dusty, smelly and cockroach ridden, Morgan and his students did ground-
breaking genetic research using millions of fruit flies (Drosophila melanogaster),
and a combination of statistical-genetic methods (per Mendel) and microscopy
to characterize mutations. Though initially against the idea that the behavior of
chromosomes can explain inheritance, Morgan became the leading supporter of the
idea. Morgan and his students (Alfred Sturtevant, Calvin Bridges, Hermann Muller
and others), developed the ideas, and provided the proof for the chromosomal
theory of heredity, genetic linkage, chromosomal crossing over and non-disjunction
(the occasional failure of homologous chromosomes to segregate during and after meiosis). In 1915, they
published a seminal book, The Mechanism of Mendelian Heredity (Henry Holt and Company, New York,
1915). Its major tenets regarding the functions of the chromosomes as the bearers of heredity were:
1. Discrete pairs of factors located linearly on chromosomes like beads on a string bear hereditary
information. These factorsMorgan would soon call them genessegregate in germ cells and
recombine during reproduction, essentially as predicted by Mendelian laws. However:
Thomas Hunt Morgan (1866-1945) 2. Certain characteristics are sex-linkedthat is, occur together because they are located on the same
chromosome that determines gender. More generally:
Nobel laureate Eric Kandel wrote of Morgan: Much as Darwins 3. Other characteristics are also sometimes associated because, as paired chromosomes separate during
insights into the evolution of animal species first gave coherence germ cell division, genes proximate to one another tend to remain together. But sometimes, as a
to nineteenth century biology as a descriptive science, Morgans mechanistic consequence of reproduction, this linkage between genes is broken, allowing for new
findings about genes and their location on chromosomes helped combinations of traits.
transform biology into an experimental science. These rigorously proven tenants revolutionized our understanding of the processes of life, and of the cause
of many diseases. By demonstrating that genes are the driving forces for the evolution of all life forms,
page 103 Morgan established the correctness of Darwins hypothesis put forward in 1859.

1910> Thomas Hunt Morgan discovery of the nature and role of the chromosome (using Drosophila)
John F. Anderson joined the Marine Hospital Service in 1898, and
in 1909 was appointed director of the Hygienic Laboratory, the
predecessor of the National Institutes of Health. He was among the
early scientists who made this laboratory well-known in scientific
circles.
Joseph Goldberger joined the U.S. Public Health Service in 1899.
From 1902-1906, he held a variety of epidemiology postsin Mexico,
Puerto Rico, Mississippi, Louisiana, all in efforts to combat yellow
fever, typhus, dengue, and typhoid fever. He survived yellow fever and
dengue infections and almost died of typhus.
In 1911, Goldberger and Anderson transmitted measles to monkeys,
showed that it was caused by an ultrafilterable virus, and that infection
was transmitted from nasal and throat secretions. Their 1912 paper
was a landmark. Until then, there had been no experimental system
in which measles virus had been demonstrated to replicate or to
produce a clinical disease similar to that in humans. Indeed, it was
not until 1954, 43 years later, that measles virus was first propagated
successfully in the laboratory in cell culture by John Enders and his
colleagues. Goldberger and Anderson were successful in producing
a febrile response and varying degrees and extent of rash and they
successfully passed the virus to further monkeys. Their experimental
protocols and the descriptions of their work were remarkably
thorough, a model for future investigations.

Anderson JF, Goldberger J. Recent advances in our knowledge of


measles. Am J Dis Children 1912;4:20-26.

John F. Anderson (1873-1958) and Joseph Goldberger (1874-1929)

Multinucleated giant cells (syncytia), typical of


measles virus infection in cell culture
page 104

1911 John Anderson, Joseph Goldberger discovery of measles virus


Joseph Goldberger, middle row, fourth from left.

In 1914, Joseph Goldberger was asked by US Surgeon General Rupert Blue to investigate pellagra,
an endemic disease in the Southern U.S. Goldbergers theory that pellagra was associated with diet
contradicted the then commonly-held medical opinion that pellagra was an infectious disease. After
multiple restricted-diet experiments with groups of volunteers spanning several years, Goldberger
was able to demonstrate that individuals who consumed heavily corn-based diets (to the virtual
exclusion of other foods) were at a greatly increased risk of contracting pellagra. Despite his careful
experiments, Goldbergers discovery proved socially and politically untenable and he made little
progress in securing support for treating pellagra. However, with further research, in 1926 Goldberger
was able to demonstrate that a vitamin B deficiency was the cause of pellagra. Conrad Elvehjem, in
Joseph Goldberger (1874-1929) 1937, discovered the specific deficitpellagra is caused by a lack of the B vitamin niacin along with
insufficient levels of the essential amino acid tryptophan.

Goldberger was nominated five times for the Nobel Prize for his work on the etiology of pellagra.
Unfortunately, he died from cancer in January 1929, so his nomination was disqualified. Christiaan
Eijkman and Frederick G. Hopkins, who were selected for the Nobel Prize later that year for their work
on vitamins, stated that Goldberger would have been a most deserving companion.

page 105

1911 John Anderson, Joseph Goldberger discovery of measles virus


Francis Peyton Rous (1879-1970) Joseph Willis Beard (1906-1983) Joseph Beard and his wife Dorothy
with chickens used for research
Peyton Rous, Nobel Lecture, 1966: In 1910 I described a malignant chicken sarcoma which could be propagated by transplanting its
cells, these multiplying in their new hosts and forming new tumors of the same sort. In other ways the growth showed itself to be a
neoplasm of a classical sort, yet, as reported in 1911, its cells yielded a causative virus. Numerous workers had already tried by then
to get extraneous causes from transplanted mouse and rat tumors but the transferred cells had held their secret close. Hence the
findings with the sarcoma were met with down-right disbelief, though soon several other, morphologically different, spontaneous
chicken tumors were propagated by transplantation and from each a virus was got causing growths of its kind. Not until after some
15 years of disputation amongst oncologists were the findings with chickens deemed valid, and then they were relegated to a category
distinct from that of mammals because from them no viruses could be obtained. Only in 1925, through the efforts of a British worker,
W.E. Gye, was much attention given them by scientists. The virus causing the chicken sarcoma first studied, now generally termed
RSV, has been maintained for more than fifty-five years and is still studied in many countries.

page 106

1911 Francis Peyton Rous, Joseph Beard discovery of Rous sarcoma virus (first solid tumor virus)
Nobel Laureates Who Laid the Foundation
for the Use of Radioisotopes in Virology
1901 Wilhelm Rntgen 1936 Carl Anderson
1903 Henri Becquerel 1938 Enrico Fermi
Marie Curie 1939 Ernest Lawrence
Pierre Curie 1943 George de Hevesy
1906 Joseph Thomson 1944 Otto Hahn
1908 Ernest Rutherford 1946 Hermann Muller
1911 Marie Curie 1951 Glenn Seaborg
1918 Max Planck Edwin McMillan
1921 Frederick Soddy 1956 John Bardeen
1921 Albert Einstein Walter Brattain
1922 Niels Bohr William Shockley
1924 William Einthoven 1959 Emilio Segre
1927 Arthur Compton 1961 Robert Hofstadter
Charles Wilson 1968 Luis Alvarez
1934 Harold Urey 1977 Rosalyn Yalow
1935 Frederic Joliot Roger Guillemin
Irene Joliot-Curie Andrew Schally
1935 James Chadwick

Isotopes Commonly Used For Biological Radiolabelling

Isotope Half Life Emission Energy Detection on Gel


Hevesy G. The absorption Beta Emitters:
and translocation of lead by 3
H 12.4 years very low Fluorography
plants: a contribution to the 14
C 5740 years moderate Autoradiography
application of the method of 35
S 87.4 days moderate Autoradiography
radioactive indicators in the 33
P 25 days moderate Autoradiography
investigation of the change of 32
P 14.3 days high Intensifying Screen
substance in plants. Biochem J. Gamma Emitters:
1923;17: 439-445. 125
I 80 days high Intensifying Screen
George de Hevesy (1885-1966)
George de Hevesys introduction to practical radiochemistry came in Ernest Rutherfords laboratories at the University of Manchester. His work there, and in Vienna,
Budapest and Copenhagen, centered on the use of radium and lead isotopes. In 1923, in Copenhagen, he and Dirk Coster discovered the element hafnium (Latin Hafnia for
Copenhagen) in Mendeleevs periodic table a chemical element with 72 protons was missing and they concluded that there must be such an element a mineralogical
museum furnished the material for the research and characteristic X-ray spectra indicated that a new element was present this earned him the 1943 Nobel Prize in
Chemistry. [When Denmark was invaded in World War II, de Hevesy dissolved the gold Nobel Prizes of Max von Laue and James Franck with aqua regia (a mixture nitric
acid and hydrochloric acid) to prevent the Nazis from stealing them. He placed the resulting solution on a shelf in his laboratory at the Niels Bohr Institute. After the war, he
returned to find the solution undisturbed and precipitated the gold out of the acid. The Nobel Society then recast the Nobel Prizes using the original gold.] In 1923, De Hevesy
also started his pioneering work on the use of radioisotopic tracers to study metabolic processes of plants and animals he published the first study on the use of the naturally
radioactive 212Pb as a tracer to follow the absorption and translocation of atoms in the roots, stems and leaves of the broad bean plant. Much similar tracer biology followed.
page 107

1911> George de Hevesy, others development of radioisotopic labeling


Two technicians at work in Alexis Carrels laboratory at the Rockefeller Institute, ~1936. They are dressed in the full-
length, black, hooded gowns adopted by Carrel. Between them is a rack of Carrel flasks that are being filled by the
technician on the right while the technician on the left is flaming rubber bungs prior to sealing the flasks. Carrels critics
continued: Tissue culture, although a delicate and exacting technique and one in which vigorous asepsis is absolutely
essential, has gained a spurious and unfortunate reputation for difficulty and almost for mysticism. From: Witkowski JA.
Alexis Carrel and the mysticism of tissue culture. Med Hist. 1979;23:279-296.

From Time magazine, 1938: A secluded labyrinth of black, dustless, germless laboratories
zigzags across the top floor of the main building of the Rockefeller Institute for Medical
Research in Manhattan. Black are the floors, black the furniture, dark grey the windowless
walls, shadowless the bleak illumination that comes through the skylights. Entrance to this
aseptic, dustless, reflectionless hideaway is by a spiral staircase from an anteroom on the
floor below. Only scientists particularly interested in fractioning life to its lowest common
denominators may mount that spiral. And all must wash their hands and faces, put on
gowns and hoods of black clothall except the master of this pure and dark domain, master
of its purified and black-clad servants. He, the most famed member of the Rockefeller
Institute, Dr. Alexis Carrel, distinguishes himself from the others with a little white
headpiece that looks like the hat of a U.S. bluejacket [U.S. Navy sailor].
page 108

1912> Alexis Carrel, Hugh and Mary Maitland, others development of many cell and tissue culture methods
Extending from his work
with cell/tissue culture,
Alexis Carrel worked with
Charles Lindbergh in the
mid-1930s to create a
perfusion pump, to allow
a diseased organ to be
removed, kept alive while
being repaired, and then
returned to the patient.
They thought that if this
process could be repeated
forever, it could potentially
render humans immortal.

Alexis Carrel (1873-1944)


In 1907, Ross Harrison of Yale University published a short note entitled Observations on the Living Developing Nerve Fibre, that aroused widespread interest, including by
Alexis Carrel and his colleagues at the Rockefeller Institute. Carrel became a pioneer in the development of tissue culture and also became its chief publicist. He made a
number of practical technical contributions, but more so greatly influenced his contemporaries. His tissue culture techniques were influenced by his surgical expertise and
they became increasingly complicated. Critics said that Carrels work, caused the method to be wrapped up from the beginning in a considerable cocoon of mumbo-jumbo,
derived from the practices that were prevalent at that time in the operating theatres of the world. Tissue culture, although a delicate and exacting technique and one in
which vigorous asepsis is absolutely essential, is gaining a spurious and unfortunate reputation for difficulty and almost for mysticism. Nevertheless, Carrel grew cells without
antibiotics for many years, which was in itself a considerable technical feat his tissue culture (explant culture) of a chicken heart embryo is said to have survived 34 years,
although the veracity of this claim is doubted by statements that chick heart cells were used in the constantly replaced nutritional medium. [Every year newspapers would run
a Happy Birthday column, celebrating the life of the culture.] Carrels development of more and more complexity in techniques dominated the field and led to a decline in
interest in tissue culture, which persisted for many years, until others developed simpler methods. The Nobel Prize in Physiology or Medicine of 1912 was awarded to Alexis
Carrel in recognition of his work on vascular sutures and the transplantation of blood vessels and organs, not for his later work with cell/tissue culture.
Abridged from: Witkowski JA. Alexis Carrel and the mysticism of tissue culture. Med Hist. 1979;23:279-296.
page 109

1912> Alexis Carrel, Hugh and Mary Maitland, others development of many cell and tissue culture methods
Except for the work of Alexis Carrel and his colleagues at the Rockefeller Institute, cell culture had little impact on virology
research prior to the 1940s. However, key technical developments were made all along, most importantly, in 1928, by Hugh
Bethune Maitland and Mary Cowan Maitland, who devised a simplified method of growing vaccinia and a few other viruses
in suspension cultures of fresh minced adult chicken kidneys, in flasks, using a substrate of chicken serum and organic salts.
Thomas Rivers, Max Theiler and their colleagues introduced further refinements to the Maitlands technique and throughout
the 1930s demonstrated the cultivation of fowlpox, foot-and-mouth disease and yellow fever viruses, and the etiologic agent of
epidemic typhus, Rickettsia prowazekii. In 1936, George Gey developed roller tube culture methods, which became standard
for years. Albert Sabin and Peter Olitsky, in 1936, successfully cultivated poliovirus in human embryonic brain tissue and
there were other pioneering successes, but there was no general usage until the breakthroughs made in the laboratory of John
Enders. In 1948, Thomas Weller, Frederick Robbins and Enders successfully propagated mumps and influenza A viruses,
using a modification of the Maitlands method, before moving on to polio and measles viruses. One key to the successes
in the Enders laboratory was the addition of penicillin and streptomycin to culture media. Others rapidly followed these
initial, landmark successes. Enders coined the term cytopathic effect (CPE), that is, the visualization by low magnification
microscopy of cell death, a key to the practicality of titrating and serotyping viruses. Colorimetric indicators of pH change of
media caused by cell death were also introduced at this time, again making practical large surveys of the presence of viruses
and/or antibodies. Minimum Essential Medium (MEM), developed in 1959 by Harry Eagle, was a monumental breakthrough;
it still is one of the most widely used of all synthetic cell culture media. Early attempts to cultivate normal mammalian
fibroblasts and certain subtypes of HeLa cells revealed that they had specific nutritional requirements that could not be met
by Eagles initial Basal Medium (BME) additions to BME were made to aid growth of a wider variety of fastidious cells. MEM
incorporated the first of these modifications, included higher concentrations of amino acids so that the medium more closely
approximated the protein composition of mammalian cells. Since Eagles pioneering work, many, many different media have
Hugh Bethune Maitland been developed, most stemming from research into the nutritional requirements of particular cells, some for commercial
(1895-1972) proprietary purposes, and some to meet the requirements of different culture systems (e.g., suspension culture systems).

Composition of a Medium Suitable for Cultivation of Mammalian Cells [1960s]


Amino acids Vitamins Salts Other Proteins
(Serum-Free Media)
Arginine biotin NaCl glucose insulin
Cystine choline KCl penicillin transferrin
Glutamine folate NaH2PO4 streptomycin specific growth factors
Histidine nicotinamide NaHCO3 phenol red
Isoleucine pantothenate CaCl2 whole serum
Leucine pyridoxal MgCl2
Lysine thiamine
Methionine riboflavin
Phenylalanine
Threonine
Trytophan
Tyrosine
Valine

Maitland HB, Maitland MC. Cultivation of vaccinia virus without tissue culture.
Lancet 1928;2:596-597.
Eagle H. Nutritional needs of mammalian cells in culture. Science 1955;122:501-504.
Enders JF, Weller TH, Robbins FC. Cultivation of the Lansing strain of poliomyelitis
Mary Logan Cowan Maitland Harry Eagle virus in cultures of various human embryonic tissues. Science 1949;109:85-87.
(~1896-1972) (1905-1992) page 110

1912> Alexis Carrel, Hugh and Mary Maitland, others development of many cell and tissue culture methods
First cultivation of a virus in cell culture:
vaccinia virus cellular outgrowths from
explants of rabbit and guinea pig cornea

Primary rabbit cornea cells

Edna Steinhardt Harde (1874-1941)


(photo from 1920 passport application)
Steinhardt E, Israeli C, Lambert RA. Studies on
the cultivation of the virus of vaccinia. J Infect
Dis. 1913;13:294-300.

Edna Steinhardt Harde is perhaps the most


accomplished virologist / microbiologist
to remain largely unrecognized. She led an
incredibly engaged, adventuresome and creative
life including being torpedoed while crossing
the English Channel during World War I on her
way to serve with the American Red Cross in Robert A. Lambert (1883-1960)
France and later with the Institut Pasteur she [Rockefeller Archives, used with permission]
deserves a full biography!
page 111 No photo of Clara Israeli was found.

1913 Edna Steinhardt Harde, Clara Israeli, Robert Lambert first cultivation of a virus in a cell culture (vaccinia)
... and the first demonstration of virus neutralization (vaccinia
virus, rabbit corneal epithelium cell culture, and serum from
previously infected vs. uninfected rabbits) page 112

1913 Edna Steinhardt Harde, Clara Israeli, Robert Lambert first cultivation of a virus in a cell culture (vaccinia)
Y. Hiro
(bronze, Hokkaido University, Sapporo)

Rubella virus
cell culture, thin section electron microscopy

In 1914, Alfred Hess showed that rubella was caused by a virus, based on work with monkeys. In
1938, Hiro and Tasaka confirmed this by passing the virus to children using ultra-filtered nasal
washings from acute cases.
Hess A. German measles (rubella): an experimental study. Archives of Internal Medicine
Alfred Fabian Hess (1875-1933) 1914;13:913-916.
Hiro Y, Tasaka S. Die rteln sind eine viruskrankheit. Monatsschr. Kinderheilk 1938;76:328.

Hess was a staff member of University & Bellevue Hospital Medical College, New York University. He
spent mornings at the Rockefeller Institute and afternoons at the Babies Hospital and Bellevue. He
also made seminal discoveries concerning rickets and scurvy.

page 113

1914-1938 Alfred Hess, Y Hiro, S Tasaka discovery of rubella virus (the only rubivirus)
Discovery and Rediscovery of Trypsinization for the Dispersal of Cells in Culture

1916: FP Rous and SF Jones - dispersal of cell clumps using trypsin


1953: W Scherer, JT Syverton and GO Gey - dispersal of HeLa cells using trypsin
1953: AW Frisch and V Jentoft - dispersal of monkey testicular tissue using trypsin
1954: JS Youngner - dispersal of monkey kidney cells using trypsin
(a key in the production of inactivated polio vaccine)

page 114

1916> Development of trypsinization for the dispersal of cells in culture


T4 bacteriophage

Twort FW. An investigation on the


nature of ultra-microscopic viruses.
Lancet 1915;2:1241-1243.

dHerelle F. Sur un microbe invisible


antagonistic des bacilles dysenterique.
C R Acad Sci Paris 1917;165:373-375.

Duckworth DH. Who discovered


bacteriophage? Bacteriol Rev.
1976;40:793-802.
Frederick William Twort (1877-1950) Flix dHerelle (1873-1949)
In 1915, British bacteriologist Frederick Twort, working in London, discovered a glassy transformation of bacterial cultures. Among other ideas, he considered that the
transformation might be caused by a virus that grew in and destroyed the bacteria: (a) affected colonies would not grow on any medium; (b) examination of the glassy areas
revealed no bacteria; (c) if a pure culture of the bacteria was touched to one of the glassy colonies, the growth at the point touched started to become transparent and gradually
made the whole colony transparent; (d) after filtration of the glassy material through a Chamberland candle, it retained its ability to cause the same glassy transformation;
and (e) the glassy transformation could be conveyed to fresh cultures for an indefinite number of generations. Tworts work was interrupted by the onset of World War I and
a shortage of funding; seemingly he never got back to this work. Independently, French-Canadian microbiologist Flix dHrelle, working at the Pasteur Institute in Paris,
announced in 1917, that he had discovered an invisible, antagonistic microbe of the dysentery bacillus. In a flash I had understood: what caused my clear spots was in fact
an invisible microbe ... a virus parasitic on bacteria. DHrelle called the virus a bacteriophage but there was difficulty in defining this. Much work followed, leading to our
present understanding of the nature of bacteriophages, but there also developed a long-running public controversy between dHerelle and defenders of Twort over the priority
of discovery. This story has been wonderfully told by Donna Duckworth in her 1976 review, Who discovered bacteriophage?
page 115

1916-1919 Frederick Twort, Flix dHerelle discovery of bacteriophages


Montgomery RE. On a
tick-borne gastroenteritis
of sheep and goats
occurring in British East
Africa. J Comp Path.
1917;30:28-57.

Daubney R, Hudson JR.


Nairobi sheep disease.
Parasit. 1934;23:507-524.

Nairobi sheep disease virus, liver, mouse


Robert Eustace Montgomery (1880-1932) thin section electron microscopy
In 1917, Robert Montgomery described a tick-borne gastro-enteritis of sheep and goats, which was eventually named Nairobi sheep disease. Working at the Kenya Agricultural
Research Institute at Kabete, near Nairobi, he showed that the causal agent was an ultrafilterable virus, transmitted by Rhipicephalus appendiculatus ticks. In succeeding years
his work was confirmed in detail. Nairobi sheep disease has been found across large areas of East and Central Africa, causing disease in sheep and goats with mortality rates
reaching 90%. Some human disease has been associated with the livestock disease, but its geographic range, prevalence rate and clinical disease data are incomplete. One
intriguing finding has been that Ganjam virus, first isolated in 1954 in India as the cause of sheep and goat disease, and also as the cause of widespread mild and severe human
disease, is serologically closely related to Nairobi sheep disease virus this has been confirmed by genomic sequencing: Ganjam virus is just an Asian variant of Nairobi sheep
disease virus. This finding, in turn, has pointed to failings in the classical USDA regulatory system used to proscribe the import of foreign animal disease agents although
work on Nairobi sheep disease virus has always been prohibited, virologists in several laboratories worked for many years on Ganjam virus, in many instances knowingly, while
federal regulatory officals remainded oblivious of the serologic and genomic data. page 116

1917 Robert Montgomery discovery of Nairobi sheep disease virus (the first nairovirus, the first tick-borne virus)
Influenza epidemic, Boston, 1918

Camp Funston (now Fort Riley), Kansas, influenza epidemic, 1918

The only spike visible on the


20th century mortality graph
of the U.S. is the 1918-1919
influenza epidemic.

People praying, influenza epidemic, 1918

page 117

1918-1919 influenza pandemic, 40-100 million deaths worldwide


The Discovery of Influenza Virus
The history of the discovery of influenza virus is more complex than that of most other viruses, intermixed with the great pandemic of 1918, confounded by many
contradictory claims, involving more than the usual number of players, and forced to overcome the insistence at the turn of the twentieth century by the prominent
bacteriologist, Richard Friedrich Johannes Pfeiffer (1858-1945), that the disease is caused by a bacterium, Pfeiffers bacillus (then Bacillus influenza, now Haemophilus
influenzae). By 1922, the notion that influenza was caused by an ultrafilterable virus had gained ground: the great microbiologist, Hans Zinsser (1878-1940) wrote in his
Textbook of Bacteriology (D. Appleton & Co.), All evidence must be considered in connection with recent experiments upon the possibility of causation by a filterable virus
He cited several studies from 1918-1919, studies done during the great pandemic, including: (1) Charles Nicolle (1866-1936) and Charles Lebailly, of the Institut Pasteur
Tunis, who ultrafiltered human blood and nasal secretions from uncomplicated grippe patients and instilled the filtrates into conjunctival sacs and nasal cavities of monkeys
(sic Macaccus Sinicus) and human volunteers, obtaining flu-like clinical signs in most. (2) Ren Dujarric de la Rivire (1885-1969), who ultrafiltered blood from four
influenza patients and injected the combined filtrate into himself, clearly obtaining a flu-like disease. (3) E. Leschke, who also produced flu-like clinical signs in monkeys with
ultrafiltered human nasal secretions. (4) H. Selter, who ultrafiltered nasal secretions from cases and sprayed the filtrate into his own throat and that of an assistant, producing
flu-like disease. (5) T. Yamanouchi, K. Sakami and S. Iwashima, of the Government Institute For Infectious Diseases, Tokyo Imperial University (now the Institute of Medical
Science, University of Tokyo), who carried out the most extensive experiments, processing nasophyrngeal secretions from 43 patients, ultrafiltering half, and instilling each into
the nose of 24 volunteers (six of those receiving the ultrafiltered material had just recovered from influenza) all of the 24, except those who had had influenza recently, came
down with a flu-like disease after an incubation period of two or three days. They did other experiments as well, confirming their initial impressive findings. (6) Peter Kosciusko
Olitsky (1886-1964) and Frederick L. Gates (1886-1933), who early in the pandemic instilled ultrafiltered material from patients into rabbits intratracheally, causing fever,
leucopenia, minute pulmonary hemorrhage, and pulmonary edema and emphysema. [This remains unexplained, since human influenza viruses usually do not cause disease
in rabbits.] Several other research groups published papers refuting the ultrafilterability of the etiologic agent. Then, 13 years later, a key breakthrough came when Richard E.
Shope (1901-1966), of the Rockefeller Institute, discovered the etiology of swine influenza. Shopes work stimulated American and British research groups to take up, once
again, the search for the cause of human influenza. In 1933, Alphonse Raymond Dochez (1882-1964) and colleagues, also of the Rockefeller Institute, produced apparent
influenza via human nasopharyngeal inoculation and succeeded in cultivating and serially passing a virus in primary chick embryo cultures, demonstrating that passage
material still produced human disease. A British group that had been collaborating with Dochez, led by Christopher Howard Andrewes (1896-1988), Wilson Smith (1897-
1965) and Patrick Playfair Laidlaw (1881-1940), reported the isolation and serial propagation of human influenza virus in ferrets; their model system was used to confirm the
etiology of the disease by irrefutable modern methods. An explosion of research followed, which has continued unabated until the present time.

Overview: Taubenberger JK, Hultin JV, Morens DM. Discovery and characterization of the 1918 pandemic influenza virus in historical context. Antivir Ther. 2007;12: 581-591.
Nicolle C, Lebailly C. Quelques notions exprimentales sur le virus de la grippe [Certain experimental ideas about the infectious agent of influenza]. Comptes Rendus de
lAcadmie des Sciences 1918;167:607-610.
Dujarric de la Rivire M. La grippe est-elle une maladie virus filtrant? [Is influenza a disease of filter-passing microbes?]. Comptes Rendus de lAcadmie des Sciences
1918;167:606-607.
Leschke E. Untersuchungen zur aetiologie der grippe [Studies on the aetiology of influenza]. Berliner Klinische Wochenschrift 1919;56:11-12.
Selter H. Zur aetiologie der influenza [On the aetiology of influenza]. Deutsche Medizinische Wochenschrift 1918;44:932-933.
Yamanouchi T, Sakakami K, Iwashima S. The infecting agent in influenza. an experimental research. Lancet 1919;1:971.
Olitsky P, Gates F. Experimental study of the nasopharyngeal secretions from influenza patients. J Am Med Assoc. 1920;74:1497-1499.
Shope R. Swine influenza. I. Experimental transmission and pathology. J Exp Med. 1931;54:349-359; and Shope R. Swine influenza. III. Filtration experiments and etiology. J
Exp Med. 1931;54:373-385.
Smith W, Andrewes C, Laidlaw P. A virus obtained from influenza patients. Lancet 1933;2:66-68.
Dochez A, Mills K, Kneeland Y. Studies of the etiology of influenza. Proc Soc Exp Biol Med. 1934-1935;30:1017-1022.

page 118

1918-1919 Charles Nicolle, Charles Lebailly, Ren Dujarric de la Rivire, others discovery of influenza virus
Ren Dujarric de la Rivire
Charles Nicolle (1866-1936) T. Yamanouchi (1885-1969)

Peter Kosciusko Olitsky Alphonse Raymond Dochez


page 119
(1886-1964) (1882-1964)

1918-1919 Charles Nicolle, Charles Lebailly, Ren de la Rivire, T. Yamanouchi, others discovery of influenza virus
Anton Breinl (1880-1944) John Burton Cleland (1878-1971) Eric Lancelot French (1914-2004)
Murray Valley encephalitis virus was the first indigenous pathogenic virus isolated in Australia. In 1918 Anton
Breinl in Townsville infected monkeys intracerebrally with material from fatal human cases. In the same year,
John Burton Cleland, from Sydney, isolated the virus in the Murray River valley by passage of human brain
material in monkeys and sheep, but in both instances isolates were not kept. The virus was next isolated from
brain material from a fatal case by Eric French during the 1950-51 epidemic in the Murray-Darling basin. He
identified the virus as belonging to the Japanese encephalitis virus group, genus Flavivirus, family Flaviviridae.
Australian virologists established the courteous tradition of citing all three investigators when listing the
discoverers of the virus.
Breinl A. Clinical, pathological and experimental observations on mysterious disease, clinically aberrant form
of acute poliomyelitis. Med J Aust. 1918;1:209-213, 229-234.
Cleland JB, Campbell AW. Nature of recent Australian epidemic of acute encephalomyelitis. Med J Aust.
1919;1:234-236.
French EL. Murray Valley encephalitis - isolation and characterization of the aetiological agent. Med J Aust.
1952;1:100-103.
William C. Reeves and Australian researcher Malcolm Simpson collecting mosquitoes
from pools of standing water in the Murray Valley, Australia, during the 1952 epidemic page 120

1918 Anton Breinl, John Cleland, Eric French discovery of Murray Valley encephalitis virus
Herpes simplex virus 1
Arnold Lwenstein (1882-1952) budding from nuclear membrane, cell culture, thin section electron microscopy
In 1913, Wilhelm Grter (1882-1963), director of an ophthalmology clinic in Marburg, Germany initiated a series of studies in rabbits that demonstrated the infectious nature
of herpes simplex virus. He showed that the etiologic agent could be transmitted serially from rabbit to rabbit, and then in 1919 he transmitted the etiologic agent back to
the cornea of a blind man. [This led to the Grter test for herpes, used up until the 1940s, which involved swabbing vesicular fluid from a patients herpetic lesions onto the
cornea of a rabbit.] The following is from a 1932 review by Margaret Holden: Lwenstein showed that the infectious agent in the fluid of herpes vesicles is ultrafilterable, and
this was fully verified by later work of Luger & Lauda (1921), Blanc & Caminopetros (1921) and Levaditi, Harvier & Nicolau (1922). Many investigators had difficulty in filtering
herpes virus. Levaditi and Nicolau concluded that this was not necessarily due to the size of the virus particles, but to adsorption of the particles to the filter. According to
Rivers (1928), the electrical charge on the virus, the electrical charge on the filter, the adsorption of the virus by aggregates of protein or by cell detritus, the amount of protein
or other substances in the virus emulsion [etc.] . . . serve to influence the results of all filtration experiments. Recently Ward and Tang (1929) by emulsifying herpes virus in
broth instead of saline solution have been able to recover the virus consistently from the filtrate of a Berkefeld V candle.
Lwenstein A. Aetiologische untersuchungen ber der fieberhaften, herpes. Munch Med Wochenschr. 1919;66:769-770.
Holden M. Nature and properties of herpes virus. Journal of Infectious Diseases 1932;50:218-236.
McNair Scott TF. Historical aspects of herpes simplex infections. International Journal of Dermatology. 1986;25:63-70.
page 121

1919 Arnold Lwenstein discovery of herpes simplex virus 1


The term adjuvant is derived from the Latin word adjuvare, which means to aid or
to help it was coined by Gaston Ramon, who in 1924 developed an anti-tetanus
vaccine consisting of tetanus toxin treated with formaldehyde and heat. He proposed
that the efficacy of this vaccine was also enhanced by substances (adjuvants) such
as aluminium hydroxide. In 1926, A.T. Glenny confirmed the adjuvant activity of
aluminum compounds utilizing an alum-precipitated diphtheria toxoid. In the
mid-1930s, Jules Freund (1890-1960) developed a powerful adjuvant composed of
a water-in-mineral oil emulsion containing killed mycobacteria [Freunds complete
adjuvant (FCA)]. Although FCA is one of the most potent adjuvants known, it is
quite toxic, precluding its use in human vaccines, and now in most experimental
animal studies. Currently, only aluminum salt/gel-based (alum) adjuvants are
licensed for human use in the United States. Many of the newer vaccine candidates
are based on protective antigens, which are inherently less immunogenic than the
inactivated or live-attenuated virus vaccines of the past. Therefore, adjuvants have
become an increasingly important component of vaccines being developed today.
A number of novel adjuvants have been under development or are in preclinical
and clinical trials some have been approved in other countries. Adjuvants may
be divided into two classes: those with mechanisms affecting delivery systems and
those that are immunopotentiators:

Examples of Adjuvants, Divided by Their Class of Action


Antigen delivery systems Immunopotentiators
Insoluble aluminum compounds MPL (monophosphoryl lipid A) and
Calcium phosphate synthetic derivates
Liposomes MDP (muramyl di-peptide) and
Virosomes derivatives
ISCOMS (structured complexes of Oligonucleotides (CpG, etc.)
saponins and lipids) Double-stranded RNA (dsRNA)
Microparticles (e.g., PLG Alternative pathogen-associated
Gaston Ramon (1886-1963) - polylactide-co-glycolide molecular patterns (PAMPs) (E.
microparticles) coli heat labile enterotoxin (LT);
Gaston Ramon, a long time staff member of the Institut Emulsions [e.g., MF59, flagellin)
Pasteur, also served as one of the first Directors General microfluidized detergent stabilized Saponins (Quils, QS-21)
of the Office International des Epizooties (OIE the squalene oil-in-water emulsion), Small-molecule immune potentiators
World Organization for Animal Health), the world Montanides [ISA-51 (stabilized (SMIPs)
regulatory body for animal disease prevention and water-in-oil emulsion) and ISA-720 Cytokines & chemokines
control. (stabilized water/squalene)]
Virus-like particles
Ramon and many others advanced (1) the use of & viral vectors
formaldehyde to inactivate viruses for vaccines and
toxins for toxoids, (2) the means for measuring vaccine Formaldehyde bottle from
potency, (3) the use of the first adjuvants, and many Jonas Salks laboratory
other technological approaches. 1952
page 122

1920s> Gaston Ramon, others improvements in many vaccine technologies


Peter Kosciusko Olitsky (1886-1964) Jacob Traum (1923-1995)
United States Department of Agriculture; Technical Bulletin TB0076; Report of the Foot-and-Mouth Disease Commission of the USDA, 1926: This extensive unsigned report
contains the results of a major research project on the nature of foot-and-mouth disease virus(es) and vesicular stomatitis virus(es). Proof that vesicular stomatitis is caused
by specific ultrafilterable agents (VSV-NJ and VSV-IND were already known as distinct entities) is presented in incredible detail. A section of the Bulletin entitled Filtration
Experiments 1925-26 states that J. R. Mohler and W. E. Cotton had earlier failed to show that VSV-NJ was ultrafilterable. In the many experiments reported in this Bulletin,
the VSV-NJ virus used was from fluid obtained from vesicular lesions on guinea pigs, horses or heifers and ultrafiltrates were tested by injecting guinea pigs which were then
evaluated for illness and lesions at 48 hours (using Bacillus prodigiosus as a filter control). In 13 of the first 18 filtration tests done in 1925 using Berkefeld type V candles the
virus passed through the filters. Then Berkefeld type N candles (finer than type V) were used: in 6 of 10 experiments the virus passed through the candles. Then Chamberland
bougies (candles) L3, L7 and L11 were used in a series of experiments also shown in tabular form: This experiment demonstrates that the virus can traverse the L3 and L7
types of Chamberland filters but not under ordinary conditions the L11.
Oltsky PK, Traum J, Schoening HW. Comparative studies on vesicular stomatitis and foot and mouth disease. J Am Vet Med Assoc. 1926;70:147-167. [Materials: Two strains
of vesicular stomatitis virus (Indiana, New Jersey), samples sent us through the kindness of Dr. W. E. Cotton, United States Bureau of Animal Industry, who had propagated the
strains in guinea pig foot pads for several years. William Edwin Cotton (1866-1951), Assistant Superintendent, Bureau of Animal Industry, USDA, Bethesda, Maryland)]
page 123 Cotton WE. Vesicular stomatitis and its relation to the diagnosis of foot-and-mouth disease, J Am Vet Med Assoc. 1926;69:313-332.

1926 Peter Olitsky, Jacob Traum, Harry Schoening, William Cotton discovery of vesicular stomatitis viruses
African swine fever virus, thin section electron microscopy
Many aspects of African swine fever virus are unusual, more than unusual enough
Robert Eustace Montgomery (1880-1932) to warrant it being placed as the sole member of a genus, Asfivirus, and family,
Asfarviridae. The virus exhibits some similarities in genome structure and strategy of
replication with the poxviruses and phycodnaviruses, but it has a quite different virion
structure. It is transmitted by ticks (soft ticks, genus Ornithodoros), and is extremely
Montgomery RE. On a form of swine
pathogenic in domestic pigs. Pigs respond to infection with such a minimal immune
fever occurring in British East Africa
response that it seems they cannot recognize the virus at all. It has been asked with
(Kenya Colony). J Comp Pathol.
tongue-in-cheek, Is this a virus from outer space? Not really, but recent focus on the
1921;34:159-191.
nucleocytoplasmic large DNA viruses (NCLDV) will bring new attention to African
swine fever virus. page 124

1921 Robert Montgomery discovery of African swine fever virus (the only asfarvirus)
Hans Gerhard Creutzfeldt (1885-1964) Alfons Maria Jakob (1884-1931)

In 1921, a German neurologist/psychiatrist, Alfons Maria Jakob, described a clinicopathological disease in 6 patients
with spasticity and progressive dementia associated with neural degeneration (characterized pathologically by neuronal
loss, spongiform changes, and astroglial proliferation). Shortly thereafter, a German neuropathologist, Hans Gerhard
Creutzfeldt, independently published on a similar case. Jakob, a famous neurological scientist, gave credit to Creutzfeldt
for describing the syndrome first without realizing he had stumbled onto a new syndrome hence, the compound name.
Creutzfeldt-Jakob disease (CJD) is closer to a neuropathological syndrome than a single etiologic-nosological entity.
Some 84 forms, (types or variants) of CJD have been recorded on the basis of clinical and neuropathological criteria.
The question has been raised whether the cases that Creutzfeldt and Jakob described were instances of classical CJD
there are clinical, neurological and neuropathological reasons for doubting several of the cases, but most neurologists
who have studied the case records in detail believe that at least most of Jakobs cases, if not Creutzfeldts initial case, were Creutzfeldt-Jakob disease, cerebrum, H&E
indeed classical CJD.
page 125

1921 Hans Creutzfeldt, Alfons Jakob initial description of Creutzfeldt-Jakob disease


The history of the development of the ultracentrifuge intersects with basic physics and
chemistry as they helped create the field of molecular biology. Simple centrifuges have
drawbacks that stem from the basic physics of a shaft-mounted rotor a spinning shaft-
mounted rotor running at speeds greater than a few hundred revolutions per second (rps) causes
a problem much like wheel imbalance in a car there is an inability to match the inertial axis
of the rotor exactly with the axis of its motor shaft resulting in uncontrollable shaking. Carl de
Laval, a Swedish engineer, overcame some of these limitations in 1883, when he developed a
centrifuge that featured a steam-powered rotor on a flexible shaft. Soon it was found that such
a centrifuge was too slow to separate the macromolecules that increasingly drew the attention
of scientists. In the 1920s, Theodor Svedberg at Uppsala University became interested in the
problems of centrifugation he was a colloid chemist working on proof of a theory by Albert
Einstein concerning Brownian motion of large molecules. It was through his studies of colloid
sedimentation that Svedberg came to invent the first ultracentrifuge, an instrument powerful
enough to separate macromolecules. By 1925, Svedberg and colleagues had constructed a
centrifuge with a maximum speed of 42,000 rpm. He later housed the rotor in a low-pressure
hydrogen atmosphere, where he was able to subject samples to forces up to a million times that
of gravity. The sample cells were constructed of rock crystal mounted on top of a vertical shaft,
all enclosed in a temperature controlled chamber. The housing had two windows for a beam of
UV light to shine through the sample cells during rotation and onto film in a special UV camera.
Using this ultracentrifuge, Svedberg calculated the molecular weights of hemoglobin and casein
as well as his colloids. The svedberg unit (symbol S, sedimentation coefficient), technically a
measure of time, defined as exactly 10-13 seconds (100 fentoseconds), is named after him.

Theodor H.E. Svedberg (1884-1971)

Th Svedberg with part of his massive oil-turbine driven


ultracentrifuge, Uppsala University, 1932. The rest of the
instrument was located in the basement below. By this time, his
ultracentrifuge was able to generate a force of 1,000,000g.
page 126

1923 Theodor Svedberg development of the ultracentrifuge


Florence Rena Sabin (1871-1953)
Florence Rena Sabin was one of the first women to build a career as a medical research scientist in the
U.S. She was the first woman on the faculty of the Johns Hopkins University School of Medicine (1902),
and the first woman elected to the U.S. National Academy of Sciences (1925). Later, at the Rockefeller
Institute she applied her interests in the role of white blood cells, especially monocytes, to studies of
host defenses against infectious agents, including viruses.
Statuary Hall, United States Capitol
page 127

1925> Florence Sabin discovery of the role of monocytes and other white blood cells in viral/bacterial infections
In 1892, the Viennese physician Janos von Bkay
suggested a possible relationship between zoster
and varicella after observing that household
exposure to zoster cases could give rise to
varicella in susceptible children. Karl Kundratitz,
also in Vienna, confirmed this connection in
1925 by showing that vesicle fluid from patients
with either varicella or zoster could produce
varicella-like rash in unexposed children and
that these children could in turn transmit
chickenpox to non-inoculated contacts.
Nonetheless, it was not until the advent of
electron microscopy in the early 1940s that a
definite viral etiology was established when the
German physician Helmut Ruska first observed
the virus particles in vesicular fluid. The virus
was finally isolated in 1952 by Thomas Weller.
Using fluorescein-labeled antibodies, he later
resolved the remaining controversy that varicella
and zoster are caused by the same virus.
Abstracted from: Nogueira RG, Traynor B. The
neurology of varicella-zoster virus. A historical
perspective. Arch Neurol. 2004;61:1974-1978.
Helmut Ruska (1908-1973) Thomas Huckle Weller (1915-2008)

Varicella-zoster virus capsids Varicella-zoster virus infection, human skin


negative contrast electron microscopy intranuclear inclusion bodies in infected cells
page 128

1925-1952 Helmut Ruska, Thomas Weller, others discovery of varicella-zoster virus


Henri Valle Otto Waldmann Herman Salomon Frenkel
(1874-1947) (1885-1955) (1891-1968)
Foot-and-mouth disease (FMD) vaccines were among the first to be developed, beginning at the end of the 19th century, by: Henri Valle (France), Otto Waldmann (Germany),
Herman Frenkel (Netherlands) and Paul Capstick (United Kingdom). FMD vaccines began to be produced on an industrial scale from 1950 onwards. The vaccine research was
done in secure government institutes: Insel Reims in Germany (founded in 1909), Pirbright Laboratory in the United Kingdom (1924), Lindholm Island in Denmark (1925),
State Veterinary Research Institute, the Netherlands (1929), Centro Panamericano de Fibre Aftsa (PanAftosa) in Brazil (1951), and Plum Island Animal Disease Center in the
United States (1953).
1925: Valle, the first vaccine, virus obtained from bovine tongue epithelial vesicles, inactivated with formalin.
1930s: Waldmann and Kbe , virus obtained by infecting cattle at an abattoir and collecting epithelium and vesicular fluid, vaccine produced by inactivation with formalin in
the presence of aluminum hydroxide gel.
1940s: Frenkel, large-scale in vitro culture system using freshly cut slices of normal bovine tongue epithelium obtained from an abattoir, the epithelium was infected in vitro
with virus, inactivated with formalin, clarified by centrifugation and filtered.
Today, the question of when and if to use foot-and-mouth disease (FMD) vaccine is incredibly complex and controversial, with policy varying by country. There are questions
whether vaccine supplies would be adequate in a large-scale, multi-country epidemic: for example, it has been stated that the total reserve held by EU-FMD member countries
is about 92 million doses. Further reserves are held in other vaccine banks (e.g., The North American Vaccine Bank). There are also questions whether additional amounts
of vaccine could be produced quickly enough to supplement present supplies, but FMD vaccine production can be ramped up very quickly (currently all FMD vaccines are
produced in BHK-21 cells grown in suspension in large bioreactors). One estimate is that the countries of the EU have the capacity to produce 155 million monovalent doses of
vaccine per year (13 million doses per month). Irrespective of whether FMD vaccine supply would be adequate in a multi-country emergency, because of its use in several parts
of the world where the disease is endemic, it is the most massive vaccine production economy in the world, except for poultry vaccines.
page 129

1925> Henri Valle Otto Waldmann Herman Frenkel, others development of foot-and-mouth disease vaccines
Frederik Cornelis Kraneveld Thomas Michael Doyle Highly pathogenic strains of Newcastle disease virus cause
(1896-1957) (1888-1981) severe, often fatal, disease in chickens, variably marked by
conjunctival reddening and swelling of the tissues of the head
The first recognized outbreaks of [velogenic (highly virulent), peracute] Newcastle disease in chickens occurred as if it were
a new disease, in 1926 in Java, Indonesia (by the work of Frederik Kraneveld) and in 1927 in Newcastle-Upon-Tyne, England
(independently, by the work of Thomas Doyle). This was a remarkable pair of discoveries in 1926-1927, considering the time
and places, the state of background knowledge of the viral diseases of poultry, and especially by the confounding presence
of fowl plague (avian influenza). There are three theories as to the explosive origin of the disease in poultry: (1) via a major
mutation of a low-virulence precursor virus; (2) emergence from a long standing presence of the virus in the village-level (rather
non-commercial) poultry environment of southeast Asia where the economic impact of the disease would not be appreciated;
and (3) emergence via transmission of virus from enzootically affected wild bird species, most likely shore birds (especially
cormorants) and/or psittacine species. Between 1926 and 1946, Newcastle disease appeared in all poultry-producing areas
of the world and became established as a major disease of chickens. Clearly, this was related to the global development of the
commercial poultry industry and its reliance on trade, including international trade. There have been three global pandemics of
velogenic Newcastle disease in the United States, these were in 1942-1946; 1971-1973 (in California alone in the eradication
campaign 12 million birds were destroyed at a cost of $56M); and 2002-2003 (in California and Arizona 3 million birds were
destroyed at a cost of $161M). In recent years, control has been impeded by the continuing presence of virus in unrecognized
backyard poultry flocks, legal and illegal pet bird import facilities and smuggling operations mainly centered on fighting cocks.
After the initial appearance of the virus in the 1920s, it became clear that other less severe diseases were caused by viruses Newcastle disease virus
indistinguishable from Newcastle disease virus [mesogenic (intermediate virulence) and lentogenic (nonvirulent) virus strains]. negative contrast electron microscopy
Kraneveld FC. A poultry disease in the Dutch East Indies. Nederlands-Indische Bladen voor Diergeneeskunde 1926;38:448-450.
Doyle TM. A hitherto unrecorded disease of fowls due to a filter passing virus. J Comp Pathol Therap. 1927;40:144-169.
page 130

1926 Frederik Kraneveld, Thomas Doyle discovery of Newcastle disease virus (the first paramyxovirus)
Journal of Bacteriology 14: 217-258, 1927

and in 1922 Cole invited Rivers to join the Rockefeller Institute faculty and pursue research
in the still poorly understood field of virology. Between 1922 and 1955, Rivers molded the
Institute into the preeminent laboratory for research on viruses, and in over 100 papers
published during these years, Rivers addressed a range of topics relating to some of the most
devastating viral diseases, including smallpox, Rift Valley fever and the viral encephalitides.
He led the way toward delineating virology as a conceptually distinct discipline. In 1937,
Rivers became director of the Rockefeller Institute hospital, a position he held until 1953,
when he became vice president and medical director of the Institute. During World War
Thomas Milton Rivers (1888-1962) II, Rivers served as commander of the Naval Medical Research Unit in the South Pacific,
[Rockefeller Archives, used with permission] where he led programs to combat infectious diseases. He returned to the Rockefeller
Thomas Milton Rivers is considered the father of modern virology and the Institute in 1946 and also served as medical director of the National Foundation for Infantile
father of American virology. He completed an MD at Johns Hopkins School Paralysis he played a major role in the testing of Jonas Salks polio vaccine. Rivers was
of Medicine in 1915. Entering the Army medical corps in 1918, he was assigned recognized widely: elected to the National Academy of Sciences (1932) and to the American
to Fort Sam Houston, San Antonio, Texas, to a project headed by Rufus Cole Philosophical Society (1942); served as president of the American Society for Clinical
of the Rockefeller Institute for Medical Research, investigating outbreaks of Investigation (1932), the American Association of Immunologists (1934), and the Society of
measles and pneumonia. The experience awakened Rivers interest in research, American Bacteriologists (now the American Society of Microbiology)(1936).
page 131

1927 Thomas Rivers publication of the paper that established virology as a distinct field of science
Rivers T. Filterable virusesa critical review. Journal of Bacteriology 1927;14: 217-258.
page 132

1927 Thomas Rivers publication of the paper that established virology as a distinct field of science
Thomas Milton Rivers (1888-1962)
[Rockefeller Archives, used with permission]

Charles E. Simon received an A.B. in 1888 from Johns Hopkins University and an M.D. in
1890 from the University of Maryland. From 1910 until 1920, he served on the faculty of
the University of Maryland. In 1920 he was appointed lecturer in medical zoology at the
Johns Hopkins University School of Hygiene and Public Health. There he started the first
known teaching program on filterable viruses in 1922 this became the first Department
of Filterable Viruses in the United States. He also compiled a large collection of virus
specimens, forming the first virus reference collection in the country. He was later
promoted to Professor of Filterable Viruses. From the inception of the American Journal
of Hygiene in 1921 to his death in 1927 Simon served as its managing editor. In 1928
Charles Edmund Simon Thomas Rivers dedicated his book, the first major virology reference work, to Simon.
(1866-1927)
page 133

1928 Thomas Rivers publication of the first major virology book, Filterable Viruses
Wilbur Augustus Sawyer Thomas Milton Rivers Frank Lappin Horsfall, Jr. Max Theiler
(1879-1951) (1888-1962) (1906-1971) (1899-1972)

Richard Shope
(1901-1966)

Wilbur Sawyer, mouse room,


Rockefeller Foundation
Virus Laboratory, 1934

[images from Rockefeller Archives, used with permission] page 134

1928 founding of the Rockefeller Foundation Virus Laboratory (RFVL) and its international research programs
Fred Griffith (1881-1941)
Fred Griffith was killed in an air-raid in London in WWII
(this poor image is thought to be the best available)

In 1927, Fred Griffith conducted what is now known as Griffiths Experiment. Published in 1928 in the Journal of Hygiene, his paper carries the first widely accepted
demonstration of bacterial transformation, whereby a bacterium can distinctly change its form and phenotype (pathotype). In his experiments, Griffith typed pneumococcus
isolates searching for patterns to clarify the epidemiology of lobar pneumonia. He found that isolates occurred in two general formssmooth and rough. The smooth form
(S) has a polysaccharide capsule, which is associated with virulence (pneumonia in humans, death within days in mice). The rough form (R), lacking a capsule, was considered
avirulent. In Griffiths Experiment, as expected, when S bacteria were killed by heat and then injected into mice, they caused no illness despite being a virulent strain. When
a large amount of heat-killed S was injected along with live R, however, mice developed pneumonia and died. Recovering bacteria from dead mice, Griffith found that the R
bacteria had acquired a capsule to become S, and thereafter maintained this phenotype over many generations. The phenomenon was at the time attributed to an unidentified
transforming factor. Over the next 16 years Oswald Avery, Colin MacLeod, and Maclyn McCarty at the Rockefeller Institute for Medical Research demonstrated that DNA
was the transforming factor, a grand departure from the prevailing belief that the protein content of chromosomes probably made up genes, the stuff of inheritance. Avery and
his colleagues remain famous for this discovery, which was a key in the advance of molecular genetics.
page 135

1928 Fred Griffith discovery of transformation in bacteria, seminal to the development of molecular genetics
Principles / Methods of Viral Disease Diagnosis
Visual information leading to a presumptive diagnosis
Case history, clinical examination, chemistry, hematology
Pathology, histopathology, ultrastructural pathology
Detection of viruses by electron microscopy
Detection and identification of viral antigens
Enzyme immunoassay methods ( e.g., antigen-capture)
Immunochromatography, immunogold binding assays
Immunofluorescence
Immunohistochemistry (immunoperoxidase staining)
Immunoelectron microscopy
Radioimmunoassay
Latex particle agglutination
Immunodiffusion
Direct detection and identification of viral nucleic acids
Hybridization: in situ hybridization, Southern blot
hybridization and dot-blot filter hybridization
Rebecca Lancefield Edwin H. Lennette Polymerase chain reaction (PCR); isothermal amplification
(1895-1981) (1908-2000) Viral genomic sequencing and partial sequencing
Oligonucleotide fingerprinting and restriction
endonuclease mapping
Virus isolation and identification
Virus isolation in cultured cells
Virus isolation in animals and chick embryos
Virus quantitation, especially plaque assay
Detection / quantitation of anti-viral antibodies
(serologic diagnosis)
Enzyme immunoassay (EIA)enzyme linked
immunosorbent assay (ELISA)
IgM class-specific antibody enzyme immunoassay
(EIA/ELISA)
Serum neutralization assay
Immunoblotting (western blotting)
Indirect immunofluorescence assay
Hemagglutination-inhibition assay
Complement-fixation assay
Nathalie J. Schmidt Immunodiffusion
(1928-1986)
page 136

1928> Rebecca Lancefield, Edwin Lennette, Nathalie Schmidt, others founding of viral disease diagnostics
Adrian Stokes (1887-1927) Johannes Bauer (1890-1961) and Wilbur
Sawyer at Rockefeller Institute, 1933
After World War I, the Rockefeller Foundation expanded its
yellow fever activities to Africa. A West African Yellow Fever
Commission was formed in 1925 it was based near Lagos and
was to determine whether African yellow fever was the same
as the South American disease, to find the causative agent (as
if James Carroll had not done this in Havana in 1901) and to
study its epidemiology. Adrian Stokes, an Irish/British professor
Noel Paul Hudson served as field director of the International
of pathology, was a member. In 1927, blood from a 28-year-old
Health Division of the Rockefeller Foundation, in which
African named Asibi, suffering from a relatively mild illness,
capacity he worked with Adrian Stokes and Johannes Bauer
was injected into a rhesus macaque (Macaca mulatta) imported
on yellow fever in Lagos. Later, he served as professor of
from India (African monkeys were resistant). The monkey
bacteriology and hygiene at the University of Chicago,
proved susceptible, establishing the infection for the first time
and then professor and chairman of the Department of
in a suitable laboratory host. This was the first time that Kochs
Bacteriology and dean of the Graduate School at the Ohio
Postulates were proven for a virus. Tragically, very shortly
State University.
afterward Stokes died of yellow fever. Hideyo Noguchi arrived
in Lagos trying to verify his theory that leptospira caused yellow
Stokes A, Bauer JH, Hudson NP. The transmission of yellow
fever he contracted yellow fever and died in 1928. A third
fever to Macaca rhesus [Macaca mulatta]. JAMA 1928;96:
investigator, William Young, who had performed Noguchis
253-254.
autopsy, also died of yellow fever.
Noel Hudson (1895-1987)
page 137

1928 Adrian Stokes, Noel Hudson, Johannes Bauer transmission of yellow fever virus to rhesus macaques
Constant Mathis (1871-1956) Andrew Watson Sellards (1884-1942) Jean Laigret (1893-1966)
At the same time that the Rockefeller Foundation West African Yellow Fever Commission was active, Andrew Watson Sellards, from the
department of tropical medicine at Harvard, was on his way to Dakar, Senegal, to study a yellow fever outbreak when he heard of the success
in infecting rhesus macaques (Macaca mulatta). Taking with him to Senegal some macaques and a strain of stegomyia mosquitoes that
came originally from Havana, he joined General Constant Mathis, director, and Jean Laigret of the Institut Pasteur Dakar, and soon they
reproduced the macaque infection. One day, Laigret noticed that the son of a woman with yellow fever did not appear for his daily visit to the
hospital. He visited the house of this man, Francois Mayali, and found that he had a fever. A sample of Mayalis blood produced severe yellow
fever in a rhesus macaque, though Mayali suffered only a relatively mild case. This strain of yellow fever virus became known as the French
strain. Sellards discovered that the virus survived freezing, allowing transport of infected macaque liver tissue (instead of monkeys) to the
United States for further study. As Sellards returned to the United States, Laigret serially passaged the French strain in mice by intrcerebral
inoculation; this led to neurotropic variants, which after hundreds of passages were used as candidate vaccines, first requiring the use of
immune serum along with vaccine to offset adverse effects due to under-attenuation. With more mouse passages (now up to 237) and other
manipulations they produced a vaccine strain they considered suitable for general use and started vaccinating large populations by 1953, 56
million doses had been used. Some severe vaccine-associated central nervous system infections continued to occur, especially in children, but
considering the risk of yellow fever this was considered acceptable until 1982 when the vaccine was replaced by 17D vaccine.
Mathis C, Sellards AW, Laigret J. Sensibilit du Macaca rhesus au virus de la fivre jaune. C R de LAcademie des Sciences 1928;188:604-606.
Harvey AM. Applying the methods of science to tropical diseases-the story of Andrew Watson Sellards. Johns Hopkins Med J. 1979;144:45-55. Yellow fever virus
negative contrast electron
Monath TP. Yellow fever vaccines: the success of empiricism, pitfalls of application, and transition to molecular vaccinology. In: Plotkin S, microscopy
Fantini B, editors. Vaccinia, vaccination and vaccinology: Jenner, Pasteur and their successors. Paris: Elsevier; 1996. Erskine Palmer, CDC
page 138

1928 Constant Mathis, Andrew Sellards, Jean Laigret transmission of yellow fever virus to rhesus macaques
Feline panleukopenia
in a kitten

Feline panleukopenia is a severe,


highly contagious, multisystemic
disease that is often fatal. Feline
panleukopenia virus is endemic
in cat populations worldwide, but
is rarely seen as a clinical entity
in developed countries today due
to the effectiveness of vaccines.
Young, unvaccinated kittens present
most commonly with this disease.
Unvaccinated feral cat colonies
and other wild felids also serve
as reservoirs of infection for the
domestic cat population.

Jean-Louis-Armand Verge (1892-1964),


with Jean-Marie Camille Gurin (1872-1961) and Gaston
Ramon (1886-1963), at Alfort, 1942
All three of these scientists were faculty members at Lcole Nationale
Vtrinaire DAlfort. Jean-Louis Verge was a distinguished professor, who
served for many years, making important discoveries pertaining to animal
viruses and veterinary vaccines. Camille Gurin worked with Albert Calmette,
discovering the tuberculosis vaccine, BCG (Bacille de Calmette et Gurin).
Gaston Ramon made seminal discoveries in vaccinology, such as the use
of formalin as an inactivating agent and the use of adjuvants that greatly
improved many vaccines for animals and humans. Feline panleukopenia virus
Verge J, Christofferoni J. La gastro-enterite infectieuse des chats est-elle due a model built from X-ray
un virus filgteerable? C R Soc Biol (Paris) 1928;99:312-314. crystallographic data
page 139

1928 Jean-Louis Verge, J. Christofferoni discovery of feline panleukopenia virus (the first parvovirus)
Stefan S. Nicolau Institute of Virology
Stefan S. Nicolau (1896-1967) Bucharest, Romania
Stefan Nicolau started his career as an assistant Constantin Levaditi (1874-1953) at the Institut Pasteur.
Their collaboration was exceptionally fruitful, ranging from viral oncolytic therapy as a novel cancer
therapy, to the discovery, along with I. A. Galloway of the etiologic virus of Borna disease, a neuro-
behavioral disease, studied in sheep, cattle and horses. The name Borna refers to the city of Borna,
Germany, the site of an equine epidemic in 1895-1896 that crippled the Saxon cavalry in a war ongoing at
that time. In the past few years, similar viruses have been found in many animals, especially birds, but their
full natural history is just beginning to be unraveled. In 1990, Janice Clements and colleagues reported that
antibodies to a protein encoded by the Borna disease virus genome are found in the blood of patients with
behavioral disorders. Researchers in Germany, the U.S. and Japan studied 5,000 patients with psychiatric
disorders and 1,000 controls they found a significantly higher percentage of patients than controls were
positive for BDV antibodies. Subsequent studies have also shown an association between Borna disease
virus and human psychiatric disorders. However, as time has gone by, this association has become more
and more controversial.
Nicolau SS, Galloway I. Preliminary note on the experimental study of enzootic encephalo-myelitis (Borna
disease) Br J Exp Pathol. 1927;8:336-341.
Nicolau SS, Galloway I. Borna disease and enzootic encephalomyelitis of sheep and cattle. Spec Rep Ser Ian A. Galloway Borna disease virus
(1900- ) thin section electron microscopy
Med Res Council 1928;121:7-90.
page 140

1929 Stefan Nicolau discovery of Borna disease virus (the first bornavirus)
Robert Green was born in 1895 in Wadena,
Minnesota. He earned four degrees from the
University of Minnesota, including an MD
in 1922. He joined the staff at the University
of Minnesota in 1918 and became professor
of bacteriology and immunology in 1929.
Green was world renowned for his research
on diseases in animals caused by viruses. From
1932-1940, he directed the Minnesota wildlife
disease research program. During his tenure,
he developed a vaccine to prevent encephalitis
in foxes (infectious canine hepatitis virus) and
a vaccine against canine distemper (the first
canine distemper virus vaccine), thereby saving
Minnesotas fur industry.
Green RG, Ziegler NR, Green BB, Dewey
ET. Epizootic fox encephalitis. I. General
description. Am J Hyg. 1930;12:109-129.
Robert Gladding Green (1895-1947)
[from the University of Minnesota Archives, used with permission]

USDA Statement, 1928 Fox Distemper Being Investigated: Frequent requests from fox breeders for assistance in controlling infectious diseases have prompted the USDA
to investigate conditions on a number of fox farms. It was found that a thorough investigation would have to be made of so-called fox distemper to determine its cause. Dr.
Robert. G. Green, Medical School of the University of Minnesota, who with a group of associates, has investigated outbreaks on large farms near Minneapolis for the past three
years. Dr. Newell R. Ziegler, Ms. Beryl Green and Dr. Earle T. Dewey have been associated with Dr. Green since the time he began his investigations. Dr. Green has tentatively
called the disease epizootic fox encephalitis. [The infection in commercially reared foxes is much more neurotropic than in dogs; hence, the common name, fox distemper.
Later, Green and his colleagues found that the virus that they had discovered was indistinguishable from infectious canine hepatitis virus, the important virus of dogs.]
page 141

1930 Robert Green, Newell Ziegler, others discovery of infectious canine hepatitis virus (the first adenovirus)
Infectious canine hepatitis virus, MDCK cell Adenovirus, cell culture, thin section electron microscopy
[stains: anti-NPCP (keratin, epithelial tight junctions) and anti-virus antibody]
page 142

1930 Robert Green, Newell Ziegler discovery of infectious canine hepatitis virus (the first adenovirus)
Leslie Tillotson Webster (1894-1943) The Genealogy of Swiss Mice (Swiss-Webster Mice)
[from Leslie Webster III, used with permission]

Leslie Webster graduated from the School of Medicine at Johns Hopkins University and shortly afterward joined the Rockefeller Institute for Medical Research, where he
remained for the rest of his life. His work aimed to understand epidemiologic phenomena, especially the phenomenon that in a given population individuals differ greatly in
their susceptibility to infection, and that host factors play an important role in this regard. His work involved development of strains of laboratory mice having different levels
of resistance or susceptibility to particular infectious agents. He showed that such traits are inheritable and that strains of mice can be developed that are highly resistant
or exceedingly susceptible. He showed that there is no need for an infective agent of exceptionally high virulence to be present to start an epidemic; it is sufficient that the
number of susceptible hosts constitute a critical proportion of the population. In an epidemic, when the number of susceptibles reaches a certain low level, the infective agent
is not transmitted well enough to continue the infection chain. Individuals surviving an epidemic are resistant, not only through a process of active immunization but also
through their inherited resistance. In the course of his work, especially on rabies and the viral encephalitis viruses, Webster greatly advanced our understanding of viral disease
pathogenesis and at the same time advanced the use of laboratory bred mice (line-bred and inbred mice).
page 143

1930 Leslie Webster, Charles Armstrong, Max Theiler, others development of the mouse for virus research
In 1947, at the Institute of Cancer Research (ICR) at Fox Chase
Center, 100 mice from a stock bred from 6 different sources,
including Websters lines of Swiss mice, were selected for
high production and high growth rate characteristics. They
achieved exceptional production parameters, with an average
Charles J. Armstrong (1886-1967) 15 pups per litter, >99% of pups weaned, <23 day interval
between litters. No sibling mating was used this line was
Charles Armstrong graduated from the School of Medicine at Johns Hopkins University and became a continued for 20 years, over 60 generations. Today, commonly
commissioned officer in the U.S. Public Health Service. In the fall of 1918 he served on a team investigating used outbred / linebred Swiss mouse stocks derive from these
pandemic influenza and soon became a life-long staff member of the Hygienic Laboratory as its name mice, and are identified as to whether they descended through
changed to the National Institutes of Health. As chief of the Division (now Laboratory) of Infectious ICR (ICR Swiss mice) or NIH (NIH Swiss mice) or Websters
Diseases he did seminal work on polio (transmitting the virus to cotton rats and then to mice), lymphocytic colonies (Webster Swiss mice). Most of the common inbred
choriomeningitis (discovery and characterization of the virus), Coxsackie viruses and psitticosis (nearly mice used today also derive from these mice.
dying of a laboratory-acquired infection). At the presentation of the Sedgwick Memorial Medal of the From: Goios A, Pereira L, Bogue M, Macaulay V, Amorim A.
American Public Health Association in 1941, Dr. T. E. Parran, the Surgeon General, said: He is unique in mtDNA phylogeny and evolution of laboratory mouse strains.
that he has made a distinct contribution to our knowledge of every disease with which he has worked. Genome Res. 2007;17:293-298.
page 144

1930 Leslie Webster, Charles Armstrong, Max Theiler, others development of the mouse for virus research
Meyer KF, Haring CM,
Howitt B. The etiology of
epizootic encephalomyelitis
of horses in the San Joaquin
Valley, 1930. Science
1931;74:227-228.

Beatrice Howitt
(1891-1982)

Karl Friedrich Meyer (1884-1974)

The Structure of the Recombinant Alphavirus, Western Equine Encephalitis


Virus, Revealed by Cryoelectron Microscopy. Sherman MB, Weaver SC.
Journal of Virology, published online ahead of print 14 July 2010.

A 3D map of WEEV, with its 2-fold axis oriented perpendicular to screen.


Six trimer spikes are clearly visible. Green layers visible through the holes in
the outer glycoprotein layer represent the outer leaflet of the lipid membrane
Clarence Melvin Haring
separating the glycoprotein shell from the inner nucleocapsid. (1878-1951)
[Dean, School of Veterinary Medicine
University of California, Davis]
page 145

1930 Karl Meyer, Beatrice Howitt, Clarence Haring discovery of western equine encephalitis virus (the first alphavirus)
Karl F. Meyer: Medical Research and Public Health An Oral History.
The Bancroft Library, University of California Berkeley Regional Oral History Office. Interview conducted by Edna Tartaul Daniel in 1961 and 1962.
ENCEPHALOMYELITIS (Abstracted) and sure enough about twenty minutes past nine the shade went up. Within about two minutes
Reports and Suspicions I was over the fence and in another two minutes the strychnine was under the skin of the horse
In July of 1930 a large number of horses were reported dying in the San Joaquin and in another two or three minutes the horse was down, and in another five minutes the head
Valley, particularly in the vicinity of Fresno, from botulism. Well, the moment that word is was off.
mentioned I have to investigate, and I had my theoretical reservations because in summertime It was a heavy head, but I threw it over the fence and wrapped it in burlap and we
you couldnt have botulism, there would not be an adequate amount of moisture in the feed vanished as fast as we could to the most remote corner on the other side of the town of Merced
to permit botulinus to grow and produce its toxin. I sent Dr. Geiger down to a ranch outside where Haring had located an old abandoned chicken coop, and there with the help of flashlights
of Fresno where there were 670 horses. He came back and said, Yes, these horses are partly I did a careful dissection of the brain and wrapped it up so that it was not contaminated, etc.
paralyzed, they are unable to walk or if they walk they walk in circles. This was all done and we were about ready to go home by midnight. We drove back, and I tell
So, I went out in the field. I took my moving picture camera with me and began you, naturally, I was fantastically excited. Haring drove ahead of me, and he had the head of the
to photograph the horses, and when I projected them, after coming back, I realized from the horse without the brain in the trunk of his car and the burlap began to turn loose and it was
motions that this was really the result of inflammation of the brain and not any toxin action, flying just like a flag. Oh, it was a dramatic sight. We got back to the lab about six oclock in
so botulism was out. Now, what was it? That was the question. These cases multiplied at a the morning. I immediately got busy and made a suspension of the brain material. I was over
terrific rate. The late Dr. Clarence H. Haring, who was in charge of the Division of Veterinary in Berkeley by about 9:30 and by ten oclock I had made two inoculations. I inoculated some
Science, convinced us that this was an important thing because there were hundreds of deaths of the suspension directly into the eye of a horse and another part of the suspension I put into
of horses. Dr. Haring and I went to the valley together and standing on street corners just the brain of the horse, which was a tricky techniquenobody ever had done that before, but I
counting the number of legs on trucks as they carted the horses to rendering plants, I roughly had figured it out and I knew that if I could make a trephine hole just above the eye at a triangle
estimated that there were between three and four thousand horses dead from this thing. At point I would get into the brain....
any rate, I had carefully collected at least twelve horse brains; I did that all myself in the field, This was done in the canyon. Just behind the stadium [University of California,
and none took. By that time, it was the latter part of October and the number of cases became Berkeley] was the veterinary science department and there we did this. We said Boo to nobody
less and less, and I was afraid this would begin to disappear with no solution. So, it was agreed because there could have been a lot of Meyer: criticism for bringing in an infection like this. The
with Dr. Haring that we would set up a kind of listening post in the area of Merced where there rest of the brain was prepared by Miss Howitt, who was with me and who was very, very good,
were still some cases. I thought that my failure to isolate this agent out of the brain was perhaps and we had agreed that instead of merely using rabbits we would use mice, we would use guinea
attributable to the fact that I used only the brains from dead horses. I had already known from pigs, and we would even use monkeys and we would put the material directly into the brain.
the twenties that sometimes if there is a virus present in the brain, at the time of autopsy its This gave us the virus. Both horses came down and both of them died from encephalitisthe
gone. virus was in the brainone monkey came down with typical encephalitis manifestations but
Procuring Fresh Tissue recovered.... Here we were really in the midst of a most amazing type of observation. We had
I said that we must get a horse which has the first signs of it. Sure enough, one of the a virus which was inoculable, passed from brain to brain. The next important thing was to try
stooges located a horse outside of Merced, and he went to the rancher and asked if he could buy to repeat it, but then the cases had disappeared. It had become cold. So it was most important,
it and this fellow said, I wont sell the horse, and if you ever do anything to the horse I shoot during the winter months, to continue the passages, to keep it going. There were no difficulties
you. This was the news which came to me over the telephone, and I said, All right, let me try at all, it was there.
my hand in this game. I went down and I had a $20 bill in my pocket. This was a depression Then I began to plot on maps where the cases had occurred and what relationship
year and I was sure they would be glad to get rid of the horse for $20. When I walked into the there was to the environment. In the course of doing this plotting I had received reports
hotel in Merced there was a gloomy crowd sitting around. You shouldnt dare to do anything that in Kern County Hospital there had been fantastic increases in cases of polio, all from
out there. Youre going to get shot. I said, Im not going to talk to him. Im going to talk to his rural areas. I went down and saw some of these cases and said, This is not polio, this is
wife. So I went out. encephalitis. Then cases of human deaths occurred, and I had told every pathologist in valley
I talked to her and I said, look here, this horse is going to die anyhow, and when its areas, If you do an autopsy, let me have the brain. Well, what these boobs did, instead of giving
dead you havent anything. It just goes to the rendering plant and you get a couple of dollars. the brain in the frozen state, they pickled it in formalin so naturally you couldnt do anything
On the other hand, you see, you could contribute to the knowledge of what this is and perhaps but get a section and the sections again showed typical encephalitis. [Beatrice Howitt eventually
to its prevention. Well, she said, My husband is just irate about this. I said, Yes, I can readily made an isolate from a fatal human case.]
understand, but look here, suppose I trust you, and I give you $20 and the next morning you Then, by looking at the map I began to see one crazy thing, that most of the cases
will find in the backyard the horse without a head? How are you going to do this? Look here, were in an irrigated area. The moment you went in the foothills, no cases. I said, Hmm, this
about nine oclock at night when it is dark, Ill be over here behind some bushes on the corner looks very much as if this might be transmitted by an insect; this might perhaps be mosquito-
of the street, and I can see the window of your house. When your husband is sound asleep you borne. But in 31, nobody had any techniques, nobody knew much how to do these things, but
lift up the shade. I was at the corner of the street absolutely prepared for everything. I had a it crystallized more and more that this was a mosquito-borne affair.
syringe with strychnine, I had a good sharp knife, and I sat around there and smoked a pipe,
page 146

1930 Karl Meyer, Beatrice Howitt, Clarence Haring western equine encephalitis virus (the first alphavirus)
Shope RE. Swine influenza.
Filtration experiments
and etiology. J Exp Med.
1931;54:373-385.

Swine influenza virus infection in pigs


is usually seen as a mild respiratory
disease, rather like that seen in humans
with seasonal influenza. Histopathology:
Richard Edwin Shope (1901-1966) mild bronchiolitis and alveolitis with
mononuclear inflammatory infiltration.
[from Rockefeller Archives, used with permission]

Pig farmers in Iowa had reported two outbreaks, one in 1918 and another in 1929, of a highly contagious, influenza-like disease. The disease bore such a remarkable
resemblance to human flu that it was named swine influenza. In 1930, following a hunch, Richard Shope and his mentor Paul Lewis took mucus and lung samples from
infected pigs and attempted to isolate the etiologic agent. They isolated a bacterium that was quite similar to Hemophilus influenzae (then called Pfeiffers bacillus), which many
investigators still thought to be the cause of human influenza. They called the pig bacterium Bacillus influenzae suis, but when they injected it into pigs, it caused no disease.
Shope then ultrafiltered the samples and found that the filtrate contained the infectious agent. Shopes filtrate caused a highly contagious, influenza-like disease in pigsalbeit
a milder disease than seen in naturally infected animals. Mixing the filtrate with the bacterium reproduced the severe disease. He concluded that the filterable agent caused the
infection, which then facilitated secondary infection with the bacterium. Using Shopes technique, Wilson Smith, Christopher Andrewes and Patrick Laidlaw, of the National
Institute for Medical Research in London, soon developed the ferret model of human influenza. Both Shope and the British trio later demonstrated that sera from humans that
were infected with the 1918 influenza virus neutralized the swine virus, leading them to conclude that the swine virus was a surviving form of the 1918 human pandemic virus.
page 147

1930 Richard Shope discovery of swine influenza virus


William Elfords Filtration System

Approximate Sizes of Certain Viruses as Determined by


Filtration Through Graded Collodion Membranes, 1934
(Table Prepared by Dr. J. H. Bauer of the International
Health Division of the Rockefeller Foundation)
William Joseph Elford Christopher Howard Andrewes
(1900-1952) (1896-1988)
The technology and equipment for producing, using and testing collodian membranes to estimate the size
of virus particles advanced very quickly, with several key papers published in the early 1930s that allowed
investigators in many laboratories to replicate Elfords work. One of the most remarkable things about the
early use of collodion membranes was its reproducability and its accuracy most of the virus particle size
estimates made using the method were remarkably close to measurements made many years later by more
sophisticated methods.
Elford WJ. A new series of graded collodion membranes suitable for general bacteriological use, especially in
filterable virus studies. J Pathol Bacteriol. 1931;34:505-521.
Elford WJ, Andrewes CH. Filtration of vaccinia virus through graded collodion membranes. Brit Jour Exp
Path. 1932;13:13-36.
Elford WJ, Andrewes CH. The sizes of different bacteriophages. Brit Jour Exp Path. 1932;13:446-456.
Bauer JH, Hughes TP. The preparation of the graded collodion membranes of Elford and their use in the study
of filterable viruses. Jour Gen Physiol. 1934;18:143-162.
page 148

1931 William Elford, Christopher Andrewes use of graded collodion membranes to determine virion size
In 1931, when Alice Miles Woodruff joined the
Department of Pathology at Vanderbilt University,
Ernest Goodpasture asked her to try to grow fowlpox
virus in tissue cultures made from chick kidney
cells this did not work. Well, lets try the chick
embryo itself, Goodpasture suggested, its made up
of living cells, and there are no bacteria within it.
Woodruff, using fertilized chicken eggs, cut a flap
in the shell with a tiny blade, injected fowlpox virus
into the embryo, sealed the window with a thin piece
of glass held in place with Vaseline, and replaced the
egg in an incubator. But this did not work because,
without exception, bacteria gained entrance and killed
the embryo. It was agreed that bacteria were being
transferred to the embryo together with the virus.
Henceforth all fowlpox samples were tested for an
absence of bacteria before transfers were made. Still,
the embryos died from bacterial infection. Every time
she and Goodpasture thought about abandoning the
experiments someone would say, Lets give it just one
more try. If we can only manage to get the virus to
grow in one embryo we can use that as a pure source
for all other experiments.
As luck would have it, Alice Woodruff discovered an
inoculated egg in which the embryo was thriving; it
also had a slightly swollen claw where the virus had Ernest W. Goodpasture
been placed. When the swollen tissue was transferred (1886-1960)
to another embryo it produced more swollen tissues.
Soon she had a number of embryos that contained
pure virus. Under the microscope the swollen tissues
showed inclusion bodies. To make sure that this was
the same virus she started out with, she took some of
the swollen tissue from an embryo and injected it into
a grown chick; it developed typical fowlpox.
Based on this work, Max Theiler of the Rockefeller
Institute, in 1937, grew yellow fever virus in eggs and
Alice Miles Woodruff (1900-1985) passed it through hundreds of mice and chicks until
and Ernest Goodpasture he had a mutant that could be used in a vaccine
[from Vanderbilt University Archives, used with permission] 17D vaccine. Theilers work led to several other viral
vaccines as well.
From: Podolsky ML. Cures out of chaos. How Embryonating hens egg
unexpected discoveries led to breakthroughs in from work of James Bumgardner Murphy
medicine and health. Chur, Switzerland, Harwood
and Francis Peyton Rous, 1930s
page 149
Publishers; 1997.

1931 Alice Woodruff, Ernest Goodpasture development of embryonating hens eggs as host for viruses
Ernest W. Goodpasture (1886-1960) Herald Cox (1907-1986)
and Frank Macfarlane Burnet (1899-1985)
By 1932, Alice Miles Woodruff and Ernest Goodpasture used embryonating hens eggs for the isolation of herpes simplex virus
and shortly thereafter variola virus (grown on the choriallantoic membrane, CAM). By 1940, influenza A & B viruses were being
grown in the amniotic and allantoic cavities of embryonating eggs, and by the end of World War II when antibiotics (penicillin
and streptomycin) became available, embryonating eggs were a very important part of laboratory diagnostics and research. W. I.
B. Beveridge and F. M. Burnet published a book in 1946, The Cultivation of Viruses and Rickettsiae in the Chick Embryo [Medical
Research Council, Special Report 256, London], in which 37 viruses were described as cultivable in eggs, including the viruses
of smallpox, yellow fever, rabies, St. Louis and Japanese encephalitis, mumps and rinderpest. The usefulness of the method for
preparing vaccines was also emphasized. The embryonating egg reached its peak in the laboratory by the 1970s since then,
except for its use in influenza research and vaccine manufacture, it has
largely been replaced by cell culture methods. Myxoma virus, chorioallantoic membrane,
embryonating hens egg
page 150

1931> Frank Macfarlane Burnet, Herald Cox development of embryonating hens eggs as host for viruses
Robert Daubney (1891-1977) John Richard Hudson (1904-1990) Percy Cyril Claude Garnham (1901-1994)
The first recorded outbreaks of Rift Valley fever occurred in Kenya in 1910 and 1913 in imported sheep; there were very large
numbers of abortions and deaths in newborn lambs and parturient dams. In 1930, Robert Daubney and John Richard Hudson
investigated a very large outbreak and carried out a classic study, isolating Rift Valley fever virus and describing the pathology
of the disease and much about its natural history. Subsequently, as imported animal numbers increased, further outbreaks
were reported in South Africa, sub-Saharan Central Africa and North Africa. The transmission cycle of Rift Valley fever virus
in eastern Africa involves virus survival in floodwater Aedes spp. mosquito eggs infected transovarially. These eggs survive
long periods of drought in dry water holes (called dambos) in fact, these eggs require a period of dehydration before they
will hatch. When dambos flood after heavy rainfall, the eggs hatch and the adults infect nearby domestic and wild animals. The
same flooding later leads to a population explosion of Culex spp. mosquitoes, which become infected by feeding on the viremic
animals these mosquitoes disperse widely, leading to extensive dissemination of virus and epidemics which often involve
humans as well as sheep and cattle. Since epidemics in East Africa are closely associated with the heavy rainfall that occurs
during the warm phase of the El Nio/Southern Oscillation, satellite-based mapping of excess surface water has been used
successfully to predict epidemics and guide intervention using livestock vaccine. In 1977, a large outbreak spread from Sudan
to Egypt along the Nile affecting 25%-50% of all sheep and cattle. The outbreak involved hundreds of thousands of human
infections with 18,000 officially confirmed clinical cases and 600 deaths (actual numbers were considered to be much higher).
There have been repeated epidemics in many different localities in Africa ever since. Following the opening of the Diama Dam
in 1987, an outbreak occurred in Mauritania. The relation between major irrigation projects and outbreaks clearly is due to an
increase in mosquito breeding habitat. Because of the importance of the livestock and human diseases, there is much ongoing
effort to develop / improve Rift Valley fever virus vaccines.
page 151

1931 Robert Daubney, J. Richard Hudson, Percy Garnham discovery of Rift Valley fever virus
Rift Valley fever virus, liver, mouse, thin section electron microscopy
[from an unpublished study by the author and Joel McKeith Dalrymple (1939-1992)]
Daubney R, Hudson JR, Garnham PC. Epizootic hepatitis or Rift Valley fever: an undescribed
virus disease of sheep, cattle and man from East Africa. J Pathol Bacteriol. 1931;34:545-579.
This classic paper influenced viral disease pathology / pathogenesis / natural history studies for
many years. For example, here is a snippet of text from the paper: Histologically the liver lesion
is in every way as diagnostic as the naked eye appearance of the organ. One might go further
and state that whenever the macroscopic changes are insufficiently pronounced to enable a
diagnosis to be made, which is not infrequently the case in adult sheep, histological examination
will enable one to establish the diagnosis beyond any possibility of error. Although the general
picture may vary with the susceptibility of the subject and with the degree of rapidity with
Rift Valley fever, liver, human, from an epidemic in eastern Africa in which a fatal termination or recovery is reached, the nature of the process remains essentially
2006-2007 studied by Sherif Zaki of the Centers for Disease Control and the same in all cases. It may be characterised briefly as a focal degeneration of liver cells
Prevention. Immunohistochemistry, paraffin section. The most distinctive closely followed by infiltration of the lesion with phagocytic cells, chiefly polymorphonuclear
histopathologic change in liver tissue from fatal cases is extensive focal leucocytes and histiocytes. The general sequence of these changes can most easily be followed
hepatocellular necrosis without prominent inflammatory cell infiltration. in adult sheep, since in lambs the whole process is so rapid and the destruction of tissue is so
[from Sherif Zaki, used with permission] enormous as to obscure completely the actual nature of the lesion. In hyperacute cases in young
lambs a section of the organ may be unrecognisable as liver. page 152

1931 Robert Daubney, J. Richard Hudson, Percy Garnham discovery of Rift Valley fever virus
Mammary adenocarcinoma, mouse mammary
tumor virus, prominent acinar architecture, H&E
Clarence Cook Little (1881-1971) John Joseph Bittner (1904-1961)
Mouse mammary tumor virus (MMTV) is a milk transmitted retrovirus. It was formerly known as Bittner
virus, and before that as the milk factor, referring to the extra-chromosomal vertical transmission of
murine mammary cancer by adoptive nursing, as demonstrated in 1936 by John Bittner, while working at the
Jackson Laboratory in Bar Harbor, Maine. Bittner, a geneticist and cancer biologist, developed the idea that
a cancerous agent, a milk factor, a virus in milk, could be transmitted from mothers to their young. Several
mouse strains carry the virus endogenously, but in other mouse strains the virus is only transmitted via milk.
In the former, the virus is carried as a DNA provirus integrated in the DNA of lymphocytes in the mammary
glands. The virus is transported through the gastrointestinal tract of pups to Peyers patches where it infects
the new hosts macrophages, and then lymphocytes. In recent years, MMTV-like DNA sequences have been
found in human breast cancer tissue and most recently the virus has been shown to be able to productively
infect human cells, possibly suggesting that an MMTV like virus may play a role in human breast cancer
but this is quite controversial and largely unconfirmed.
Staff, Jackson Memorial Laboratory. The existence of non-chromosomal influence in the incidence of
mammary tumors in mice Science 1933;78:465-466.
Bittner JJ. Some possible effects of nursing on mammary gland tumor incidence in mice Science 1935;84:162.
page 153

1932 John Bittner, Clarence Little Jackson Memorial Laboratory discovery of mouse mammary tumor virus
Albert Bruce Sabin (1906-1993) Frederick Parker Gay (1874-1939)
[from the U.S. National Academy of Sciences, used with permission]
The first documented case of human B virus infection occurred in 1932 when a researcher (patient W.B.) was bitten on the hand by an apparently healthy rhesus macaque
(Macaca mulatta) and died of progressive encephalomyelitis 15 days later. Two research groups obtained samples from patient W.B.: Frederick Gay and Margaret Holden
(University of California, later Columbia University) and Albert Sabin and Arthur Wright (Cincinnati Childrens Hospital). Both groups demonstrated a similar disease
progression in rabbits inoculated with nerve tissue from patient W.B. and characterized the agent as a herpesvirus. Neither group was able to produce disease in rhesus
macaques, presumably because the monkeys were already immune. Sabins group named the virus B virus (after patient W.B.). Formal and vernacular nomenclature have left
several synonymous names for this virus: Macacine herpesvirus 1, Cercopithecine herpesvirus 1, herpesvirus simiae, as well as B virus.
Gay FP, Holden M. The herpes encephalitis problem. J Infect Dis. 1933;53:287-303.
Sabin AB, Wright WM. Acute ascending myelitis following a monkey bite, with the isolation of a virus capable of reproducing the disease. J Exp Med. 1934;59:115-136.
page 154

1932 Frederick Gay, Albert Sabin, Margaret Holden, Arthur Wright discovery of B virus
Wilson Smith Christopher Howard Andrewes Patrick Playfair Laidlaw
(1897-1965) (1896-1988) (1881-1940)
Even though influenza was shown during the great pandemic of 1918-1919 to be caused by an ultrafilterable virus, by Charles Nicolle and quite a few others, and this was taken
as common knowledge by Thomas Rivers in his seminal book, Filterable Viruses, in 1928, there seemed to be lingering uncertainty. When in 1931, Richard Shope discovered
swine influenza virus and concluded that the disease in swine required the interaction of the virus and a bacterium, now called Haemophilus parasuis, the wish for further
proof of the etiology of the human disease became more vocal. It was Wilson Smith, Christopher Andrewes and Patrick Laidlaw at the National Institute of Medical Research
(NIMR) Mill Hill who settled the matter, irrefutably. Through the 1920s, Laidlaw had worked on canine distemper, culminating in discovery of the viral etiologic agent and
development of the first distemper vaccine. The key to this work was a ferret experimental animal model. When a severe influenza epidemic erupted in 1929, Laidlaw recruited
Smith and Andrewes and they infused ultrafiltered throat washings from eight influenza patients into rats, mice, guinea pigs, rabbits, monkeys, pigs, and horses. These efforts
failed. Curiously, ferrets, Laidlaws favorite model, were not among the first animals tested. However, soon after, following upon reports of an outbreak of a flu-like disease
among ferrets at Wellcome Laboratories, where the animals were being used to manufacture canine distemper vaccine, Smith instilled intranasally into two ferrets ultrafiltered
nasal washings taken from Andrewes, who had himself caught the flu. Within 48 hours the animals started sneezing and displaying signs of flu-like disease. Washings from
seven other patients also induced the disease. However, the etiologic agent was lost when canine distemper broke-out among the ferrets. By a twist of fate, shortly afterward
Smith developed influenza and Andrewes used his throat washing to infect a new batch of ferrets, now maintained in an isolation facility. The virus isolated from these ferrets
became the first influenza virus isolate (designated WS after Smith), the one used for validation of ultrafilterability, serial passage in ferrets, neutralization by convalescent
ferret serum, and transfer to mice. Most importantly, this work was seminal in shifting virological research from the perspective of its bacteriological roots to that of
experimental pathology the NIMR researchers identified themselves as experimental pathologists. This was the perspective of virology until the rise of molecular biology.
Smith W, Andrewes C, Laidlaw P. A virus obtained from influenza patients. Lancet 1933;2:66-68.
Bresalier M. Uses of a Pandemic: Forging the Identities of Influenza and Virus Research in Interwar Britain. Social Hist Med. 2011;25;400-424.
page 155

1933 Wilson Smith, Christopher Andrewes, Patrick Laidlaw ferret model confirming the viral etiology of influenza
Richard Edwin Shope Francis Peyton Rous Joseph Willis Beard
(1901-1966) (1879-1970) (1906-1983)
In 1933, Richard Shope learned from hunters in Kansas and Iowa of tumors around the head and shoulders of cottontail rabbits
(Sylvilagus spp.), appearing as large horny protruberances sometimes resembling antlers. The antlers were shown to be
epidermal papillomas, which because cornified cells continued to form but did not slough off, grew to appear as tough horns
[thought to be the origin of the mythical jackalope the jackrabbit with antelope horns or deer antlers]. Shope and E. Weston
Hurst showed that soluble extracts from the abnormal epdermal tissues from wild cottontail rabbits contained an ultrafilterable
(Berkefeld V filter) agent, the Shope papillomavirus (now cottontail rabbit papillomavirus), which can be transmitted and cause
disease in wild cottontail and domestic rabbits (Oryctolagus cuniculus). As covered later in this book, in 1935, Peyton Rous and
Joseph Beard showed the tumorigenic potential of the Shope virus, thereby recentering much of virus-cancer research for many
years. Meanwhile, other papillomaviruses were discovered and later became important research subjects: canine papillomavirus
was discovered by John McFadyean and Frederick Hobday in 1898; the first human papillomavirus was discovered by Giuseppe
Ciuffo in 1907; and, importantly because of its capacity to grow in cell culture, bovine papillomavirus was discovered by O.
Magelhaes in 1920.
Shope RE, Hurst EW. Infectious Papillomatosis of rabbits: with a note on the histopathology. J Exp Med. 1933;58:607-624.
McFadyan J, Hobday F. Note on the experimental transmission of warts in the dog. J Comp Pathol Ther. 1898;11:341-344. Horned hare by Benard. Summa
Dubiorum Circa Classes Quadrupedum
Magalhaes O. Verruga dos bovideos. Brasil-Medico 1920;34:430-431. et Amphibiorum, 1743 page 156

1933 Richard Shope, Francis Peyton Rous, Joseph Beard discovery of rabbit (Shope) papillomavirus
Max Knoll (1897-1969), Ernst Ruska and their first electron microscope
One half of the 1986 Nobel Prize in Physics was awarded to
Ernst Ruska for his fundamental work in electron optics and
for the design of the first electron microscope, one of the most
important inventions of this century. Its development began at
the end of the 1920s with work carried out by Ruska as a young
student at the Berlin Technical University, under the supervison
of Max Knoll. Ruska found that a magnetic coil could act as a
lens to focus electrons, and that such electron lenses could be
used to obtain an image of an object irradiated with electrons. By
Ernst Ruska (1906-1988) coupling two electron lenses he produced a primitive microscope.
He quickly improved on this and in 1933 built the first electron
Ruska E, Knoll M. Die magnetische sammelspule fr schnelle microscope with a performance clearly superior to that of the
elektronenstrahlen. (The magnetic concentrating coil for fast electron light microscope. After completing his PhD in 1933, Ruska
beams.) Zeit Physik 1931;12:389-400 und 448. joined Siemens, where in 1939 he led the development of the
first commercially-produced electron microscope. He also set up
Knoll M, Ruska E. Das elektronenmikroscop. Zeit Physik a laboratory for visiting researchers, which was initially headed
1932;78:318-329. by his brother Helmut, a physician who took the first electron
page 157 micrograph of a virus.

1933 Ernst Ruska, Max Knoll invention of the transmission electron microscope
The first transmission electron
microscope, Ernst Ruska and
Max Knoll (1933)
(replica) The first commercially built transmission electron microscope
the Siemens bermikroskop M 100 (1939)

The first commercially produced electron microscope was produced by Siemens in 1939, as described by Ernst Ruska: As first collaborators [Max Knoll and I] started in
1937, and in 1938 we had completed two prototypes with condenser and polepieces for objective and projective lenses as well as airlocks for specimens and photoplates. The
maximum magnification was 30,000x. One of these instruments was immediately used for the first biological investigations by [my brother] Helmut Ruska and several medical
collaborators. By the end of 1939 the first serially produced Siemens instrument had been delivered to Hoechst [Hoechst AG, now Sanofi-Aventis]. By February 1945 more than
30 electron microscopes [bermikroskop] had been built in Berlin and delivered. By the late 1930s electron microscopes with theoretical resolving power of 10nm were being
produced. By 1946, resolving power had reached ~1.0nm, by 1970, 0.6nm, and by 1990, 0.1-0.2nm. [The theoretical resolving power of a an optical light microscope is 200 nm.]

page 158

1933 Ernst Ruska, Max Knoll invention of the transmission electron microscope
Ralph S. Muckenfuss (1899-1979) St. Louis encephalitis virus, Culex pipiens salivary gland
The Metropolitan Health Council of St. Louis appointed Ralph Muckenfuss, Assistant
Professor of Medicine at Washington University School of Medicine, chairman of the
committee tasked with finding the cause of a major encephalitis epidemic still ongoing
in the area. [Muckenfuss later became the first director of The Public Health Research
Institute of the City of New York and director of the laboratories of the New York City
Department of Health.]
page 159

1933 Ralph Muckenfuss, Leslie Webster, Charles Armstrong, others discovery of St. Louis encephalitis virus
Leslie Tillotson Webster (1894-1943) Charles J. Armstrong (1886-1967)
[from Leslie Webster III, used with permission]

Muckenfuss RS, Armstrong C, McCordock HA. Encephalitis: Studies on experimental transmission. Public Health Reports 1933;48:1341-1343. [virus isolation via intracerebral
inoculation of rhesus macaques]
Webster LT, Fite GL. A virus encountered in the study of material from cases of encephalitis in the St. Louis and Kansas City epidemics of 1933. Science 1933;78:463-465. [virus
isolation via intracerebral inoculation of Webster-Swiss mice]
In 1933, in five weeks in autumn an encephalitis epidemic of explosive proportions broke out in the vicinity of St. Louis, Missouri and the neighboring St. Louis County.
Over 1,300 cases were reported to the local health departments and the newly constituted National Institute of Health was appealed to for epidemiological and investigative
expertise. Time Magazine, 1937: The only scientific fact known about this disease is that it is caused by a specific virus. This was ascertained during the 1933 epidemic by one
of the most vigorous and concentrated attacks on a disease ever made by Medicine. Immediate discoverers of that virus were Dr. Howard McCordock, Washington University
School of Medicine, St. Louis; Dr. Charles Armstrong, virus expert of the U. S. Public Health Service; Dr. Leslie Webster of the Rockefeller Institute; and Dr. Ralph Muckenfuss,
then of Barnes Hospital, Washington University School of medicine, St. Louis, now director of NYCs Bureau of Laboratories. page 160

1933 Ralph Muckenfuss, Leslie Webster, Charles Armstrong, others discovery of St. Louis encephalitis virus
Roberto Franco Jerome Austin Kerr Lus Patio Camargo Jorge Boshell-Manrique Fred L. Soper
(1874-1958) (1925-1974 (1891-1978) (1903-1976) (1893-1977)
The control of the urban mosquito vector of yellow fever, Aedes aegypti, in the major port cities of the western hemisphere in the
early decades of the twentieth century led to the notion that the disease could be eradicated. However, unexplained outbreaks in
inland rural forested areas continued after urban centers had been freed of Aedes aegypti. Roberto Franco (1874-1958) first studied
this in Colombia and coined the term yellow fever of the forests. In 1910-1911 he reported in an obscure journal that yellow fever
was transmitted by mosquitoes that bit during the day while men were working in the forest. From 1926 to 1932, Jerome Austin Kerr
(1925-1974)(Rockefeller Foundation staff member) and Lus Patio Camargo (1891-1978)(Rockefeller Foundation employee) found a
high level of yellow fever seropositivity in a forested region where there was no Aedes aegypti, but were unable to identify the vector.
Fred L. Soper (1893-1977), head of the Brazilian office of the International Health Division of the Rockefeller Foundation, prevented
the publication of the Colombian work, due to the dangerous nature of the findings, but then sent his own article to the American
Journal of Hygiene, intending to establish priority for the discovery of jungle yellow fever. For many years, Soper did indeed hold this
legacy, but now we understand the proper shared legacy. Soper went on to become the main architect of the Rockefeller Foundations
Victory Over Yellow Fever campaign in Brazil. He directed the program toward the nearly complete eradication of the urban
mosquito, Aedes aegypti, from Brazil and other Latin American countries. Soper later also led the Rockefeller Foundations yellow
fever vaccination campaign in the region. In 1936, Jorge Boshell Manrique (1903-1976)(also a Rockefeller Foundation employee)
confirmed the role of arboreal Haemagogus spp. mosquitoes and the endemic forest canopy / monkey
cycle of the virus. Haemagogus spp. mosquitoes live at the top of the jungle canopy, but are brought to
ground level when trees are felled, thereby transmitting virus to forest workers. In Africa, several species
Studies of arboviruses, such as yellow fever
of Aedes mosquitoes were later shown to maintain a cycle of jungle yellow fever and carry virus to the
virus, were carried out for many years at the
Instituto Evandro Chagas, Belem, Brazil, edges of urban habitats, where again Aedes aegypti (and other urban Aedes species) cause epidemics.
supported by the Rockefeller Foundation. Quevedo E, Manosalva C, Tafur M, Bedoya J, Matiz G, Morales E. Knowledge and power: the
More than 50 different viruses were found,
many from tree platforms different
asymmetry of interests of Colombian and Rockefeller doctors in the construction of the concept of
mosquito species harboring different jungle yellow fever, 1907-1938. Can Bull Med Hist. 2008;25:71-109. (History of Medicine Center,
viruses were found at particular levels in Universidad Nacional de Colombia)
the canopy, Haemagogus spp. at the top of
the canopy. Soper FL, Penna HA, Cardoso E, Serafim J, Frobisher, M, Pinheiro J. Yellow fever without Aedes aegypti:
study of rural epidemic in Valle do Chanaan, Espirito Santo, Brazil. Am J Hyg. 1932;18:555-587. Hemagogus janthinomys
page 161

1910-1933 R Franco, JA Kerr, L Patio Camargo, F Soper, J Boshell Manrique discovery of jungle yellow fever cycle
William W. Dimock Elvis Roger Doll
(1880-1953) (1912-1967)
There are now 9 equine/equid herpesviruses:
Equine herpesvirus 1 exists in horse populations
worldwide and is associated with acute respiratory
disease. It is also responsible for abortion in mares and
neurological disease, though the latter is rare.
Equine herpesvirus 2 is of unclear clinical significance,
but it is consistently found in the respiratory mucosa,
conjunctiva and leukocytes of clinically normal horses
and has been associated with keratoconjunctivitis, upper
respiratory tract disease, generalized malaise and fever,
pharyngitis and lymphadenitis.
Equine herpesvirus 3, also called equine coital
exanthema virus, is the cause of a sexually-transmitted Equine fetus, lung, equine herpesvirus 1. (top) Necrotizing
genital vesicular disease of some importance, especially in bronchiolitis with intranuclear eosinophilic inclusions and
breeding mares and stallions. chromatin margination (H&E) (bottom) Same, with virus antigen
detected using a monoclonal antibody recognizing a specific
Equine herpesvirus 4, like EHV-1 causes acute
viral structural glycoprotein. (Immunohistochemistry
respiratory disease in horses worldwide and is rarely the
and hematoxylin)
cause of neurological sequelae in infected horses.

Michael J. Studdert The other viruses mostly infect other equid species.
page 162

1933> William Dimmock, Elvis Doll equine abortion virus and equine rhinopneumonitis virus (herpesviruses)
The Etiology of Mumps.
C.D. Johnson and E.W. Goodpasture, 1934:
We have recently reported the production of a non-
suppurative parotitis in the rhesus monkey (Macaca mulatta)
by the inoculation of the glands, through Stensons duct, with
saliva collected from early cases of epidemic parotitis. The
etiological agent of the experimental parotitis was found to be
a filterable, cytotropic virus which is resistant to freezing,
drying and glycerination. This virus was not found in normal
saliva nor did it correspond to any known virus with which we
were familiar.
The difficulty of proving the etiological relationship of a virus
to a particular disease is apparent when one realizes that the
two means of establishing microbial causation, heretofore so
successfully applied to infectious diseases, namely cultivation
and microscopic morphology, are completely lacking in the
study of viruses. Therefore, the only means one has to rely
on are the specific inhibition of a particular virus by immune
serum from an animal or man recently recovered from an
attack of the specific infection, and the reproduction of the
specific disease in the natural host by inoculation with the
virus under investigation.

Mumps N protein (turquoise) in


the cytoplasm of a cultured cell.
Immunofluorescence

Ernest W. Goodpasture (1886-1960)


Mumps virus, with prominent
Johnson CD, Goodpasture EW. An investigation of the etiology of nucleocapsids in budding virions
mumps. J Exp Med. 1934;59:1-19. thin section electron microscopy
Johnson CD, Goodpasture EW. Experimental immunity to the virus
of mumps in monkeys. Am J Hyg. 1936;23:329-339.
Claud D. Johnson (1910-1976)
Johnson CD, Goodpasture EW. The histopathology of experimental Vanderbilt University School of Medicine,
mumps in Macacus rhesus. Am J Pathol. 1936;12:495-510. Class of 1932
[from Vanderbilt Archives, with permission]
page 163

1934 Claud Johnson, Ernest Goodpasture discovery of mumps virus


Beckman Acidimeter (1934)
with lemons

Model G pH Meter (1935)


Arnold O. Beckman (1900-2004)
The California citrus industry needed an accurate gauge of the level of acidity of fruit and derived products. At the time, early in the 1930s, the most common method for
determining acidity was litmus paper (litmus is a soluble powder derived from lichens). When the paper turned color, color charts helped the examiner determine the acidity
of the solution. Chemists also used electrochemical methods to measure the concentration of hydrogen ions, but these did not work well on citrus products. When approached
about the problem, Arnold Beckman recommended using a vacuum tube voltmeter to amplify weak electrical signals from a glass electrode submerged in the sample to be
tested. In 1934, Beckman decided he would build the instrument himself it worked so well that he was asked to make another one just like it. He soon concluded that all
the parts should be put together in an integrated instrument, initially called an acidimeter. Beckman not only figured out how to measure pH accurately, he revolutionized
instrumentation by building the first chemical instrument that consisted of one compact unit that was portable. This simplified research as the user no longer had to assemble
various components an instrument could just be purchased. [The pH (hydrogen potential) scale, ranging from 0 to 14, was developed in 1909 by Sren Srenson, a Danish
biochemist.]
page 164

1934 Arnold Beckman invention of the electronic pH meter


Malcolm H. Merrill (1903-1987) Charles Lacaillade, Jr. (1904-1978) Carl Ten Broeck (1885-1966)

Eastern equine encephalitis virus, Aedes triseriatis mosquito salivary gland, thin section electron microscopy
page 165 [Ten Broeck photo from Rockefeller Archives; Lacaillade photo from St. Johns University Archives, used with permission]

1934 Malcolm Merrill, Charles Lacaillade, Carl Ten Broeck discovery of eastern equine encephalitis virus
Japanese encephalitis (JE) is the leading cause of viral encephalitis in
Asia, with an estimated annual incidence of 45,000 human cases and
10,000 deaths. However, JE cases are under-reported the true annual
incidence of encephalitis cases is estimated to be closer to 175,000.
Case-fatality rates (hospitalized) range from ~25-40%, with serious
neurologic sequelae occurring in ~45-70% of survivors depending
on the population and age of infected individuals. Outbreaks of JE in
northern India and Nepal between 2005 and 2007 have resulted in at
least 11,000 cases and over 2,000 deaths, highlighting the continued
burden of disease in developing countries and the continuing westward
spread of the virus.
Seminal experiments conducted near Tokyo in the 1950s elucidated
the transmission cycle of JE virus, with pigs and wild birds identified
as amplifying hosts and Culex tritaeniorhynchus incriminated as the
primary vector mosquito species. In Japan, it has been shown that pigs
are necessary for pre-epizootic amplification of the virus, although
some epidemics do occur in the absence of large pig populations (as is
certainly the case in India and Pakistan). Humans and horses develop
fatal encephalitis, but they are only incidentally infected and are dead-
end hosts of the virus.

Michitomo Hayashi (1885-1973) Shiro Kasahara


[from Kitasato Institute, used with permission]

Hayashi M. Ubertragung des virus von encephalitis epidemica auf affen. Folia Psych et Neurol
Japonica 1935;1:419-465.
Kasahara S, Ueda M, Okamoto Y, Yoshida S, Hamano R, Yamada R. Experimental studies on
epidemic encephalitis. I. Transmission tests of the Japanese encephalitis in 1935 and some
characteristics of the infectious agent. Kitasato Arch Exp Med. 1936;13;48-65.[origin of prototype
Nakayama strain of virus]
Kasahara S, Yamada R, Hamano R. Experimental studies on epidemic encephalitis. 4.
Immunological comparison of viruses of the Japanese (B type) and the American (St. Louis) types.
Kitasato Arch Exp Med. 1937;14:229-233.
Hashimoto H, Kudo M, Uraguchi K. Experiences in the summer epidemics of acute encephalitis in
Tokyo. JAMA. 1936;106:1266.

page 166

1934 Michitomo Hayashi, Shiro Kasahara, others discovery of Japanese encephalitis virus
Top: TMV, purified, negative contrast electron microscopy, minimum exposure study, Robley
Wendell Meredith Stanley (1904-1971) Williams, 1970s. Bottom: TMV in paracrystalline array in the cytoplasm of a tobacco plant cell,
thin section electron microscopy.
In 1935, Wendell Stanley of the Rockefeller Institute for Medical Research at Princeton emerged as a scientific celebrity. The media hailed his work as revolutionary, providing
the key to the riddle of life. By crystallizing tobacco mosaic virus (TMV) he seemed to have demonstrated that viruses are protein molecules, a special class of enzymes, and to
have proved the chemical nature of life. This was the symbolic beginning of molecular biology. Yet Stanleys work was flawed by technical errors and misconceptions. His virus
sample contained water and impurities and thus was not a true crystal. It also contained a significant fraction of nonprotein material: about 6% ribonucleic acid (RNA), which
Stanley had missed entirely, and which turned out to be the crucial component, the stuff of heredity. Furthermore, the property of self-replication in viruses was not based on
any enzymatic activity and crystal growth, as Stanley claimed, but was a direct consequence of the structure of the nucleic acid. Nevertheless, the wonder of crystalline life
and living molecules captured the imagination of many scientists, especially the Rockefeller Institute and the University of Uppsala, where the crystallization of TMV was
seen as a validation of the chemical nature of life and validation of the merit of research based in new technologies.

page 167
Stanley WM. Isolation of a crystalline protein possessing the properties of tobacco-mosaic virus. Science 1935;81:644-645.

1935 Wendell Stanley purification and crystallization of tobacco mosaic virus


Patient Asibi, Dakar, 1935

Max Theiler Hugh Hollingsworth Smith


(1899-1972) (1902-1995)

In 1935, the Asibi strain of yellow fever virus (isolated by Adrian Stokes in Dakar in 1928) was adapted to grow in mouse
embryonic tissue by Max Theiler and Hugh Smith. After 17 passages the virus was further cultivated until passage 58 in
whole chicken embryonic tissue and thereafter, until passage 114, in denervated chicken embryonic tissue. At this stage,
Theiler and Smith injected the virus intracerebrally into monkeys the virus showed a marked reduction in its viscero-
and neuro-tropism as compared to wild type. This virus was further subcultured until passages 227 and 229 and these
viruses were used to immunize 8 human volunteers. Results were satisfactory there were no adverse reactions and all 8
recipients seroconverted within 2 weeks.
Theiler M, Smith HH. The effect of prolonged cultivation in vitro upon the pathogenicity of yellow fever virus. J Exp Med.
1937;65:767-786.
page 168

1935 Max Theiler, Hugh Smith development of yellow fever vaccine


Max Knoll Manfred von Ardenne Vladimir Zworykin Charles Oatley
(1897-1969) (1907-1997) (1888-1982) (1904-1996)

The theory of the scanning electron microscope (SEM) was


developed by Max Knoll in 1935 he then built one, but did
not patent it. Two years later Manfred von Ardenne built an
electron microscope with a probe for scanning transmission
electron microscopy (STEM) and also tried it as an SEM. Soon
afterwards, Vladimir Zworykin and colleagues built a dedicated
SEM. The general use of the SEM began in 1965 when the
Cambridge Instrument Company marketed its Stereoscan 1,
which was developed by Charles Oatley, Gary Stewart and
Dennis McMullan at Cambridge University (this was followed
about 6 months later by an instrument built by JEOL in Japan).
By 1952 the Stereoscan had achieved a resolution of 50 nm
and produced the first micrographs showing the striking Ebola virus, scanning electron microscopy,
three-dimensional imaging characteristics of the modern-day from Cynthia Goldsmith, Centers for
SEM. [Vladimir Zworykin also invented the iconoscope, Disease Control and Prevention
kinemascope, and storage principle that became the basis of Stereoscan 1
TV as we know it, and Manfred von Ardenne achieved the first Cambridge Instrument Company
transmission of a television picture.] 1965, the first commercially produced
scanning electron microscope
page 169

1935 Max Knoll, Manfred v Ardenne, Vladimir Zworykin, Charles Oatley the scanning electron microscope
Frederick Charles Bawden (1908-1972) Norman Wingate Pirie (1907-1997)
Wendell Stanley was nominated for a Nobel Prize in chemistry for the first time in 1938 and 13 times after that until he was awarded it in 1946 it was the first Nobel Prize
in virology. The basis for the award was his crystallization of tobacco mosaic virus (TMV). Stanley concluded that the crystals contained protein, and he likened TMV to
an autocatalytic protein which, for the present, may be assumed to require the presence of living cells for multiplication. Frederick Bawden and Norman Pirie also purified
TMV: their results in 1936 were dramatic. Pirie later stated, In a few weeks using the methods that had been standard in protein chemistry for half a century, we got liquid-
crystalline preparations of an infective nucleoprotein. They showed that TMV particles contain about 95% protein and 5% RNA, that the virus particles are rod-shaped,
can form liquid crystals, and have a regular substructure detectable by X-ray diffraction. Stanley had not detected nucleic acid in his preparations and at first disputed the
evidence for its presence later he admitted that he was wrong and agreed with Bawden and Pirie. However, this admission was not much aired in public venues. Later, some
knowledgeable people suggested that the Nobel Prize should have gone to Bawden and Pirie.
Bawden FC, Pirie NW, Bernal JD, Fankuchen I. Liquid crystalline substances from virus infected plants. Nature 1936;138:1051-1052.
Bawden FC, Pirie, NW. The isolation and some properties of liquid crystalline substances from solanaceous plants infected with three strains of tobacco mosaic virus.
Proc R Soc Lond. 1937;B123:274-320.
page 170

1936 Frederick Bawden, Norman Pirie tobacco mosaic virus is comprised of nucleoprotein, the nucleic acid is RNA
Jean Cuill (1872-1950) Paul-Louis Chelle (1902-1943)

By 1755, the industrial revolution was in full swing and wool was in short supply. The British
parliament asked about the economic effects of a spreading disease in sheep. Thus began the recorded
history of scrapie. By 1850, C. Besnoit and his colleagues, at the Ecole Vtrinaire de Toulouse,
recognized the characteristic neuronal vacuolation in scrapie; they attempted to transmit the disease
via brain and blood from affected sheep. They kept animals for only a few months and the results
were negative. Jean Cuill and Paul-Louis Chelle, taking note of epidemiological studies indicating
incubation periods of >18 months, succeeded in 1936 in transmitting scrapie to two healthy sheep by
intraocular inoculation of brain tissue. They then transmitted the disease via intracerebral, epidural and
subcutaneous routes, and by passing brain tissue through an ultrafilter. The incubation period varied
from one to two years. This was the first step in the long path from thinking that the etiologic agent of
scrapie, and later the agents of other spongiform encephalopathies, was a virus, to the work of Stanley
Prusiner and his colleagues and their conclusion that the agent is a prion, an infectious protein.
Cuill J, Chelle P-L. La maladie dite tremblante du mouton est-elle inoculable? C R Acad Sci. Scrapie, sheep brain (geniculate nucleus)
1936;203:1552-1554. [Ecole Vtrinaire de Toulouse] (top) H&E; (bottom) H&E, counterstained with
antibody to glial fibrillary acidic protein (GFAP)
[micrographs courtesy of Robert Higgins, University of California,Davis]

page 171

1936 Jean Cuill, Paul-Louis Chelle transmission of scrapie prion in sheep using ultrafiltered CNS tissue
Charles J. Armstrong (1886-1967) Thomas Milton Rivers (1888-1962) Erich Traub (1906-1985)
[from Rockefeller Archives, used with permission]
In the 1930s, lymphocytic choriomeningitis (LCM) virus was discovered independently in three laboratories.
Charles Armstrong and Ralph Lillie, at the National Institutes of Health, isolated the virus along with St. Louis
encephalitis virus during the 1933 encephalitis epidemic in St. Louis. They showed that LCM virus was quite
distinct based on unusual pathological changes in the brains of experimentally infected monkeys and mice
(especially a massive lymphocytic infiltration of the choroid plexus and meninges). Armstrong went on to do
extensive virologic and pathogenetic research on LCM. Meanwhile, Erich Traub at the National Naval Research
Laboratory found the same virus in laboratory mice and went on to do the first epidemiological research on
the virus in mouse colonies. Thomas Rivers and T.F. McNair Scott at the Rockefeller Institute isolated the virus
from two cases of aseptic meningitis and went on to do the first prevalence studies of the infection in humans.
Armstrong C, Lillie RD. Experimental lymphocytic choriomeningitis of monkeys and mice produced by a virus
encountered in studies of the 1933 St. Louis encephalitis epidemic. Pub Health Rep. 1934;49:1619-1627.
Traub EA. Filterable virus recovered from white mice. Science 1935;81:298.
Rivers TM, Scott TFMcN. Meningitis in man caused by a filterable virus. Science 1935;81:439. page 172

1936 Charles Armstrong, Thomas Rivers, Erich Traub lymphocytic choriomeningitis virus (the first arenavirus)
During the years 1911-1914, Peyton Rous worked at
disproving objections to generalizations made from
his discovery of virus-induced tumors in chickens. He
wrote, The findings with the chicken tumors largely
demolish the theoretical basis in which objections to an
extrinsic cause for cancer have been built up (1912).
He and Joseph Beard showed that tumors induced by
different viruses were capable of invading tissues and
of metastasizing to distant organs; thus they were true
cancers. Moreover, each independently isolated virus
caused a tumor of a different kind. In order to find
more generally acceptable evidence, Rous and Beard
attempted to extend their observations especially
through carefully devised experiments with the tumors
of other species of animals (1911). Evidently, for the
viral etiology of cancers to be accepted, similar findings
were needed in mammals. The strategy of their work
was determined at that time, but the extension of their
ideas to other species came only after Richard Shopes
discovery of rabbit papillomavirus in 1933. Shope
reported the first experimental studies on cottontail
rabbit papillomavirus (CRPV) infection and its life
cycle. From then on, the CRPV rabbit model was used
extensively to study virological aspects of papillomavirus
infections in vivo, virus-induced carcinogenesis,
immunological features of virus infection leading to
host immune-mediated tumor regression, interactions
between papillomas and co-carcinogens, viral gene
function, etc.
Papillomaviruses infect epithelial tissues of cutaneous
Francis Peyton Rous (1879-1970) Joseph Willis Beard (1906-1983) and mucosal origin and are ubiquitous throughout the
animal kingdom from reptiles to primates. In humans,
more than 100 papillomavirus (HPV) types have been
Rous FP, Beard JW. The neoplastic traits of a mammalian growth due to a filterable virusthe defined genetically, with approximately 15-20 types
Shope rabbit papilloma. Science 1934;79:437-438. associated with various cutaneous and mucosal cancers,
Rous FP, Beard JW. A virus-induced mammalian growth with the characters of a tumor (the most notably cervical cancer.
Shope rabbit papilloma). I. The growth on implantation within favorable hosts. J Exp Med.
1934;60:701-722.
Rous FP, Beard JW, Kidd JG. Observations on the relation of the virus causing rabbit
papillomas to the cancers deriving therefrom. II. The evidence provided by the tumors; general
considerations. J Exp Med. 1936;64:401-424.
Rous FP. The virus tumors and the tumor problem. Am J Cancer 1936;28:233-271.
page 173

1936 Francis Peyton Rous, Joseph Beard progression of rabbit papillomavirus lesions to carcinomas
Vesicular exanthema virus of swine
negative contrast electron microscopy

Vesicular exanthema of swine first occurred in 1932 and was thought to be foot-and-
mouth disease because of the clinical similarities. In 1935, Jacob Traum of the University of
California Davis first isolated the etiologic agent, vesicular exanthema virus of swine (VESV).
The outbreaks were associated with the feeding of uncooked garbage to pigs. Following
legislation requiring the cooking of garbage fed to domestic animals, by 1956 the disease
was eliminated and the virus was declared extinct. Epidemiologic evidence had prompted
Stuart Madin of the Naval Biological Laboratory at the University of California Berkeley to
postulate that the virus had its origin in some natural reservoir and was only incidentally
transmitted by feeding uncooked garbage to swine. The later identification of San Miguel sea
Jacob Traum (1923-1995) lion viruses (SMSV) from aborted sea lion fetuses on the island of San Miguel in California
confirmed his belief the viruses appeared to be nearly identical to VESV. It was found
Traum J. Vesicular exanthema of swine. JAVMA 1936;88:316-327. that sea lion carcasses were fed to pigs causing the initial outbreaks the virus was then
spread widely via uncooked pork scraps from railroad dining cars being fed to pigs at distant
locations. There are 13 genotypes of VESV, and presently 17 genotypes of SMSV recent
phylogenetic comparison of these viruses has shown that they are, indeed, very closely
related, so closely that they should be classified as a single genotype, not just separate viruses
within a genus of the family Caliciviridae.
page 174

1936 Jacob Traum discovery of vesicular exanthema virus of swine (the first calicivirus)
Nothing in Biology Makes Sense
Except in the Light of Evolution
Theodosius Dobzhansky was a prominent geneticist and evolutionary biologist, and in his day the
central figure in developing the unifying modern evolutionary synthesis. The following is a summary
of the modern synthesis, which bridged the gap between experimental geneticists and naturalists, and
between both and paleontologists. It states that:
1. All evolutionary phenomena can be explained
in a way consistent with known genetic
mechanisms and the observational evidence of
naturalists.
2. Evolution is gradual: small genetic changes,
recombination ordered by natural selection.
Discontinuities amongst species (or other taxa)
are explained as originating gradually through
geographical separation and extinction (not
saltation).
3. Natural selection is by far the main mechanism
of change; even slight advantages are important
when continued. The object of selection is the
phenotype in its surrounding environment.
4. The role of genetic drift is equivocal. Though
strongly supported initially by Dobzhansky,
it was downgraded later as results from
ecological genetics were obtained. [The role of
genetic drift in the evolution of viruses, such as
influenza viruses, came much later.]
5. Thinking in terms of populations, rather than
individuals, is primary: the genetic diversity
existing in natural populations is a key factor
in evolution. The strength of natural selection
in the wild is greater than previously expected;
the effect of ecological factors such as niche
Theodosius Dobzhansky (1900-1975) occupation and the significance of barriers to
gene flow are all important.
Dobzhansky T. Genetics and the origin of species. New York: 6. In paleontology, the ability to explain
Columbia University Press; 1937. historical observations by extrapolation from
microevolution to macroevolution is proposed.
Historical contingency means explanations at
different levels may exist. Gradualism does not
mean a constant rate of change.
page 175

1936 Theodosius Dobzhansky publication of Genetics and the Origin of Species, seminal in evolutionary synthesis
Fred Robert Beaudette (1897-1957) Avian infectious bronchitis virus
thin section and negative contrast electron microscopy
Fred Beaudette started his career in avian pathology in 1919 after receiving a DVM from Kansas State College. He left Kansas in 1923 for Rutgers University, where he rose
through the ranks becoming professor, a position he held for the rest of his life. Dr. Beaudette was an interesting character. He was a brilliant man, spoke fluent Russian and
Portuguese. The original poultry pathology books were all written in Portuguese so he had learned the language. Avian infectious bronchitis costs the U.S. poultry industry
millions of dollars annually and it remains one of the top research priorities for the commercial poultry industry worldwide. It is highly contagious and is extremely difficult
to control because immunological variants emerge often due to mutations and recombination events. In addition to genetic drift, genetic shift (recombination via template
switching) can lead to dramatic phenotypic (pathotypic) changes and can result in unique variant viruses requiring new vaccine formulations.
Beaudette FR, Hudson CB. Cultivation of the virus of infectious bronchitis. JAVMA. 1937;90:51-58.
page 176

1937 Fred Beaudette, Charles Hudson discovery of avian infectious bronchitis virus (the first coronavirus)
Viruses and Kochs Postulates
From Thomas Rivers Presidential address delivered before the
Society of American Bacteriologists (now the American Society for
Microbiology) at its Thirty-eighth Annual Meeting, Indianapolis,
Indiana, December 29, 1936:
...very early in the bacteriological era a few discerning individuals
appreciated the fact that there was no reason, except analogy, for
assuming that all infectious agents must be living autonomous
organisms. Through the activities of these investigators a group of
disease-producing agents, known as viruses, has gradually become
recognized. The exact nature of these agents is not known; some may
be the midgets of the microbial universe, others may represent forms
of life unfamiliar to us, while still others may be inanimate incitants
of disease. Regardless of lack of complete knowledge of their nature,
it is decidedly incorrect to say that these agents are unknown. The
incitants of smallpox, vaccinia, poliomyelitis, yellow fever, fowl plague
and tobacco mosaic are known Thus, to the initiate the term virus
used in connection with an infectious agent has lost its old indefinite
meaning and has acquired a new significance similar in exactness to
that borne by the words bacterium and spirochete. Such a statement
does not imply that all viruses are alike in nature and that a subdivision
of the viral group is not essential. The proper time for this subdivision,
however, has not yet arrived.
...At the time when they were formulated Kochs postulates were
essential for the progress of knowledge of infectious diseases; but
progress having left behind old rules requires new ones which some
day without doubt will also be declared obsolete. Thus, in regard
to certain diseases, particularly those caused by viruses, the blind
adherence to Kochs postulates may act as a hindrance instead of an
Thomas Milton Rivers (1888-1962) aid. It is obvious that Kochs postulates have not been satisfied in viral
[from Rockefeller Archives, used with permission]
diseases. Moreover, it is equally evident that proof of the etiological
significance of viruses has been obtained without their satisfaction.
Such a statement, however, does not imply that certain conditions do
not have to be met before the specific relation of a virus to a disease is
Rivers Criteria for Proof of Viral Disease Causation established. The conditions are [as listed in the separate box]
A specific virus must be found associated with a disease with a degree of regularity. ...To summarize, Kochs postulates as proposed by him do not have
The virus must be shown to occur in the sick individual not as an incidental or accidental to be fulfilled in order to prove that a virus is the cause of a disease.
finding but as the cause of the disease under investigation. However, the spirit of his rules of proof still holds in that a worker must
demonstrate that a virus is not only associated with a disease but that
Information concerning the presence of antibodies against the agent and the time of
it is actually the cause. The methods of doing this are different from the
their appearance in the serum of patients is equally important as evidence of etiological
ones used by Koch but are equally efficient.
significance of the virus.

page 177

1937 Thomas Rivers criteria for proof of viral disease causation: the Henle-Koch postulates revisited
Zilber LA, Levkovich EN,
Shubladze AK, Chumakov MP,
Soloviev VD, Shboldaeva AD,
Safonova TI. Etiology of spring-
summer encephalitis. Arch Biol Sci.
1938;52:162-183. (in Russian)
The discovery of tick-borne
encephalitis in the far east of
the USSR in 1937 was marked
by courage and tragedy there
were infections in members of the
expedition and deaths when the
first vaccine was tested in 1938. Lev
Zilber and two others were arrested
on charges of subversive activities:
spreading Japanese encephalitis in
the guise of discovering a new virus.

Lev Alexandrovich Zilber Elizabeth N. Levkovich Mikhail Petrovich Chumakov


(1894-1966) (1900-1982) (1909-1993)

Tick-borne encephalitis virus, mouse brain, thin section electron microscopy page 178

1937 Lev Zilber, Mikhail Chumakov, Elizabeth Levkovich discovery of tick-borne encephalitis virus
Plaque honoring Elisabeth Wollman and Eugne Wollman August H. Doermann (1918-1991)
at the Institut Pasteur, Paris
From Esther Zimmer Lederberg: ...regarding the famous geneticist, lie Leo Wollman, long time staff member and
deputy director of the Institut Pasteur: His parents, Elisabeth Wollman and Eugne Wollman, were researchers at
the Institut Pasteur. It was during World War II. The Gestapo arrested these Jewish scientists (they were sent to
Auschwitz and never seen again). Their son, lie, was walking towards the Institut to meet his father. Friends of the
parents quickly grabbed the young boy, and hid him from the Gestapo officers who were waiting for him, with his
father. Later, these friends hid the young boy in the catacombs of the Institut Pasteur, and took care of him until
the end of the war. He survived the Holocaust. Andr Lwoff had known the Wollmans in the thirties as scrupulous
and dogged experimentalists: they did seminal work leading to the development of the concept of the eclipse phase
in virus (bacteriophage) replication. lie Wollman later fought with the maquis in the south of France, and in 1945
joined Lwoff s unit at the Pasteur, just a few weeks before Jacque Monod. From: Judson, HF. The eighth day of
creation: the makers of the revolution in biology. New York: Simon and Schuster; 1979.
Doermann AH. The eclipse in the bacteriophage life cycle. In: Cairns J, Stent GS, Watson JD, editors. Phage and the
origins of molecular biology. Cold Spring Harbor, NY: Cold Spring Harbor Laboratory Press; 1992. p. 79-87.

page 179

1937-1943 Elizabeth Wollman, Eugne Wollman, August Doermann discovery of the eclipse period
In quantitating a virus by serial dilution assay, the measured endpoint is usually taken as
the dilution of virus in which half of treated animals or cells show the desired reaction
(death LD50, infection ID50, cytopathology TCID50, per volume of specimen, etc.),
and the other half do not. That is, the dilution endpoint is taken as the highest dilution that
will produce the desired reaction in 50% of the animals or cells used. Accurate endpoint
dilution measurements are very difficult and expensive to perform, so reasonable numbers
of replicate samples at dilutions around the predicted endpoint are tested in animals or cells
and the results are interpolated by classic methods: the methods of Reed and Muench, from
1938, or the method of Spearman-Krber, from 1931. More modern methods are available,
including several based in complex computer programs, but in most virology laboratories
the old classic methods are still used. Most virologists never think about the statistical bases
for the Reed-Muench and Spearman-Krber methods the methods lend themselves to
plugging data-points (dilutions vs. effects) into the formulae as shown in the two boxes.
These are taken from the book, Diagnostic Procedures for Viral and Rickettsial Diseases, by
Edwin H. Lennette, Nathalie J. Schmidt, several editions of which were published over the
years by the American Public Health Association.
Krber G. Beitrag zr kollektiven behandlung pharmakologischer reihenversuche. Arch Exp
Path Pharmak. 1931;162: 480-487.
Reed LJ, Muench H. A simple method of estimating fifty percent endpoints. American
Journal of Hygiene 1938;27: 493-497.

Lowell J. Reed (1886-1966) Hugo Muench (1894-1972) page 180

1938 G. Krber, Lowell Reed, Hugo Muench development of methods for estimating fifty percent endpoints
Vladimir Kubes
(1904-1982)

Journal of Virology
Francisco A. Rios Ralph Walter Wyckoff cover art from Scott Weaver
(1897-1994)
from Scott Weaver, used with permission
page 181

1938 Vladimir Kubes, Francisco Rios discovery of Venezuelan equine encephalitis virus
Handbuch der Virusforschung. Edited by Prof. Dr. R. Doerr
and Prof. Dr. C. Hallauer. With Contributions by F.M.
Burnet, R. Doerr, W.J. Elford, G.M. Findlay, M. Haitinger,
C. Hallauer, M. Kaiser, and W.M. Stanley. Erste Hlfe.
Illustrated, pp. I-XII, 1-546. Wien:Julius Springer; 1938

Robert Doerr (1871-1952) Curt Hallauer (1900-1994)


From an unsigned review, Journal of the American Medical Association, 1939: This is a
collective work by a most competent group of specialists in a field of remarkable activity, of
astounding results and of greatest scientific significance. The viruses lie at the lowest margin
of the living world or beyond it, depending on the concept of the limits of living substance.
They appear to be protein substances capable of self propagation within living cells and to
be composed of units mostly but not wholly beyond the limits of visibility or photographic
detection even with the ultramicroscope. They can be centrifuged, filtered through graded
collodion membranes, purified by filtration and high speed centrifugation from organic
contaminants, and inactivated and reactivated by various chemical procedures. Their
dimensions can be determined within limits and their activity retained between a series of
crystallizations. They differ among themselves not only in their pathologic consequences
but in dimensions and in physical and chemical properties. They can be cultivated in tissue
cultures and grown on chick allantoic membranes Perhaps this is a realistic view of the
virus world in 1939 from outside the citadel even today, the viruses are mysterious to many
people who know quite a bit about biology in general.
page 182

1938 Robert Doerr, Curt Hallauer publication of the major virology book, Handbuch der Virusforschung
The First Virus Visualized by Electron Microscopy
There is a large, confused literature on this, with many claims of primacy, but
the actual record is quite clear Bodo von Borries, Ernst Ruska and Helmut
Ruska published the first paper, which clearly showed the virions claimed.
Helmut Ruska went on to study several viruses by the same methods:
1938: von Borries B, Ruska E, Ruska H. Bakterien und virus in
bermikroskopischer aufnahme. Klin Wochenschr. 1938;17:921-925. [images
of ectromelia virus and vaccinia virus]
1939: Kausche GA, Pfankuch E, Ruska H. Die sichtbarmachung von
pflanzlichem virus [TMV] im bermikroskop. Naturwissenschaften
1939;27:292-299.
1940: Ruska H, von Borries B, Ruska E. Die bedeutung der bermikroskopie
fr die virusforschung (The significance of electron microscopy for virus
research). Arch Ges Virusforsch. 1940;1:155-169.
1940: Ruska H. Die sichtbarmachung der bakteriophagen lyse im
bermikroskop. Naturwissenschaften 1940;28:45-46.
1942: Ruska H. ber das virus der varicellen und des zoster. Klin Wochenschr.
1942;22:703-704.
Bodo von Borries (1905-1956) Ernst Ruska (1906-1988)

Ectromelia (mousepox) virus T2 bacteriophage

page 183 Helmut Ruska (1908-1973)


1938 Bodo von Borries, Ernst Ruska, Helmut Ruska first electron micrographs of viruses (ectromelia, vaccinia)
Archiv fr die Gesamte Virusforschung / Archives of
Virology, 1939, Volume 1, Number 1 Table of Contents
P. Jordan. Die Stellung der Quantenphysik zu den aktuellen
Problemen der Biologie. Archives of Virology, 1939, Volume
1, Number 1, Pages 1-20.
A. Gratia and P. Manil. Recherches sur les virus des plantes.
Archives of Virology, 1939, Volume 1, Number 1, Pages 21-
45.
Erich Khler. ber das Auftreten abweichender Varianten bei
den Cs-Stmmen des Kartoffel-X-Virus. Archives of Virology,
1939, Volume 1, Number 1, Pages 46-69.
C. Hallauer. Studien ber die Variabilitt des Hhnerpestvirus
im Gewebsexplantat. Archives of Virology, 1939, Volume 1,
Number 1, Pages 70-84.
M. Kaiser. Die Trockenkonservierung von Virusarten. Archives
of Virology, 1939, Volume 1, Number 1, Pages 85-90.
Helga Jahn. Experimentelle Untersuchungen ber das Virus
der Ektromelia infectiosa. Archives of Virology, 1939,
Volume 1, Number 1, Pages 91-103.
W. Gavrilov and A. Fester. Quelques proprits du virus
de sarcome Rous. Archives of Virology, 1939, Volume 1,
Number 1, Pages 104-113.
R. W. Fairbrother and A. E. Martin. Further observations
on the value of heated elementary body suspensions in
immunization against experimental influenza. Archives of
Virology, 1939, Volume 1, Number 1, Pages 114-119.
Robert Doerr (1871-1952) Curt Hallauer (1900-1994)
A. Paillot and A. Gratia. Essai disolement du virus de la
grasserie des vers soie par lultracentrifugation. Archives of
In the first volume of Archiv fr
Virology, 1939, Volume 1, Number 1, Pages 120-129.
die gesamte Virusforschung, there 1984
was an article on the significance of A. Gratia and A. Paillot. tude srologique du virus de la
ultramicroscopy in the evaluation of the grasserie des vers soie isol par ultracentrifugation.
nature of viruses by Helmut Ruska (1908- Archives of Virology, 1939, Volume 1, Number 1, Pages 130-
1973). The paper was written with his 139.
brother, Ernst Ruska (1906-1988), and his K. Burckhardt. Die Durchlssigkeit der Blut-Liquor-Schranke
brother-in-law Bodo von Borries (1905- und der Placenta fr Bakteriophagen. Archives of Virology,
1956). 1939, Volume 1, Number 1, Pages 140-154.
Helmut Ruska, Bodo v. Borries and Ernst Ruska. Die
Bedeutung der bermikroskopie fr die Virusforschung.
Archives of Virology, 1939, Volume 1, Number 1, Pages 155-
169.

page 184

1939 First international virology journal, Archiv fr die gesamte Virusforschung (now Archives of Virology)
Charles J. Armstrong (1886-1967)

While still doing research work on lymphocytic


choriomeningitis (LCM) virus in 1939, Charles Armstrong
adapted a human strain of poliovirus (Lansing type 2 virus)
from monkeys to small rodents, first to cotton rats and then
to mice. This accomplishment was considered revolutionary. Scientists who played leading roles in poliomyelitis research met beneath bronze busts at the dedication
His bust is included in the Polio Hall of Fame at Warm of the Polio Hall of Fame in Warm Springs, Georgia. Left to right: Dr. Thomas M. Rivers, Dr. Charles
Springs, Georgia. Armstrong, Dr. John R. Paul, Dr. Thomas Francis, Jr., Dr. Albert B. Sabin, Dr. Joseph L. Melnick, Dr. Isabel
Morgan, Dr. Howard A. Howe, Dr. David Bodian, Dr. Jonas E. Salk, Mrs. Eleanor Roosevelt, and Mr. Basil
Armstrong C. The experimental transmission of OConnor. February, 1958.
poliomyelitis to the Eastern cotton rat, Sigmodon hispidus
hispidus. Pub Hlth Rep. 1939;54:1719-1721.

page 185

1939 Charles Armstrong adaptation of poliovirus to cotton rats and mice


The Polio Hall of Fame, Warm Springs, Georgia

(left to right)
1. Jakob Heine (1800-1879) Described infantile paralysis in 1840.
2. Karl Oskar Medin (1847-1927) Described polio as an acute infectious disease (1890).
3. Ivar Wickman (1872-1914) Described epidemic polio and non-paralytic polio (1907).
4. Karl Landsteiner (1868-1943) Discovered poliovirus and transmitted the virus to monkeys.
5. Thomas M. Rivers (1888-1962) Planned and ran the successful 1954 inactivated vaccine field trials.
6. Charles Armstrong (1886-1967) Transmitted poliovirus to cotton rats and mice.
7. John R. Paul (1893-1972) Described polio natural history, spread and epidemiology.
8. Albert Sabin (1906-1993) Developed the attenuated live-virus polio vaccine that bears his name.
9. Thomas Francis, Jr. (1900-1969) Conducted the field trials of the Salk inactivated vaccine.
10. Joseph L. Melnick (1914-2001) Developed assays of polio immunity in populations.
11. Isabel Morgan (1911-1996) Tested an experimental inactivated-virus vaccine in monkeys.
12. Howard A. Howe (1901-1976) Showed that chimpanzees can acquire polio infection by mouth.
13. David Bodian (1910-1992) Showed initial viremia before central nervous system invasion.
14. John F. Enders (1897-1985) Developed cell culture methods for growing poliovirus in volume.
15. Jonas E. Salk (1914-1995) Developed the inactivated-virus vaccine that bears his name.
16. Franklin D. Roosevelt (1882-1945) Founded the National Foundation for Infantile Paralysis.
17. Basil OConnor (1892-1972) The architect of the first national polio vaccination campaign.

Charles J. Armstrong (1886-1967) page 186

1939 Charles Armstrong adaptation of poliovirus to cotton rats and mice


DDT, dichloro-diphenyl trichloroethane, was first synthesized
in 1874, but it gathered dust until 1939 when Paul Mller, of JR
Geigy AG of Basle, Switzerland, discovered its amazing insecticidal
qualities. DDT had a major role in improving the health of military
personnel in World War II . A 1945 military document states, At
present the armed services are using all the DDT available and it
is not expected that any will be available for civilian use soon
Already the army has checked a typhus epidemic in Naples and
is preventing malaria and dengue epidemics in the Pacific. After
the war, more than 4 billion pounds of DDT were used throughout
the world (1.35 billion pounds in the U.S.), until 1973 when the
Environmental Protection Agency stopped all use in the U.S. and
Paul Hermann Mller most other countries rapidly followed suit. The largest amount of
(1899-1965) this usage was in agriculture, where as the illustrations here show,
very high concentrations were used regularly. The ecologically
damaging effect of this usage in turn affected the scientific-political
debate whether usage at very low concentration to repel mosquitoes
is safe when DDT is applied to the inside wall of homes it is
very effective as a mosquito repellent; there is little environmental
effect and little development of DDT resistance of mosquitoes that
carry malaria, dengue and other pathogens. Nevertheless, very few
jurisdictions are willing to allow its use. The mode of action of DDT
has never been clearly established, but in some complex manner
p,p-dichloro-diphenyl- it opens sodium and potassium ion channels in neurons of insects
trichloroethane and prevents normal nerve impulse transmission.
[DDT] Mller P. Triclorethane insecticidal composition and methods. US
patent 2329074, issued 1943, assigned to JR Geigy AG.

page 187

1939 Paul Mller discovery of the insecticidal qualities and use of DDT for the control of vector-borne diseases
Max Ludwig Henning Delbrck Emory Leon Ellis (1906-1993)
(1906-1981) [from Caltech Archives, used with permission]
[from Caltech Archives, used with permission]

In 1937, Max Delbrcks initial introduction to Caltech was quite difficult. One day he inadvertently failed to attend a seminar on bacteriophages by Emory Ellis, and went
to him later to find out what he had missed: I had sort of the vaguest notions that viruses might be an interesting experimental object for a study of reproduction at a basic
level. Ellis showed him the simple materials and techniques needed for experiments, and Delbrck saw for the first time plaques made in a lawn of bacteria where single
bacteriophage had initiated infection. Ellis also demonstrated one step-growth curves, revealing the kinetics of the cycle of bacteriophage multiplication. According to
Ellis, Delbrcks first comment was, I dont believe it, but Delbrcks recollection was, This seemed to me just beyond my wildest dreams of doing simple experiments on
something like atoms in biology and I asked him whether I could join him in his work, and he was very kind and invited me to do so. Ellis and Delbrck invented and greatly
refined the one-step growth curve (the single-burst experiment), which permitted study of bacteriophage multiplication in individual cells, a key in later research.
Ellis EL, Delbrck M. The growth of bacteriophage. J Gen Physiol. 1939;22:365-384. page 188

1939 Emory Ellis, Max Delbrck the one-step growth experiment (now called the single burst experiment)
Kenneth C. Smithburn John Rodman Paul Thomas P. Hughes (1900-1995)
(1904-1973) (1893-1971) with Wilbur Sawyer at Rockefeller Institute, 1930s
West Nile virus (WNV) was first isolated from a feverish adult
woman in the West Nile District of Uganda in 1937 during research
on yellow fever. Serosurveys in 1939 found antibody ranging from
1.4% (Congo) to 46.4% (White Nile region, Sudan). The virus was
subsequently identified in Kenya (1942) and India (1953); a 1950
serosurvey in Egypt found 90% of those over 40 years of age had
WNV antibodies. The ecology of the virus was characterized in
1953 in studies in Egypt and Israel. The virus became recognized
as a cause of severe human meningoencephalitis in elderly patients
during an outbreak in Israel in 1957. In the 1960s the disease was
found to be widespread in southern Europe and southwest Asia.
Smithburn KC, Hughes TP, Burke AW, Paul JH. A neurotropic virus
isolated from the blood of a native of Uganda. Am J Trop Med Hyg.
1940;20:471-492.
West Nile Virus, Global Distribution
page 189 Alexander W. Burke (1886-1944)

1940 Kenneth Smithburn, Thomas Hughes, Alexander Burke, John Paul discovery of West Nile virus
Ernst Mayr was the leading evolutionary biologist of
his day. He was also a renowned taxonomist, tropical
explorer, ornithologist, historian of science and naturalist.
His work contributed to the conceptual revolution that
became the modern evolutionary synthesis, unifying
concepts from (a) Mendelian genetics, (b) systematics, (c)
paleontology, and (d) the original Darwinian principles.
From Mayrs book, Systematics and the Origin of Species,
Columbia University Press, New York, 1942, and from
What Evolution Is, Basic Books, New York, 2001: ... I
was much struck how entirely vague and arbitrary is the
distinction between species and varieties. Darwin,
1859.
In many of his writings, Mayr rejected reductionism in
evolutionary biology, arguing that evolutionary pressures
act on the whole organism, not on single genes, and that
genes can have different effects depending on the other
genes present. He advocated a study of the whole genome
rather than of isolated genes only. After articulating
the biological species concept in 1942 (acknowledging
Theodosius Dobzhanskys earlier work in this area), Mayr
played a central role in the problem of defining species.
He staunchly defended the biological species concept
against the many definitions that others proposed. Mayr
insisted throughout his career that the gene as the target
of selection cannot and should not be considered a valid
idea in modern evolutionary thought: The idea that
a few people have about the gene being the target of
selection is completely impractical; a gene is never visible
Ernst Walter Mayr (1904-2005) to natural selection, and in the genotype, it is always
in the context with other genes, and the interaction
with those other genes make a particular gene either
more favorable or less favorable. Therefore people
like Dawkins in England who still think the gene is the
target of selection are evidently wrong. In the 30s and
40s, it was widely accepted that genes were the target of
selection, because that was the only way they could be
made accessible to mathematics, but now we know that
it is really the whole genotype of the individual, not the
gene. Except for that slight revision, the basic Darwinian
theory hasnt changed in the last 50 years.

page 190

1940 Ernst Mayr, others development of the conceptual basis of the modern evolutionary synthesis
Barbara McClintock (1902-1992)
Barbara McClintock began her studies at the College of Agriculture, Cornell University, in 1919. Her interests became focused when she took her first genetics course taught by
C. B. Hutchison in 1921. He was impressed by her interest, and telephoned to invite her to participate in his graduate genetics course; later, she said that Hutchinsons invitation
was the reason she continued in genetics: Obviously, this telephone call cast the die for my future. I remained with genetics thereafter. In 1948, working as always with maize,
she discovered the first transposons she noticed insertions, deletions, and translocations, caused by these transposons. She hypothesized that gene regulation via transposons
(sequences of DNA that can move to different positions within the genome of a cell) could explain how complex multicellular organisms made of cells with identical genomes
can have cells with vastly different functions. These changes in the genome could, for example, lead to changes in the color of corn kernels. About 50% of the total genome of
maize consists of transposons. [The most common transposon in humans is the Alu sequence, which is ~300 bases long and can be found between 300,000 and a million times
in the human genome.] The importance of McClintocks contributions came to light in the 1960s, when Franois Jacob and Jacques Monod described the molecular basis for
genetic regulation of the bacterial lac operon, a parallel system to that which she studied years earlier. Their work also explained the mechanism of transposition
McClintock B. The origin and behavior of mutable loci in maize. Proc Natl Acad Sci USA. 1950;36:344-355.
page 191

1940> Barbara McClintock development of the concept of transposable elements, transposons


Katherine K. Sanford (1915- ) Wilton R. Earle (1902-1964)

The fibroblast-like L cell line was established in 1940 by Wilton Earle from normal C3H/An mouse subcutaneous areolar
and adipose tissue; it was one of the first cell lines to be established in continuous culture. Clone L929 was the first cloned
cell line to be established; this was done by Kay Sanford, Wilton Earle, and G.D. Likely in 1948, using Sanfords capillary
technique for single cell isolation. The cloned cell line was given the designation ATCC CCL-1 in the catalog of the newly
founded American Type Culture Collection. Both the cell line and its cloned derivative became widely used in virology
because of their broad spectrum of virus susceptibility: pseudorabies virus, vesicular stomatitis viruses, herpes simplex
virus, B virus, vaccinia virus, and many others replicate well in these cells.
page 192

1940-1948 Katherine Sanford, Wilton Earle development of the L cell line and L929 cloned cell line
Keith Porter is considered the father of cell biology. In the 1940s, using thin-section
electron microscopy, he initiated studies of the fine structure of cells and contributed
to an appreciation of the importance of understanding the relationship between cell
structure and cell function. He was the driving force behind the founding of the Tissue
Culture Society, the American Society for Cell Biology, the Electron Microscopy
Society of America (now the Microscopy Society of America), the Journal of Cell
Keith Roberts Porter (1912-1997) Biology, and the International Federation for Cell Biology. In 1977, Porter was awarded
the U.S. National Medal of Science, presented by President Jimmy Carter.

page 193

1940s> Keith Porter, others description of the fine structure and biochemistry of cellular organelles
U.S. National Medal of Science
(Recipients Cited in this Book)

David Baltimore
Paul Berg Copley Medal of the Royal Society
J. Michael Bishop (Recipients Cited in this Book)
Herbert W. Boyer
Mario R. Capecchi 1753: Benjamin Franklin
Stanley N. Cohen 1845: Theodor Schwann
Theodor O. Diener 1864: Charles Darwin
Harry Eagle 1874: Louis Pasteur
U.S. Presidential Medal of Freedom Donald A. Henderson 1892: Rudolf Virchow
(Recipients Cited in this Book) Maurice R. Hilleman 1902: Lord Lister
Leroy Hood 1906: Elias Metchnikoff
Carl Ten Broeck Robert J. Huebner 1917: Emile Roux
James E. Darnell Har Gobind Khorana 1933: Theobald Smith
William McDowall Hammon Arthur Kornberg 1939: Thomas Hunt Morgan
John R. Paul Joshua Lederberg 1945: Oswald Theodore Avery
John F. Enders Ernst Mayr 1949: George Charles De Hevesy
Thomas Francis Jr. Barbara McClintock 1959: Frank Macfarlane Burnet
James D. Watson Daniel Nathans 1969: Peter Medawar
Jonas E. Salk Severo Ochoa 1975: Francis Crick
Albert Bruce Sabin George Emil Palade 1977: Frederick Sanger
D.A. Henderson Keith Roberts Porter 1979: Max Perutz
Anthony Fauci Stanley Prusiner 1983: Rodney Porter
William Foege Albert B. Sabin 1985: Aaron Klug
Phillip A. Sharp 1989: Cesar Milstein
Maxine F. Singer 1991: Sydney Brenner
Howard M. Temin 1993: James D. Watson
Harold Varmus 1995: Frank Fenner
James D. Watson 2001: Jacques Miller
Carl R. Woese 2009: Martin Evans
Rosalyn S. Yalow
page 194

Recipients of U.S. Presidential Medal of Freedom, National Medal of Science and Copley Medal of the Royal Society
Albert Claude (1899-1983) Christian de Duve (1917-2013) George Palade (1912-2008)

The Nobel Prize in Physiology or Medicine of 1974 was awarded jointly to Albert Claude, Christian de Duve and George Palade for their discoveries concerning the structural
and functional organization of the cell. The cell had been studied since the middle of the 19th century, but its structure, composition and functions were quite limited until
two different approaches, both introduced at The Rockefeller Institute, changed everything. One was the application of electron microscopy, the other was biochemical
analysis of cellular components (organelles) that were being seen in the electron microscope. Tissues or cells were homogenized and components separated by differential
ultracentrifugation. Fractions were re-examined by electron microscopy and then subjected to many biochemical analyses. The two technologies taken together gave rise
to the discipline of cell biology. Albert Claude, from Belgium, working at the Rockefeller Institute, was the first to apply the two technologies for the study of animal cells.
He published the first electron micrographs of cells and cell components that provided new and relevant biological information in 1945; shortly thereafter he published the
methodology of differential ultracentrifugation. His approach was taken up by George Palade, from Romania, also working at the Rockefeller Institute, in 1947. He added
important methodological improvements both to differential centrifugation and electron microscopy. His early work, done in collaboration with Keith Porter, was mainly
descriptive of the cytoplasmic organelles the endoplasmic reticulum (originally discovered by Claude and Porter), ribosomes (showing with others that they are the site
of protein synthesis), secretory granules and secretory processes, and the Golgi complex. Christian de Duve, from Belgium by way of the United Kingdom, also working at
the Rockefeller Institute, studied the distribution of different enzymes among the four major differential ultracentrifugation fractions [nuclei, mitochondria, microsomes
(fragmented endoplasmic reticulum) and the soluble fraction]. He found that particular enzymes sedimented with particular fractions, and discovered lysosomes and their
complex, compartmentalized proteolytic degradative functions.

page 195

1940s> Albert Claude, Christian de Duve, George Palade fine structure and biochemistry of cellular organelles
Many viruses can agglutinate erythrocytes, usually under specific conditions
(species of mammal or bird providing the erythrocytes, temperature, pH, etc.).
Examples of viruses that hemagglutinate include influenza viruses, parainfluenza
viruses, adenoviruses, rubella virus, alphaviruses, bunyaviruses, flaviviruses and
some picornaviruses. Influenza viruses have an envelope protein complex, the
hemagglutinin (HA), which binds to sialic acid receptors on erythrocytes (and the
same receptors on airway epithelial cells). Other virion surface protein complexes
comprise the hemagglutinin of other viruses. Virions bind to erythrocytes in a
cross-linking fashion, causing the formation of a compact mass of erythrocytes
that settles to the bottom of the well or tube as a pellet or button. This is the
basis of a classical rapid assay to determine levels of virus present in a sample.
To conduct the assay, two-fold serial dilutions of a sample containing virus are
prepared, mixed with a specific amount of erythrocytes, and added to tubes or the
wells of a plastic tray. Where the erythrocytes are cross-linked by virions a pellet
or button is seen at the bottom of the tube or well. Where the erythrocytes are
not cross-linked by virions they settle out smoothly covering the whole bottom of
the tube or well. The assay can be performed within 30 minutes, and is therefore a
quick indicator of the relative quantity of virus present in a sample.
The hemagglutination assay can be easily modified to determine the level of
antibodies present in a serum sample this is called the hemagglutination-
George K. Hirst (1910-1994) inhibition (HAI) assay. In HAI assays the specificity is usually rather narrow:
with influenza it is strain specific, with flaviviruses it is somewhat broadly cross-
reactive. A fixed amount of virus is added to each tube or well; then, two-fold
Hirst GK. The agglutination of red cells by allantoic fluid of chick dilutions of each serum to be tested are added, and finally erythrocytes are added
embryos infected with influenza virus. Science 1941;94:22-23. and left at a given temperature for 30 minutes. Antibodies present in the serum
sample inhibit the formation of a cross-linked pellet of erythrocytes the highest
Hirst GK. The nature of hemagglutination by viruses. Harvey Lect
dilution of serum that prevents hemagglutination is called the HI titer of the
Series 1948;44:84-98.
serum. While less accurate than a plaque assay or plaque-inhibition assay, HA and
HAI assays are cheaper and quicker and for years were widely used. page 196

1941 George Hirst discovery of influenza virus hemagglutination, hemagglutination-inhibition, neuraminidase


The notion that rubella was only a mild illness of children was dispelled in 1941, when Norman
Gregg, an Australian ophthalmic surgeon, reported the devastating teratogenic effects of the
virus. In 1940, Australia had experienced an exceptional epidemic of rubella, the magnitude
of which was probably enhanced by wartime mobilization. Gregg observed an unusually large
number of cases of congenital cataracts in newborn children, and the cataracts were often
associated with other deformities. Today we know that the congenital rubella syndrome includes
deafness, eye abnormalities (especially cataract, microphthalmia), congenital heart disease
(especially patent ductus arteriosus), mental retardation, and other less common defects. Gregg
obtained careful histories from the mothers of the affected babies in an attempt to assign a cause
to the episode. He had 13 such cases in his own practice, and with colleagues he managed to
collate a total of 78 cases of children with cataracts; of these, 68 mothers gave a history of rubella
Norman McAllister Gregg (1892-1966) infection early in pregnancy. Soon after, he unraveled the same relationship between congenital
deafness and maternal rubella infection. Thus, the idea that viruses could be teratogenic agents
was introduced. Greggs report initially drew little attention. It was supported by his Australian
Gregg NM. Congenital cataract following German measles in the mother.
colleagues, who further reported other defects associated with maternal rubella, but it was
Trans Ophthalmol Soc Aust. 1941;3:35-46.
not until the syndrome was confirmed in Europe and the U.S. that the significance of Greggs
page 197 observations was accepted.

1941 Norman Gregg discovery of congenital abnormalities caused by rubella virus infection during pregnancy
Culex tarsalis

Hammon WM, Reeves WC,


Brookman B, Izumi EM,
Gjullin CM. Isolation of the
viruses of Western equine and
St. Louis encephalitis from
Culex tarsalis mosquitoes.
Science 1941;94:328-330.

William McDowall Hammon William Carlisle Reeves


(1904-1989) (1917-2004)
News Item: Mosquitoes and Encephalitis in the Yakima Valley (JAMA. 1942;119:1430-1431.)
In a series of recent studies by Hammon, Reeves and their colleagues much new information has been made
available on the transmission of encephalitis in the Yakima Valley of Washington state. In a four month period
during the summer of 1941 over 15,000 arthropods were collected, frozen and inoculated into laboratory animals
for the purpose of isolating the encephalitis virus. From Culex tarsalis Coquillett three strains of St. Louis
encephalitis virus and five strains of western equine encephalomyelitis virus were isolated. Virus was not isolated
from other species of mosquitoes. For the isolation intracerebral inoculation of five Swiss mice of a suitable strain
was proved by comparative tests to be more satisfactory than intracerebral inoculation of one 200 Gm. guinea pig
or two 8 to 12 day chick embryos. By means of precipitin tests it was demonstrated that C. tarsalis feeds in nature
on cows, horses, man, pigs, dogs, chickens (other birds?) and sheep. Since C. tarsalis has been shown to be an
efficient vector of western equine and St. Louis viruses, this widespread feeding makes it possible for the species
to spread the infection among many animals and birds. In the course of these investigations an efficient trap for
collecting live mosquitoes was developed. Finally the investigators summarize the evidence against Culex tarsalis
and conclude that it is the most important vector of western equine and St. Louis encephalitis viruses in the Bill Reeves and Bill Hammon, Yakima Valley
Yakima Valley. Its possible role elsewhere is wisely left to be judged on the basis of local observations.
page 198

1941 William Hammon, William Reeves quantitative study of natural history of arboviruses in vector arthropods
The Beckman Model DU Ultraviolet Spectrophotometer

Beginning in 1940, the first in what became a series of Beckman spectrophotometers was
developed at the National Technical Laboratories Company headed by Arnold Beckman.
This later became the Beckman Instrument Company. Initially, the sole product of the
company was the worlds first pH meter, which Beckman had invented. Next, in 1941,
came the Beckman Model DU ultraviolet spectrophotometer, which was considered the
Model T of laboratory instruments. It was referred to by Nobel laureate Bruce Merrifield
as probably the most important instrument ever developed towards the advancement of
bioscience. The Model DU was an immediate success and it had a commercial lifetime
of 35 years, with more than 30,000 sold. Among other uses, the Model DU was used in
quantifying protein and DNA concentrations in all sorts of samples. Several amino acids
found in most proteins, including tryptophan, absorb light in the 280nm range and DNA
absorbs light in the 260nm range. Reasonable estimates of protein or DNA concentration
were made by simple measurements of absorbance of light at these wavelengths relative
to standards. Additionally, the ratio of 260/280nm absorbance was found to be a good
Arnold O. Beckman (1900-2004) general indicator of the relative purity of a solution in terms of protein and DNA.
Eventually, every virology laboratory in the world had, or had access to, a Beckman
pH meter, a Beckman DU spectrophotometer, a Beckman analytical ultracentrifuge, a
Beckman preparative ultracentrifuge, and other Beckman instruments.

page 199

1941 Arnold Beckman development of the ultraviolet spectrophotometer (the Beckman DU)
Thomas Foxen Anderson (1911-1991)

The earliest electron micrographs of many


bacteriophage particles showed them to be tadpole-
shaped structures with round or oval heads to which
tails of various thicknesses and lengths were attached
this was realized from the early work of Helmut Ruska
in 1941, but was made much clearer by the work of
Thomas Anderson. The secret of his success, before the
invention of negative contrast electron microscopy, was
his development of the critical point drying method
and Robley Williams development of a freeze-drying
technique, both reducing distortions in specimens. These images illustrate negative contrast electron microscopy of various bacteriophages. This method was invented
Anderson stated in 1953 that, It turns out that the in 1959 by Sydney Brenner and Robert Horne it was not available when Thomas Anderson did his work in
heads of all the phage particles so far prepared by these the 1940s. His images were not as revealing, but he made the right interpretation of the images he produced by
methods are polyhedral in shape. He studied T1, T2, T5 the methods he invented, especially his critical point drying technique. Clockwise: Escherichia coli T1 phage,
and P1 bacteriophages. Staphylococcus aureus 3A phage, Listeria A118 phage, Salmonella phage. page 200

1941 Thomas Anderson discovery of the amazing variety of bacteriophage morphologies


John F. Enders
The Cultivation of the Poliomyelitis Viruses in Tissue Culture
Nobel Lecture, December 11, 1954
These indices of viral multiplication in suspended cell cultures, useful as they
were in our earlier investigations, proved inconvenient... Accordingly, other
means were sought by which viral cytopathogenicity might be conveniently and
rapidly demonstrated. At first we explored the method of explanation which
had been used in the past by other workers, in particular by C.H. Huang, to
test the effect of viruses on cell viability. This consists in placing the fragment
taken from a suspended cell culture in a drop of clotted chicken plasma and
observing whether or not cell outgrowth occurs upon subsequent incubation.
Results of experiments with poliomyelitis virus carried out in this manner were
unequivocal...
To quantitate virus, this approach was adapted to serial dilutions of virus
inoculated into cell culture tubes, with the endpoint determined by color change
using a pH indicator. C.H. Huang first did this using Western equine encephalitis
virus and primary chick embryo cells, while at the Rockefeller Institute in 1942,
where he worked with Richard Shope.
Chen-Hsiang Huang (~1914-1988),
Huang CH. Further studies on the titration and neutralization of the Western
Frederick A. Murphy, strain of equine encephalomyelitis virus in tissue culture. J Exp Med.
Robert E. Shope (1929-2004) 1943;78:111-126.
WHO Regional Office-New Delhi, 1980

page 201

1942 Chen-Hsiang Huang development of the quantitative virus neutralization assay


CDC, Savannah, Georgia, 1946

CDC, Virus & Rickettsia Section


Montgomery, Alabama, 1950s
Joseph W. Mountin (1891-1952) Left to Right: Alan Eschenbreriner, Alan Bernstein,
Matt Bucca, Roy Chamberlain, ?, ?, ?, ?, Brownie
Yarbrough, James Paine, Rachel Gorrie, ?, Robert
The U.S. Centers for Disease Control and Prevention Kissling, Morris Schaeffer, Don Nelson, Dorothy
originated as the Office of National Defense Malaria Control Reese, Gerald Taylor, Lila Pope, Sy Kalter,
Activities (1942); it then became the Office of Malaria Control W. Daniel Sudia. CDC, Downtown Atlanta, Georgia, 1952
in War Areas (1942); then later, the Communicable Disease
Center (1946); then the National Center for Disease Control Etheridge EW. Sentinel for health: a history of the Centers for Disease Control. Berkeley:
(1967); then the Center for Disease Control (1970); then the University of California Press; 1992.
Centers for Disease Control (1980); and finally the Centers for
Historical perspectives: history of CDC. MMWR Morb Mortal Wkly Rep. 1996;45;526-530.
Disease Control and Prevention (1992) but, since 1946 it has
retained its initials, CDC. 60 Years of public health science at CDC. Morb Mortal Wkly Rep. 2006;55(Sup2);1-24.
page 202

1942 Joseph Mountin, others founding of the U.S. Centers for Disease Control and Prevention
CDC, 1960, view from the front of the campus CDC, 1965, view from the back of the campus
Virology building (Bldg 7) at back-right Virology building (Bldg 7) at left, Alex Langmuir at front

page 203 CDC, 1988, Virology building (Bldg 15) at top-left CDC, 1988, Virology building (Bldg 15)

1942 Joseph Mountin, others founding of the U.S. Centers for Disease Control and Prevention
(left) Mumps virus, rhesus macaque parotid gland, frozen section immunofluorescence,
Coons & colleagues, 1949 (colorized); (right) Cell labeled with five fluorophores, 2006,
from The fluorescent toolbox for assessing protein location and function. Giepmans BN,
Adams SR, Ellisman MH, Tsien RY. Science 2006;312:217.
Coons AH, Creech HJ, Jones RN, Berliner G. The demonstration of pneumococcal antigen in
tissues by the use of fluorescent antibody. J Immunology 1942;45:159-170.
Coons AH, Kaplan MH. Localization of antigen in tissue cells. II. Improvements in a method
Albert Hewett Coons (1912-1978) for the detection of antigen by means of fluorescent antibody. J Exp Med. 1950;91:1-13.
Albert Coons initiated a major revolution in immunology cell biology and virology that continues to this day he developed the immunofluorescent technique for labeling specific
antibodies with fluorescent dyes, thus permitting the detection of antibodies and antigens in cells and tissues. His methods are also the basis for all immunohistochemical methods
now in such wide use. The development of immunofluorescence in the early 1940s was a technical tour de force, taking several years to accomplish after he had the casual idea that
putting a visible label on antibody molecules would provide a valuable research tool. With the idea in mind, Coons received encouragement from John Enders and Hans Zinsser,
among others, and expert assistance from a number of researchers including Hugh Creech and Norman Jones, who were working on the conjugation of isocyanates to proteins.
After false starts with other molecules, Coons obtained from Louis Fieser and Ernst Berliner fluorescein isocyanate, which fluoresces with a brilliant apple green color not seen in
normal tissues. Coons had further good luck Allan Grafflin was just assembling a fluorescence microscope, not a common instrument at the time. Coons labeled antibody to a
pneumococcal polysaccaride and in phagocytic cells of mice injected intravenously with the same pneumococcus he detected large numbers of fluorescing bacteria. He published
two papers, but then the work was interrupted by World War II. After the war, he had to synthesize the fluorescent compounds himself and solve many other technical problems,
the most troublesome being background tissue fluorescence in the frozen sections of tissues from animals infected with various bacteria and viruses (solved using acetone-dried
tissue extracts as an absorbing agent). The papers reporting this work had great impact, most notably on virologists at a time when pathogenesis research was booming.
page 204

1942 Albert Coons development of immunofluorescence labeling methods


A one-million liter sphere at Fort
Detrick, called the Eight-Ball, is the
largest aerobiology chamber ever built.
The sphere is on the National Register
of Historic Places it has not been used
since 1969.

Camp Detrick, later Fort Detrick, the U.S. Army installation in Frederick, Maryland, was from 1943 to 1969 the home of the U.S. Army Biological Warfare Laboratories.
During World War II, it became the site of intensive biological warfare research using various pathogens, including many viruses. The work was cloaked in the deepest
wartime secrecy, matched only by the Manhattan Project. For example, 5,000 bombs containing anthrax spores were produced there during the war. In 1969, the United
States ratified the 1925 Geneva Protocol prohibiting the use of chemical and biological weapons, and President Richard Nixon issued an executive order outlawing offensive
biological warfare research. At the same time the U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID) was formed to carry out a defensive mission. From
then on work has focused on diagnostics, prevention and therapeutics against infectious agents used in biowarfare and bioterrorism.
page 205

1943 U.S. Army, Camp Detrick development of military high-containment virology laboratories
Kenneth C. Smithburn Alexander John Haddow
(1904-1973) (1912-1978)
Smithburn KC, Haddow AJ. Semliki Forest virus. I. Isolation and pathogenic
properties. Journal of Immunology 1944;49:141-157. [From the Yellow Fever
Research Institute, Entebbe, Uganda]
Kenneth Smithburn (abridged): Between 1937 and 1948 a number of viruses
were encountered by staff members of the International Health Division of The
Rockefeller Foundation and their colleagues during long-term investigations of
yellow fever in West Africa... No systematic studies were possible until 1949,
when a comprehensive study of them was undertaken by means of a complete
series of cross-neutralization tests. The results of the study indicate that 11 of the
agents are hitherto unknown viral entities [this includes Semliki Forest virus]. All
of the agents were discovered, so to speak, accidentally by virtue of the fact that
the methods employed in the isolation of yellow fever virus are effective also for
other viral entities which are neurotropic for Swiss mice [this is] without intent
to imply that they are necessarily neurotropic in their natural hosts, whatever
these may be.

Semliki Forest virus, purified virus, thin section of ultracentrifuge pellet, by Carl von Bonsdorff page 206

1944 Kenneth Smithburn, Alexander Haddow discovery of Semliki Forest virus


Colin Munro MacLeod Maclyn McCarty
(1909-1972) (1911-2005)
Rough avirulent type II pneumococcal
colonies (left) and Smooth virulent type III
colonies (right).
Avery OT, MacLeod CM, McCarty M. Studies
on the chemical nature of the substance inducing
transformation of pneumococcal types. Induction
of transformation by a desoxyribonucleic acid
Oswald Theodore Avery (1877-1955) fraction isolated from pneumococcus type III.
J Exp Med. 1944;79:137-158.
Fred Griffiths 1928 experiment, cited earlier in this volume, described the discovery of bacterial transformation, whereby a bacterium (Streptococcus pneumoniae) was made
to change its form and pathogenicity. The phenomenon was attributed to an unidentified transforming factor. After years of work, in 1944, Oswald Avery, Colin MacLeod,
and Maclyn McCarty of the Rockefeller Institute identified the factor as DNA. They found that they could induce transformation not only with killed bacteria, but also with
extracts of bacterial cells. Treating the extract with enzymes to destroy polysaccharides (including the polysaccharide of the bacterial capsule) had no effect; neither did a lipase
to destroy bacterial lipids, proteases to destroy bacterial proteins, and RNase to destroy bacterial RNA. But, treating the extracts with DNase to destroy bacterial DNA did
abolish the transforming activity. Exhaustive studies confirmed that only DNA was capable of transforming bacteria from one type (type II rough avirulent) to another (type
III smooth virulent). The paper was met with skepticism it was a departure from the prevailing belief that the protein content of chromosomes probably made up genes. It
took another decade, and the discovery by Watson and Crick, for DNA to be widely accepted as the stuff of inheritance.
page 207

1944 Oswald Avery, Colin MacLeod, Maclyn McCarty identification of DNA as the material of inheritance
Erwin Schrdinger was an Austrian theoretical
physicist who was one of the fathers of
quantum mechanics he was awarded the
Nobel Prize in physics in 1933. In 1939, he
was dismissed from his university position in
Germany for political unreliability. He ended
up in Dublin, where in addition to continuing
his career in physics he wrote the book, What
is Life? This was in 1944, at a time when DNA
was not yet accepted as the stuff of heredity,
but a time when the concept of complex
molecules containing the genetic code for
living organisms was being discussed in
scholarly circles. Schrdinger assumed that the
gene is a one-dimensional crystal, which lacks
a periodic repeat, i.e., an aperiodic crystal.
Not being aware of Avery, MacLeod and
McCartys proof that the gene is composed of
DNA (the book was based upon lectures given
in 1943), Schrdinger believed that the gene
was composed of protein. He concluded, We
are faced with a mechanism entirely different
from the probabilistic one of physics, one
that cannot be reduced to the ordinary laws
of physics... Living matter, while not eluding
the laws of physics ... is likely to involve other
laws of physics hitherto unknown... The book
received many uncomplimentary reviews,
but nevertheless became quite popular and
influential. Eventually, the book was translated
into seven languages, and total sales exceeded
well over 100,000. The molecular virologist, Schrdinger E. What is life? Cambridge:
Gunther Stent, later stated that the book Cambridge University Press; 1944.
Erwin Schrdinger (1887-1961) probably had little influence on professional
biologists, but it had a great impact on
physical scientists, who were only too happy
to focus their intellect on a new and refreshing
problem in the post-war years when physics
was seen only as contributing to the specter
of nuclear war. Many of the first molecular
virologists came to biology from backgrounds
in physics, influenced by Schrdingers book
Max Delbrck, James Watson, Francis Crick
and others cited the book in their memoirs as
inspirational and career changing.
page 208

1944 Erwin Schrdinger publication of book, What is Life? motivating many scientists toward molecular biology
Saul Krugman, a nurse and visitors, including the
NYC mayors wife, at Willowbrook State School
Much of the early work in the United States on viral hepatitis
(hepatitis A and hepatitis B) was done at the Willowbrook State
School, a notoriously overcrowded institution for mentally
disabled children in Staten Island, New York. At a time before
much was known about these infections, Saul Krugman and his
colleagues discovered that nearly all patients and staff became
infected shortly after arrival at the institution unless they had
Frederick O. MacCallum Saul Krugman been previously infected. Most infections had gone unrecognized.
( ~1912-1994) (1911-1995) They found that hepatitis A and B co-existed and many cases
In the 1940s, F. O. MacCallum realized that a number of soldiers who received yellow fever vaccine of relapse were actually infections with two different viruses.
developed hepatitis a few months later. The yellow fever vaccine contained human serum as a stabilizing Krugman and his colleagues showed that injecting patients
agent, and MacCallum was aware of other hepatitis cases reported in the medical literature that followed upon admission with immunoglobulin provided protection from
inoculation with vaccines containing human serum. He also knew of cases that followed the reuse of illness. This was an important advance, but in a contentious set
unsterilized syringes, needles and instruments that could be contaminated by human blood. MacCallum of experiments, viruses were injected and fed as well Krugman
came to suspect that a virus carried in human blood caused hepatitis. argued that the development of a vaccine would outweigh any
A series of observations of volunteers by MacCallum and others during and shortly after the war minor harm to the children. He also argued that the children were
strengthened this hypothesis and made it clear that hepatitis could also be spread by other means than bound to be infected anyway, and only children whose parents
through blood. MacCallum coined the term hepatitis A for the form of the disease that is spread primarily who gave consent were included. The Willowbrook experience
through food and water contaminated with fecal material and the term hepatitis B for the form that is contributed importantly to the public debate over research
transmitted mainly by exposure to contaminated blood. ethics and gave impetus to regulation. In the 1970s, the ethical
controversy reached medical journals and newspapers, and led
MacCallum FO, Bauer DJ. Homologous serum jaundice transmission experiments with human volunteers. to Congressional hearings. Many regulations of institutions
The Lancet 1944;1:13:622-627. [Virus Reference Laboratory, Public Health Laboratory Service, Colindale, like Willowbrook were instituted at state and national levels in
London, UK] succeeding years.
page 209

1944-1958 Frederick MacCallum, Saul Krugman, others separation of two forms of hepatitis, hepatitis A and B
Max Ludwig Henning Delbrck Salvador Edward Luria Gunther Stent
(1906-1981) (1912-1991) (1924-2008)
In 1945, Max Delbrck initiated the first course
at the Cold Spring Harbor Laboratory the
Phage Course, which covered the fundamentals of
bacteriophage biology and methodology. It instilled
the Phage Groups distinctive math- and physics-
oriented approach to biology and played a seminal
role in the global development of molecular biology
and molecular genetics. Faculty and participants
in the first years included Salvador Luria, Alfred
Hershey, Seymour Benzer, Gunther Stent, James
Watson, Frank Stahl, Renato Dulbecco, and other
young scientists who went on to build the science
we know today. The course has continued for more
Franois Jacob, Max Delbrck, Matthew Meselson, Salvado Luria Alfred Hershey than 60 years, now under the title, the Molecular
Ronald Rolfe,Gunther Stent and Sydney Brenner, 1960 and Max Delbrck and Seymour Benzer
Genetics of Bacteria & Phages.
page 210

1945> Max Delbrck, Salvador Luria, others founding of the Phage Course at Cold Spring Harbor Laboratory
Thomas Francis, Jr. George K. Hirst Frederick Davenport Gordon Meiklejohn Edwin Kilbourne
(1900-1969) (1910-1994) (1914-1982) (1911-1997) (1921-2011)

The first influenza vaccine was developed by Thomas Francis, Jr., working at the Rockefeller Institute in 1933, just as Smith, Andrewes and Laidlaw developed the ferret
model of influenza Francis was the first American to isolate the virus (by instillation of ultrafiltered respiratory secretions from patients intranasally in ferrets). In 1937, he
first attempted to vaccinate humans using a virus grown in minced chick embryo cultures. Francis was appointed director of the Commission on Influenza of the U.S. Army
Epidemiological Board in 1941, from where he conducted what are now called phase one clinical trials, involving volunteers including conscientious objectors. The trials,
which would be considered unethical today, proceeded to a successful double-blinded field trial in soldiers (12,000 subjects). By 1943, the method of vaccine production
evolved into that which is still used today: vaccine virus is inoculated into the allantoic cavity of embryonating eggs, incubated, harvested, inactivated using formalin or
b-propriolactone, and purified. By 1945, all 7 million U.S. military personnel were immunized and vaccine was made available to the public. The vaccine of the 1940s was
monovalent; in the 1960-70s vaccines were bivalent; since 1978 they have been trivalent, containing one strain of influenza B and two of influenza A virus.
page 211

1945> Thomas Francis, George Hirst, Fred Davenport, others development of influenza vaccines
Crimean-Congo hemorrhagic fever, human liver,
massive focal necrosis and hemorrhage. H&E

Mikhail Petrovich Chumakov Ghislain F. G. J. Courtois


(1909-1993) (1912-1971)
Crimean hemorrhagic fever was first observed by Russian scientists in 1944-1945 when about 200 Soviet
military personnel were infected during an epidemic in the Crimea. At that time it was established by
Mikhail Chumakov in studies in human volunteers that the etiological agent was ultrafilterable and
transmitted by the tick Hyalomma marginatum. The virus was not maintained in the laboratory. Congo
virus was first isolated in 1956 in Zaire by Ghislain Courtois and his colleagues from the blood of a febrile
patient. The virus was isolated using newborn mice and with difficulty was serially passaged. Jordi Casals
later showed that the viruses isolated in the Crimea and in Zaire were antigenically indistinguishable
hence, the hyphenated name. Since then, the virus has been shown to have an extremely wide geographic
distribution and is a significant cause of severe disease and substantial mortality.
Chumakov MP, Butenko AM, Shalunova NV, Martianova LI, Smirnova SE, Bashkirtsev IuN, Zavodova TI,
Rubin SG, Tkachenko EA, Karmysheva VIa, Reingold VN, Popov GV, Savinov AP, New data on the viral
agent of Crimean hemorrhagic fever. Vopr Virusol. 1968;13: 377 (in Russian).
Woodall JP, Williams MC, Simpson DI. Congo virus: a hitherto undescribed virus occurring in Africa. II.
Identification studies. East Afr Med J. 1967;44:93-98.
page 212

1945> Mikhail Chumakov, Ghislain Courtois, colleagues discovery of Crimean-Congo hemorrhagic fever virus
Joshua Lederberg (1925-2008) in his lab, 1958 Edward Lawrie Tatum (1909-1975)

1958 Nobel Prize


in Physiology or
Medicine

Plaque at Yale University


page 213

1946 Joshua Lederberg, Edward Tatum discovery of genetic recombination in bacteria


In 1946, at Yale University Joshua Lederberg and Edward Tatum carried out a series it. I was scared. I knew it was a very important finding, but I didnt want to be out
of experiments demonstrating genetic recombination in bacteria for this in 1958 on a limb until I could be absolutely certain and I didnt want to commit myself
they shared the Nobel Prize in Physiology or Medicine. Joshua Lederberg was a genius emotionally to the consequence until then. So it was a matter of going back to the
(at 33 he was the second youngest Nobel laureate ever), but he also was the most grindstone and repeating the experiment many times, doing it different ways and
wonderful writer; the following are quotes clipped from his autobiographical pieces: being sure it was a totally reliable and reproducible result. Put in every control that I
could think of to be sure it had controlled for possible artifacts. Happily, with bacteria
The important biological discovery of 1944 was the identification by Avery, one can do these experiments, you can run two or one or two cycles a day in this kind
MacLeod & McCarty, at the Rockefeller Institute, of the substance responsible for of experimentation so within a few weeks it was possible to get total validation.
pneumococcal transformation. This phenomenon, which Fred Griffith had stumbled
on in 1928, appeared to be the transmission of a gene from one bacterial cell to The experiment was as follows: two strains of Escherichia coli with different
another... Sleepwalking, we were all groping to discover just what was important nutritional requirements were used. Strain A would grow on a minimal medium only
about the chemical basis of this biological specificity. My immediate private response if the medium was supplemented with methionine and biotin; strain B would grow on
to reading the 1944 paper was that the research was unlimited in its implications a minimal medium only if it was supplemented with threonine, leucine, and thiamine.
What could be done to incorporate this dramatic finding into the mainstream Each strain of bacteria was plated into dishes containing only unsupplemented
of biological research; how could one further advance these new hints about the minimal medium. Some of the dishes were plated only with strain A bacteria, some
chemistry of the gene?... It is difficult to find a clear instance of a scientific revolution only with strain B bacteria, and some with a mixture of strain A and strain B bacteria
in the history of biology, in the strict sense of a paradigm shift barely coupled to that had been incubated together for several hours in a liquid medium containing
experimental evidence The Darwinian revolution comes very close, especially in all the supplements. No colonies arose on plates containing either strain A or strain
its application to microbiology My work of 1946-1947 became my dissertation I B alone, showing that back mutations cannot restore prototrophy. However, the
spent the summer of 1947 at Woods Hole (and the magnificent library of the Marine plates that received the mixture of the two strains produced colonies at a frequency
Biological Laboratory), completing my dissertation. The stacks gave me a wonderful of 1 in every 10,000,000 bacteria plated (i.e., 1x107). This observation showed that
opportunity to explore the history of microbiology: how its pioneers had sought to recombination of genes had taken place between the genomes of the two strains to
cope with the perplexities of bacterial variability, totally isolated from the intellectual produce prototrophs.
apparatus of modem genetics.
Between 1946 and 1952, Lederberg and his lab group, then at the University of
My first important discovery came about through a rather unusual process. More Wisconsin, significantly reshaped the field of bacterial genetics. By showing that
than almost everything else that Ive ever done, it was theory driven rather than data certain strains of bacteria reproduce by mating that is, by recombining their
driven. It was theory driven in the sense that a postulate had arisen out of the course genetic material, he overturned prevailing assumptions that bacteria were primitive
of examination of the contemporary scene, namely Avery et al., had shown transfer organisms not suitable for genetic analysis. Rather, he demonstrated that bacteria
of heritable characteristics in a bacterium. One was deeply motivated to want to could serve as a powerful experimental system, with widespread application in genetic
know more about whether there were things like genes in bacteria. The approach research. He also entered the world of virology, making seminal discoveries such as
that speculatively arose and how to respond to it is: could one find out that there is a transduction, using bacteriophage.
possibility of genetic recombination, of crossing between bacterial cells?... One was
able to put together a thought experiment that said, If bacteria can be crossed, and In 1950, Bernard Davis (1916-1994) showed that the donor and recipient bacteria
if you start out with two different nutritional mutants, and if they exchange with one involved in genetic recombination must be in contact, and in 1952 William Hayes
another, they will form prototrophic [wild type, able to grow on unsupplemented (1918-1994) showed that the mechanism of transfer of genes between bacteria is quite
minimal medium] genotypes, you will be able to select for them, and define their complex, unidirectional and employs a unique filamentous mating structure.
occurrence even if they happen very, very rarely [this could be done] by a rather Lederberg J, Tatum EL. Novel genotypes in mixed cultures of biochemical mutants of
simple procedure plating the mixtures on minimal agar. So here was a case where bacteria. Cold Spring Harbor Symp Quant Biol. 1946;11:113-114.
the experimental design was worked out [in theory] it was a theoretical postulate: Lederberg J, Tatum EL. Gene recombination in Escherichia coli. Nature 1946;158:558.
does genetic recombination occur? Does it not occur? And that was all worked out in
advance of ever doing the experiment. Lederberg J. Genetic recombination in bacteria: a discovery account. Annu Rev
Genet. 1987;21:23-46.
Well somewhat to my surprise it worked very, very promptly. I was rather fearful Lederberg J. Forty years of genetic recombination in bacteria. A fortieth anniversary
when the first positive results came in. I was worried deeply that this was going to reminiscence. Nature 1986;324:627-628.
be an artifact, that my hopes would be dashed. I didnt want to get too excited about
page 214

1946 Joshua Lederberg, Edward Tatum discovery of genetic recombination in bacteria


Max Ludwig Henning Delbrck Alfred Day Hershey
(1906-1981) (1908-1997)
[with the same model fraction collector used by the
author in his thesis research at the University of
California Davis]
In 1946, Alfred Hershey demonstrated the independence of different mutation types occurring in the same bacteriophage: this was the first indication that a virus may contain
more than one gene. In the same year, Max Delbrck discovered an unexpected genetic interaction between viruses infecting the same cell. He and Hershey independently
advanced this work and demonstrated that the phenomenon was due to genetic recombination and that it could be used to construct the genetic map of the bacteriophage.
This finding led, about 10 years later, to the complete mapping of the T2 bacteriophage genome by Seymour Benzer. Salvador Luria was able to support Delbrck and Hersheys
interpretation of their findings as showing genetic recombination with experiments in bacteriophages in which genomic damage caused by radiation could be repaired by gene
exchange following infection of the same host bacterium with several damaged bacteriophage particles. These findings opened tremendous possibilities for analysis of the
structure of the genetic material, DNA.
Delbrck M, Bailey WT Jr. Induced mutations in bacterial viruses. Cold Spring Harb Symp Quant Biol. 1946;11:33-37.
Hershey AD, Rotman R. Genetic recombination between host range and plaque-type mutants of bacteria in single bacterial cells. Genetics 1949;34:44-71.

page 215

1946 Max Delbrck, Alfred Hershey discovery of genetic recombination in viruses (bacteriophage)
Smithburn KC, Haddow AJ, Mahaffy AF. A neurotropic virus
isolated from Aedes mosquitoes caught in the Semliki forest. Am J
Trop Med. 1946;26:189-208.
The virus was isolated in September 1943 from a large lot of Aedes
mosquitoes caught in an area in the Semliki Forest [Uganda], known
as Bunyamwera, hence the name provisionally adopted, Bunyamwera
virus. In recent years a considerable number of previously
unknown filterable viruses have been discovered, many of them
being neurotropic in action. These include the viruses of eastern,
western and Venezuelan equine encephalomyelitis; lymphocytic
choriomeningitis; St. Louis, Japanese B and Russian spring-summer
encephalitis; and louping ill. Also, in this laboratory, and quite
incidental to our main problem (yellow fever), we have encountered
four hitherto unknown viruses and have found humoral immunity
to each to occur in man. Some of these agents have been isolated
from man or animals, others from insects and a few from both.
Some have been discovered during the course of investigations of
specific diseases in man or animals, while others were encountered
accidentally, so to speak. Some are related antigenically, others are
not known to be. The significance of at least three of the agents in
human and/or veterinary medicine is not yet known. All of these
facts indicate that a concerted attack on the problem of virus etiology
of disease (including pyrexias of unknown origin) might not only
increase our knowledge of the known viruses and their properties
but also uncover further viruses at present unknown as the causative
agents of disease.

Kenneth C. Smithburn Alexander John Haddow


(1904-1973) (1912-1978)
Uganda Virus Research Institute
(UVRI), Entebbe, Uganda
The UVRI was established in 1936 as
the Yellow Fever Research Institute by
the Rockefeller Foundation. In 1977,
it became a Ugandan government
public health research institution.
The Institute has a grand record in
virus discovery; more than 20 new
arboviruses were discovered by its
staff members, including West Nile
virus, Semliki Forest virus, and here,
Bunyamwera virus.
Alexander Francis Mahaffy (1891-1952)
Yellow fever vaccination campaign, Uganda, 1944 page 216

1946 Kenneth Smithburn, Alexander Haddow, Alexander Mahaffy discovery of Bunyamwera virus
Lloyd Florio (1911-1976)
Florio L, Stewart MO, Mugrage ER. The etiology of Colorado tick fever.
J Exp Med. 1946;83:1-10. [Department of Public Health and Laboratory
Diagnosis, University of Colorado School of Medicine]
Florio and his colleagues reported the transmission of the etiologic
agent of Colorado tick fever to Syrian hamsters and human volunteers
after ultrafiltration through a series of Gradocol (collodion)
membranes, thereby showing that the agent was a virus. However, even
though the experiments were repeated and controls were satisfactory,
the findings were not cut-and-dried and their interpretation of their Colorado tick fever virus: (top) BHK-21 cell culture, thin section electron
findings did not predict the virion size later determined by others. microscopy; (bottom) negative contrast electron microscopy.
page 217

1946 Lloyd Florio, Mabel Miller, Edward Mugrage discovery of Colorado tick fever virus (the first coltivirus)
Frank Macfarlane Burnet (1899-1985) Alfred Gottschalk (1894-1973) Ernst Klenk (1896-1971)

The interaction of viruses with cellular receptors initiates a chain of dynamic events that enables entry of the virus into the cell. This is a multistep process, usually
with multiple attachment receptors used sequentially with or without co-receptors. As more and more research has been done on this subject, it has gotten more and
more complex, with less and less merit in generalizations. Extrapolation from cell culture studies, to in vivo studies, to natural host studies, has further confounded our
understanding. Nevertheless, it is virus+receptor interactions that provide keys to our understanding of virus tissue tropism, host range, disease pathogenesis and the antiviral
immune response (protective or immunopathogenic). Since the discovery of the first cellular receptors for viruses, so many investigators have made so many discoveries that
this subject cannot be covered properly here. Schneider-Schaulies J. Cellular receptors for viruses: links to tropism and pathogenesis. J Gen Virol. 2000;81:1413-1429. page 218

1947> Frank Macfarlane Burnet, Alfred Gottschalk, Ernst Klenk discovery of viral receptors on target cells
Comparison of readouts from (a) Schlieren, (b) interference, (c)
absorbance, and (d) photoelectric optical systems. Schachman H.
Ultracentrifugation in Biochemistry. Academic Press, New York, 1959

Spinco Model E Analytical Ultracentrifuge 1947, Spinco (now Beckman Coulter)


This was the first commercial analytical ultracentrifuge

Jerome Vinograd (1913-1976),


a pioneer in the use of analytical
ultracentrifugation in virology, with
his Model E, Caltech, at the time of the
Meselson-Stahl experiments, 1956

Analytic ultracentrifugation
of foot-and-mouth disease
virus, and as controls, X174
bacteriophage and southern
bean mosaic virus,
Schlieren optics.
page 219

1947 Arnold Beckman development of the Spinco Model E analytical ultracentrifuge


Mettler Analytical Balance
Model H5, Mettler Instruments AG
Only someone who had prepared cell
culture media from scratch, weighing
out the many ingredients on a two-pan
beam balance, could fully appreciate the
advance in laboratory virology brought
about by the Mettler single-pan balance.
page 220

1947 Mettler Instruments AG development of the first single-pan precision analytical balance
Frank John Fenner (1914-2010)
John Curtin School of Medical Research, Canberra

Left: Growth curves of ectromelia virus in foot, spleen, blood,


and skin of mice inoculated in the foot with a small dose
of Moscow strain of ectromelia virus. Development and
disappearance of primary lesion and rash are shown, as is the
occurrence of inclusion bodies in skin in sections stained with
Manns stain.
Right: The original classic figure from Fenners 1948 paper in The Lancet
the first quantitative virus pathogenesis study, the model for many
that followed the figure has been copied and redrawn many times,
appearing in most virology and experimental pathology textbooks.
page 221

1948 Frank Fenner founding of viral disease pathogenesis research (ectromelia virus infection in mice)
Gilbert Dalldorf graduated from New York
University and Bellevue Hospital Medical
School in 1924, and trained as a pathologist
in Freiburg and at the New York Hospital.
He contracted tuberculosis, probably in the
morgue, and poliomyelitis, probably from
his children. These infections influenced his
research in following years. In the summer of
1947, there were outbreaks of poliomyelitis
in upstate New York. Dalldorf, then director
of the New York State Department of Health
Wadsworth Laboratory in Albany, and his
associate Grace Mary Sickles (1898-1959)
investigated these outbreaks, in particular
seeking polioviruses that would replicate in
mice. This was motivated by having heard
that suckling mice could be infected with
Theilers virus (a parvovirus). Dalldorf and
Sickles made fecal suspensions from two
children suspected of having poliomyelitis,
and inoculated these into suckling mice the
mice developed paralysis caused by widespread
necrosis of skeletal muscles, not by the central
nervous system lesions seen with polioviruses.
This was the first instance of isolation of new
viruses using suckling mice. The viruses were
named Coxsackieviruses, after the New York
village of the same name. In subsequent years
many different Coxsackieviruses, divided
into groups A or B depending upon their
pathology in suckling mice, were isolated and
Gilbert Dalldorf (1900-1979) associated with a variety of clinical syndromes: Coxsackievirus A4, mouse hind limb
In general, group A Coxsackieviruses infect muscle, thin section electron microscopy
the skin and mucous membranes, causing from: Harrison A, Murphy FA, Gary GW. Ultrastructural
herpangina, acute hemorrhagic conjunctivitis, pathology of Coxsackie A4 virus infection of mouse striated
and hand-foot-and-mouth disease. Group muscle. Exp Mol Pathol. 1971;14:30-42.
B viruses infect the heart, pleura, pancreas,
and liver, causing pleurodynia, myocarditis,
pericarditis, and hepatitis. Both group A and
group B Coxsackieviruses cause nonspecific
febrile illnesses, rashes, upper respiratory tract Dalldorf G, Sickles GM. An unidentified, filterable agent
disease, and aseptic meningitis. isolated from the feces of children with paralysis. Science
1948;108:61-62.

Coxsackievirus structural model


page 222

1948 Gilbert Dalldorf, Grace Sickles discovery of Coxsackieviruses


Thomas Milton Rivers Frank Lappin Horsfall Jr. Igor Tamm
(1888-1962) (1906-1971) (1922-1995)

Viral and Rickettsial Infections of Man


First Edition: Thomas M. Rivers, Editor. 1948.
Second Edition: Thomas M. Rivers, Editor. 1953.
Third Edition: Thomas M. Rivers and Frank L. Horsfall, Editors. 1959.
Fourth Edition: Frank L. Horsfall and Igor Tamm, Editors. 1965.

First edition: Twenty-seven chapters/contributors, including General Aspects of Viral and Rickettsial
Infections, by Thomas M. Rivers; Physical and Chemical Procedures, by W. M. Stanley and Max A.
Lauffer; Serologic Reactions in Viral and Rickettsial Infections, by Joseph E. Smadel; Chick Embryo
Techniques, by E. W. Goodpasture and G. John Buddingh; Propagation of Viruses and Rickettsiae
in Tissue Cultures, by John F. Enders; Epidemiology, by Kenneth F. Maxey; and Bacterial Viruses
(Bacteriophages), by A. D. Hershey and J. Bronfenbrenner.
page 223

1948-1965 Thomas Rivers, Frank Horsfall, Igor Tamm publication of book, Viral and Rickettsial Infections of Man
Gueh-djen (Edith) Hsiung Frances Whitman Doane Pekka Halonen
(1918-2006) (1928-2001) (1927-2001)

Phillip Gardner Kenneth McIntosh Caroline Breese Hall


(1925-1994) (1939-2012)
page 224

1948> Gueh-djen Hsiung, Phillip Gardner, Pekka Halonen, others broad application of viral disease diagnostics
Spinco Model L Ultracentrifuge
1949-1950

The first commercial preparative ultracentrifuge


Maximum speed: 40,000rpm, ~275,000g

The vacuum ultracentrifuge was invented by Edward Greydon Pickels. His was
the contribution of the vacuum system, which allowed a reduction in friction
on the rotor generated at high speeds. The vacuum system also enabled the
maintenance of constant temperature. In 1940, interest in the isolation of viruses
brought Pickels and Johannes Bauer of the International Health Division of the
Rockefeller Institute together to build the first high-speed air-driven vacuum
centrifuge suitable for the study of filterable viruses the first virus they worked
with was yellow fever virus. Later, Pickels went on to develop the more convenient,
electrically driven ultracentrifuge.
In 1946, Pickels co-founded Spinco (Specialized Instruments Corp.) and marketed
an air-driven ultracentrifuge based on his design. Pickels, however, considered his
design to be too complicated and developed a more foolproof version. But even
with the enhanced design, sales of his centrifuge remained low, and Spinco almost
went bankrupt. In 1949, Spinco introduced the Model L, the first preparative
ultracentrifuge to reach a speed of 40,000 rpm. In 1954, Beckman Instruments
(now Beckman Coulter) purchased the company, forming the basis of its Spinco
centrifuge division. Arnold Beckman immediately went to work making major
improvements in the instrument it became an incredibly rugged workhorse,
such that by the 1960s every virology laboratory had at least one Model L
running night and day.
Pickels EG, Bauer JH. Ultracentrifugation studies of yellow fever virus. J Exp Med.
1940;71:703-717. [From the Laboratories of the International Health Division of
The Rockefeller Foundation]

page 225

1949 Edward Pickels development of the Spinco Model L preparative ultracentrifuge


John Franklin Enders Thomas Huckle Weller Frederick Chapman Robbins
(1897-1985) (1915-2008) (1916-2003)
Nobel Prize Award Ceremony, Presentation Speech, by Sven Gard, member of the Staff of Professors of the Royal Caroline Institute
(abstracted):
in 1949 there appeared from a Boston research team a paper, modest in size and wording but with a sensational content. John
Enders, director of the Childrens Hospitals Research Laboratory and his associates Thomas Weller and Frederick Robbins reported
the successful cultivation of the poliomyelitis virus in test-tube cultures of human tissues. A new epoch in the history of virus
research had started. In their first experiments they used human embryonic tissue. To the great surprise of everybody except
perhaps the experimenters themselves they registered a hit in their first attempt. The virus grew not only in brain tissue but equally
well in cells derived from skin, muscle, and intestines Typical changes appeared in the cellular structure, finally leading to complete
destruction, easily recognizable under the microscope a convenient method of reading the results applicable also in immunity
tests. The virologists finally had a tool in the same class as the culture technique of the bacteriologists These discoveries incited a
restless activity in the virus laboratories the world over. The tissue-culture technique was rapidly made one of the standard methods
of medical virus research, among which it now holds an undisputed first place Weller has succeeded in cultivating the agents
causing varicella and herpes zoster, Enders that of measles, viruses previously almost inaccessible for study. The method has also been
successfully applied to several problems in the field of veterinary medicine Much effort and a considerable time will be required
for equivalent achievements in the fight against the virus diseases as has been the case with the bacteria diseases. However, thanks to
Enders, Weller and Robbins discovery we may look with confidence to the future.
Thomas Weller, Frederick Robbins and
John Enders at the Nobel Prize ceremony, 1954
page 226

1949 John Enders, Thomas Weller, Frederick Robbins development of cell culture methodology for polio, measles
In 1949, Andr Lwoff started to work on lysogeny. He had fixed his choice on a lysogenic Bacillus
megaterium, a particularly large bacterium, because he wanted to study single bacteria. With a
microscope and a micromanipulator he inoculated bacteria into individual microdrops and let
them grow. Why this methodological choice? Because, as he said, he: disliked mathematics, and
wanted to avoid formulas, statistical analysis and, more generally, calculations as much as possible.
By following the kinetics of phage production, he realized that only a relatively small fraction of
the bacteria lysed and produced bacteriophage. Why did some lyse, while others did not? With
Louis Siminovitch and Niels Kjeldgaard he tried, day after day, to obtain lysis of the entire bacterial
population in a culture vessel. After several unsuccessful months, he decided to irradiate the
bacteria with ultraviolet light. A UV lamp, used by Jacques Monod to mutagenize Escherichia coli,
was available. A bacterial culture was irradiated for a few seconds, and after an hour the culture was
entirely lysed. Lwoff wrote: As far as I can remember, this was the greatest thrill of my scientific
careerfor the first time in my life, I had the feeling of having discovered something. This was a
far-ranging discovery. First of all, a clear picture of lysogeny emerged: with the bacteriophage in a
Andr Lwoff (1902-1994) lysogenic state, the bacterium perpetuates the genome of the bacteriophage (named by Lwoff the
prophage); induction of the prophage leads to its vegetative multiplication and lysis of the host
bacterium. The subsequent discovery of induction by Franois Jacob and Elie Wollman, and the
uncovering of the mechanism of genetic repression by Jacob and Jacques Monod extended this line
of research to the eventual award of Nobel Prizes to Lwoff, Jacob and Monod.
Lwoff A. The specific effectors of viral development (The First Keilin Memorial Lecture). Biochem J.
1965;96:289-301.
page 227

1949 Andr Lwoff, Louis Siminovitch, Niels Kjeldgaard discovery of lysogeny and induction (bacteriophage)
In 1950, Erwin Chargaff discovered two rules (Chargaff s Rules) that helped lead to the discovery of the
double helix structure of DNA: (1) That in natural DNA the number of guanine molecules equals the
number of cytosine molecules and the number of adenine molecules equals the number of thymine
molecules. This strongly hinted of the base-pairing structure of DNA, although Chargaff did not make
this connection himself. This work is credited with disproving the tetranucleotide hypothesis that
was widely accepted at the time. Chargaff met Crick and Watson at Cambridge in 1952 and explained
his findings to them a seminal meeting, indeed. (2) That the composition of DNA varies from
one species to another, in particular in the relative amounts of A, G, T, and C bases. Such evidence
of molecular diversity, which had been presumed absent from DNA, made DNA a more credible
candidate for the genetic material than protein.
Chargaff E. Chemical specificity of nucleic acids and mechanism of their enzymic degradation.
Erwin Chargaff (1905-2002) Experientia 1950;6:201-209.
Chargaff E, Lipshitz R, Green C, Hodes ME. The composition of the deoxyribonucleic acid of salmon
sperm. J Biol Chem. 1951;192:223-230.

page 228

1950 Erwin Chargaff discovery that in DNA the number of molecules of A+T equals that of G+C
From his holistic view to the
study of viral pathogenesis in
experimental animals, Cedric
Mims taught the merit of a holistic
view of the infectious diseases
per se. From the Preface of his
book: It is my conviction that the
centrally significant aspects of the
subject of infectious diseases are
the mechanisms of infection and
pathogenicity, and that the principles
are the same, whatever the infectious
agent. When we consider the entry
of pathogens into body, their spread
through tissues, the damage they
cause, their exit and spread, the role
of the immune responses, etc., the
general features are the same...

Cedric Mims
Cedric Mims illustration of the power of the frozen-section
Mims CA. Aspects of the pathogenesis of virus immunofluorescence technique (colorized). A, trachea 24 hours after the
diseases. Bacteriological Reviews. 1964;28:30-71. intranasal infection of a mouse with Sendai virus. B, Section of thymus and
Mims CA, Nash A, Stephen J. Mims Pathogenesis of nearby muscle of a newborn LCM virus carrier mouse, showing infected
Infectious Disease (Fifth Edition). London: Elsevier; thymus and neighboring intercostal muscle. C, Leg muscle of a mouse showing
2001. infected muscle cells 96 hours after infection with Ross River virus. D. Uterus
of a pregnant LCM virus carrier mouse, showing infected endometrium and
page 229 glands.

1950-1970s Cedric Mims development of immunofluorescence in viral pathogenesis research


HeLa cells
The HeLa cell line was derived from a biopsy of a cervical carcinoma suffered by Henrietta Lacks,
who died from this carcinoma in 1951. The cells were propagated by George Otto Gey, who with his
wife, Margaret, had started the Tissue Culture Laboratory at Johns Hopkins University in the 1950s.
The cells were obtained without Mrs. Lacks knowledge or permission at that time permission was
not required. The issue of ownership was later brought up in a Supreme Court of California case,
Moore v. Regents of the University of California. The court ruled that a persons discarded tissue and
cells are not their property and can be commercialized without permission. HeLa cells were used
by Jonas Salk in his polio vaccine studies in the 1950s and since then in many other kinds of studies.
They are very widely used in virology.
George Otto Gey (1899-1970)
In 2010, a book, The Immortal Life of Henrietta Lacks, by Rebecca Skloot (Crown Publishing Co.),
brought the travails of the Lacks family to public attention, becoming a best-seller and the basis for
Gey GO, Coffman WD and Kubicek MT. Tissue culture studies of the
reopening the question of legal ownership of materials like biopsies and cells and molecular products
proliferative capacity of cervical carcinoma and normal epithelium.
derived from such materials.
Cancer Res. 1952;12:264.

page 230

1951 George Gey establishment of HeLa cell line (human cervical adenocarcinoma; named for Henrietta Lacks)
More than four decades passed between the
discoveries of the first avian retroviruses by
Ellermann and Bang and Rous to the isolation
of murine leukemia virus by Ludwik Gross
(1951). Gross found that cell-free filtrates
prepared from the spontaneous leukemias in
AKR strain mice could transmit the disease to
the low leukemia C3H strain. Gross success
where everyone else had failed had three main
bases: his serendipitous use of newborn mice as
recipients; his fortuitous choice of C3H mice as
recipients (the only low leukemia strain available
that was susceptible to the virus carried by AKR
mice); and Gross dogged persistence when no
one expected positive results. The scientific
community was skeptical, unbelieving until
Jacob Furth took pains to repeat the experiments
under original conditions. The virus isolated
became Gross murine leukemia virus. During
the next two decades, many such viruses that
Ludwik Gross Charlotte Friend John B. Maloney cause neoplastic disease in mice were identified
(1904-1999) (1921-1987) (1924-2007) and became important model systems, actively
studied at the cellular and molecular levels
From Wallace P. Rowe, 1973: The history to this day. Friend (1957), Maloney (1960),
of tumor virology is one of the more Rauscher (1962), Abelson (1970) and other
stormy and interesting chapters in the murine leukemia and sarcoma viruses provided
history of 20th century science. From models for the study of many aspects of
the very beginning of tumor virology, oncogenesis.
the pioneers in this field had to go Gross L. Spontaneous leukemia developing in
through successive cycles of optimism C3H mice following inoculation in infancy, with
and disillusion as each technical and AK-leukemic extracts, or AK-embryos. Proc Soc
conceptual breakthrough failed to Exp Biol Med. 1951;76:27-32.
achieve any degree of success in dealing
with the problem of human cancer. In Gross L. Mouse leukemia. Ann N Y Acad Sci.
addition to struggling with themselves, 1952;54:1184-1196.
these pioneers had to deal with intensive Rowe WP. Genetic factors in the natural history
opposition from other parts of the of murine leukemia virus infection. Cancer
scientific community. The virus-cancer Research 1973;33:3061-3068.
question has been such a highly polarized
issue that the question Do you believe Coffin JM, Hughes SH, Varmus HE.
that viruses cause cancer? has always Retroviruses. Cold Spring Harbor: Cold Spring
been as much a test of faith as Do you Harbor Laboratory Press; 1997.
believe in God?; and if the answer was Javier, RT, Butel JS. The history of tumor
Frank J. Rauscher Jr. Herbert T. Abelson Yes, it was as likely to be greeted by a virology. Cancer Research 2008;68:7693-7706.
page 231 (1932-1993)
patronizing half-smile half-sneer.

1951> Ludwik Gross, Charlotte Friend, others discovery of murine leukemia and lymphoma viruses
Esther Miriam Zimmer Lederberg (1922-2006)

The bacteriophage (lambda) has been the most widely used bacteriophage in molecular biological
research. Esther Lederberg was the first to isolate this DNA virus, from Escherichia coli K-12, in 1951.
The genome consists of a double-stranded DNA molecule with 5 twelve-base-pair sticky ends, which
permit circularization of the DNA molecule. It shows a lytic cycle and a lysogenic cycle. Studies on the
control of these alternative cycles have been very important for our understanding of the regulation of
gene transcription. Study of over the past 50 years has also provided valuable insights into virus life
cycles, the regulation and expression of DNA, and the mechanism of integration and excision of viral
genes and genomes into host chromosomes. The mechanism of integration of DNA into bacterial
DNA was first worked out by Lederbergs colleague, Allan Campbell, in 1962. has also been used as a
Bacteriophage
Negative contrast electron microscopy
vector for the cloning of recombinant DNA.
Lederberg EM. Lysogenicity in Escherichia coli strain K-12. Microb Genetics Bull. 1950;1:5-8.
Lederberg EM, Lederberg J. Genetic studies of lysogenicity in Escherichia coli. Genetics 1953;38:51-64.

page 232

1951 Esther Lederberg discovery of the bacteriophage


Linus Carl Pauling (1901-1994) John Cowdery Kendrew (1917-1997) Max Ferdinand Perutz (1914-2002)

Studies leading
to resolution of
the structure of
hemoglobin:
Max Perutz with
his diffractometer,
the crystal
diffraction pattern
of hemoglobin 1c,
and a molecular
ribbon model of
hemoglobin 1c

page 233

1951> Linus Pauling, John Kendrew, Max Perutz discovery of the structure of proteins using X-ray crystallography
In 1944, the Avery-MacLeod-McCarty experiments demonstrated that DNA rather
than proteins is the carrier of genetic information. Though the work was well done, it
met with resistance from much of the scientific community and for the next decade
Avery was forced to repel attacks. Finally, in 1952, Alfred Hershey, at Cold Spring
Harbor Laboratory, set out to settle the issue Hershey believed that proteins, with
their complicated structures, were more likely to be the carriers of genetic information
than was the simple DNA molecule. Hershey and his technician, Martha Chase, decided
to track the transfer of proteins and DNA between a virus and its host. They chose the
bacteriophage T2 as the vehicle for delivering the genetic material to the host Escherichia
coli the phage would inject its genetic material into the bacterium, leaving its protein
shell attached to the outside of the bacterial cell wall. It was thought that certain proteins
were transferred from the virus to the bacterium upon attachment; if the genetic
information was in fact carried by these proteins, it was necessary to demonstrate that at
least a portion of it was transferred to the interior of the bacterium.
In their first experiment, Hershey and Chase tagged the phage DNA with radioactive
phosphorus-32 (32P). Because phosphorous occurs in large quantities in DNA, but in
only trace amounts in protein, the location of the phage DNA could be determined.
They then allowed the labeled phage to begin infecting samples of E. coli. After the
phage was mixed with the bacteria, they used a Waring blender to violently disturb the
infected bacteria, causing the phage protein shells to detach from their hosts. Then,
using a centrifuge, they separated the bacteria from the phage and protein. Once the
separation was complete, they measured the 32P in the bacteria and the separated
phage protein shells. The 32P appeared in large quantities only in the bacterial sample,
demonstrating that DNA was transferred from the phage to the bacteria. Further, despite
the protein shells being detached while infection of the bacteria was still ongoing, the
phage replicated in the bacteria as usual. This, in turn, suggested that the protein shell
was not necessary for the replication process following the initial insertion of the genetic
Martha Cowles Chase (1927-2003) material.
Alfred Day Hershey (1908-1997)
Shocked by their findings, Hershey and Chase decided to perform the experiment again,
[Karl Maramorosch took this famous photo]
this time using a different tracer sulfur-35 (35S), because sulfur is present in proteins, but
not in DNA. After labeling the proteins, infecting the bacteria, and separating the phage
shells from the bacteria, they tested for the presence of 35S. It could only be found in the
protein shells, not in the bacteria. And again, phage replication went on normally.
Sufficiently impressed by the significance of their findings, they examined the progeny
of the 32P-labeled phage and found that they also had 32P-labled DNA, but their protein
lacked any trace of radioactivity.
At first, Hershey and Chase were inclined to believe that their experiments were flawed,
but no faults were found. The results were no mistake and the importance of their work
was clear: they had produced direct, irrefutable, biological evidence that DNA, not
protein, is the stuff of inheritance. Later that year, they reported their findings in a short
paper. As they say, The rest is history.
Hershey AD, Chase M. Independent functions of viral protein and nucleic acid in growth
of bacteriophage. J Gen Physiol. 1952;36: 39-56.
page 234

1952 Alfred Hershey, Martha Chase Waring blender experiment proof of DNA as the material of inheritance
The Virus Laboratory, University of California Berkeley, 1952 Wendell Meredith Stanley (1904-1971)
In 1948, Wendell Stanley went to the University of California Berkeley from the Rockefeller Institute to be director of a new research organization, the Virus Laboratory (always
known as the virus lab). Stanley assumed the additional task of recruiting staff for a new Department of Biochemistry to be attached to the College of Letters and Science. In
1952, this department, as well as the Virus Laboratory, moved from temporary quarters into the new Virus Laboratory building near the East Gate. The Department of Virology
was established in 1958, in recognition of the prominent role that graduate and post-doctoral training had assumed in the Virus Laboratory. The staff, under the chairmanship
of Stanley, organized a course of study and research leading to MA and PhD degrees in virology. The department emphasized in its teaching and research the biochemical,
biophysical, and biological aspects of animal, plant, and bacterial viruses. It was the second Department of Virology founded in a major university, the first being at Johns
Hopkins University. In 1962, a broader vision of the role of virology culminated in the creation of the Department of Molecular Biology. Its initial staff numbered 16: among its
members were three Nobel Prize winners and five members of the National Academy of Sciences. In 1964, the Department of Virology was subsumed into the Department of
Molecular Biology. In 2005, the Virus Laboratory building was demolished to make room for a new biosciences and bioengineering facility.
[In 1962, the author (FAM), while a graduate student at the University of California Davis, enrolled in the basic virology course at Berkeley it was taught by Gunther Stent
and Harry Rubin and was held in the lecture room in the virus lab. It was the best course ever incredible in its scope, depth and context, demanding to an extreme, and
of great influence upon everyone who ever took it. Part of the aura was that nearby in the building were leaders in all the booming facets of virology research, including
bacteriophage research (it was said that Gunther Stent and his colleagues, along with colleagues at Caltech, represented branches of the Phage School at Cold Spring Harbor
Laboratory). Of course students taking that great basic virology course did not know all this at the time!]
page 235

1952 Wendell Stanley establishment of the Virus Laboratory at the University of California Berkeley
Between 1947 and 1955, Joshua Lederberg and his colleagues at the
University of Wisconsin described several important experimental
techniques and results which transformed the science of bacterial genetics
and helped define the classical era of molecular biology. Their most
important discoveries were that of transduction the transfer of genetic
fragments from one cell to another by a virus and of extra-chromosomal
genetic elements called plasmids. Lederberg later said: We were exploring
a completely new territory that we only dimly understood. We werent
looking for transduction we bumped into it. We werent looking for
plasmids we bumped into them. Every time we turned around we found
something unexpected.
Lederberg and the other members of his small laboratory, in particular
his wife, Esther Zimmer, and graduate students Norton Zinder and Larry
Morse, conducted a systematic search for genetic recombination in bacteria.
Streptomycin-resistant mutants of Salmonella proved especially important.
In 1951, Lederberg and Zinder discovered the third mechanism of genetic
transfer in bacteria, (the first being transformation, discovered by Oswald
Avery, the second being mating, discovered earlier by Lederberg). Lederberg
named the new phenomenon transduction. They used a glass tube, bent
into a U-shape and fitted with an extremely fine glass filter (an ultrafilter)
at its crook, and filled it with broth containing none of the nutrients their
Salmonella mutants needed to grow. They then added one mutant parent
strain to each arm, and pumped the broth back and forth through the filter.
They expected to find no recombinants, because the bacteria in Lederbergs
Joshua Lederberg (1925-2008) Norton Zinder (1928-2012) original crossing experiments had to be in direct contact with one another
for conjugation to take place. To their surprise, several bacteria nonetheless
appeared and multiplied, an indication that recombination had occurred
and had enabled each mutant strain to compensate for the nutritional
deficiencies of the other. Obviously, a biological agent small enough to
pass through the filter was at work. When they purified the material, they
found that it did not consist of pure DNA or RNA; instead, it proved to be
a bacteriophage. The Salmonella Lederberg and Zinder had used turned
out to be lysogenic: it carried phage DNA integrated into its chromosome,
but occasionally this was excised and the phage replicated. Prior to lysis,
the phage particles picked up random snippets of bacterial DNA, which
they then carried through the filter The phage acted as vectors for bacterial
genes, genes that complemented the mutual genetic (nutritional) defects of
the parent bacteria.
Zinder ND, Lederberg J. Genetic exchange in Salmonella. J Bact.
1952;64:679-699.
Morse ML, Lederberg EM, Lederberg J. Transduction in Escherichia coli
K-12. Genetics 1956;41:142-156.

page 236

1952 Joshua Lederberg, Norton Zinder discovery of transduction: DNA transfer between bacteria by bacteriophage
Karl Maramorosch Tobacco necrosis virus A, negative contrast electron microscopy
Karl Maramorosch wrote in 1953: There exists a group of viruses which constitutes a link between animal and plant viruses. These viruses were shown to multiply both in
the plants they infect and in their insect vectors. The best studied of these versatile viruses is the one that causes aster-yellows disease. The aster-yellows virus requires for its
survival in nature two alternating hosts: a susceptible plant and a susceptible insect. The host range in the plant kingdom is very wide, as species in more than 50 families have
been found susceptible. However, only one species of cicadellid leafhopper, Macrosteles fascifrons Stal, the common aster leafhopper, has been found susceptible to the virus
in the Eastern United States. This pattern of rather broad plant host specificity and narrow arthropod host specificity tuned out to be quite common. Over many years, Karl
Maramorosch led studies of the natural history of many arthropod-borne plant viruses, many of which cause economically important crop and ornamental flower diseases.
Many, as well, exhibit complex, intriguing mechanisms by which they survive and propagate in nature, including: mechanical transmission (flying pin), biological transmission
(requiring virus replication in the arthropod and delivery of virus to the feeding organs), vertical transmission (requiring infection or carriage in the egg or embryo of the
arthropod), and fomite transmission (often via carriage on farm equipment and implements). [In recent years, some plant diseases thought to be caused by viruses have been
attributed to phytoplasmas (mycoplasmas); this includes aster yellows.]
Maramorosch K. Multiplication of aster-yellows virus in its vector. Nature 1952;169:194-195.
Maramorosch K. Do developmental stages occur in the reproductive cycle of aster-yellows virus? Cold Spring Harbor Symposia in Quantitative Biology 1953;18: 51-54.
page 237

1952 Karl Maramorosch discovery that some viruses can replicate in both plants and insects
Western equine encephalitis virus,
plaques in Vero cells

Dulbecco R. Production of plaques in monolayer tissue


cultures by single particles of an animal virus. Proc Natl
Acad Sci USA. 1952;38:747-752.
Dulbecco R, Vogt M. Plaque formation and isolation
of pure lines with poliomyelitis viruses. J Exp Med.
1954;99:167-182.

Renato Dulbecco (1914-2012) Marguerite Vogt (1913-2007)


Interview with Renato Dulbecco by Shirley K. Cohen, 1998. [Caltech Oral History] (Unpublished) http://resolver.caltech.edu (section abridged): You know, accidents are
wonderful in science, provided you can take advantage of them. [In ~1950 Dulbecco, then a fellow at Caltech, was called into the office by Max Delbrck regarding some new
funding for virus research.] at the time, phage work didnt appeal to me so much. I needed something new, so I said yes Immediately I thought that to do anything in the
field, you needed tissue cultures. And I already had a background in that. So I said yes [After trips to several labs, to] learn about what people were doing, I thought of how
to develop a good system for assay of animal viruses [I told Max,] You know, the virus I need is the western equine encephalitis virus, which was known to be pathogenic.
They were very worried about that, so they decided to send me to the second basement at the end of the corridor, where there was a room by itself Immediately I was
thinking that my goal was to develop a plaque system like that of phage and for that, you need a uniform layer of cells. [Dulbecco passed minced mouse embryo tissue
through a fine screen so dispersed cells could be spread on Petri plates and infected with highly diluted virus.] For a week or two, nothing seemed to happen. And then one day
I put one of the cultures in a tangent light, and saw that it was full of plaques that were not visible in transmitted light [Someone, name forgotten, suggested using a vital stain
one that stained only living cells clear holes were seen in a red background.] The paper was published in 1952 in the Proceedings of the National Academy of Sciences
Then people from the National Science Foundation came and said that I should try to develop a system for polio [important since vaccine development was then ongoing the
NSF people also told Dulbecco about the use of trypsin to disperse cells]. Of course, people here at Caltech were very, very worried about work in polio. [So, a lab was rented
in a nearby hospital. At that time, Marguerite Vogt joined Dulbecco as a research fellow. Using monkey kidney cells, they developed a plaque system which worked very well.]
We succeeded in demonstrating two things: One is that if you want to have a pure strain, you have to pick one plaque, and second, that by picking different plaque types or
size, you can get mutants. I think this was essential for Sabin vaccine development. page 238

1952 Renato Dulbecco, Marguerite Vogt development of virus quantification by plaque assay (WEE, polio viruses)
5-hydroxymethyl cytosine cytosine

Seymour Cohens work on bacterial viruses, begun in 1945, was the first systematic exploration
of the biochemistry of virus-infected cells and of how viruses multiply. In 1952, he and Gerard
R. Wyatt independently discovered that one of the T-even viruses that infect E. coli contained a
new pyrimidine, 5-hydroxymethylcytosine, in its DNA, instead of the normal cytosine. Later, in
1957, he and Joel G. Flaks found that the enzyme that induced this pyrimidine was not found in
uninfected bacteria. They reasoned that the virus caused the E. coli cells to produce the enzyme
that would then induce the formation of 5-hydroxymethyl cytosine. This discovery made it
possible to develop drugs that would inhibit the enzymes induced by the virus without harming
healthy cells. The discovery that virus infection creates new metabolic pathways and affects flux
through existing pathways was of importance because it suggested that virus-specified enzymes
could be used as biochemical targets for antiviral agents.
Separately, when it was discovered in 1952 that T2, T4 and T6 bacteriophages lacked cytosine,
it was realized that this would make the double-helix model impossible. The demonstration that
these bacteriophages contained the modified form 5-hydroxymethylcytosine and that this was in
Seymour Stanley Cohen equal quantities to guanine removed this obstacle to the double-helix model.
In recent years, 5-hydroxymethylcytosine has been found in large amount in mouse, bovine,
rabbit and human zygotes and in human stem cells. It is eventually enzymatically converted to
cytosine, but research is ongoing questioning the ancient phylogenetic basis for this seeming
anomaly in a key building block of the stuff of inheritance.
Wyatt GR, Cohen SS. The bases of the nucleic acids of some bacterial and animal viruses: the
occurrence of 5-hydroxymethylcytosine. Biochem J. 1953;55:774-782.
page 239

1952 Seymour Cohen discovery that the DNA of some bacteriophages contains 5-hydroxymethylcytosine
Wallace Rowe (1926-1983)
Human adenovirus 5, cell culture
thin section electron microscopy

From a memoir by Harry Ginsberg: Robert Huebner of the National Institutes of Health visited Western Reserve to present a seminar. Afterward he came to my laboratory to
hear about my research, but before I could begin, he told the following exciting news: Dr. Wallace Rowe, a postdoctoral fellow in his laboratory, had made a thrilling discovery.
In an attempt to isolate the virus of the common cold, he had used an explant of an adenoid in cell culture, into which he inoculated secretions from a patient with a cold.
Dr. Rowe being a very smart young scientist also used control uninoculated cultures. After only a few days he noted that the cells in the control uninoculated cultures, as well
as the cells in those inoculated with patient secretions, became rounded and formed grape-like clusters, but the cells did not lyse. If he disrupted the cells, he could obtain
and pass on the cytopathic agent to other cells. Hence, the adenoids were obviously infected with a latent virus I asked Dr. Huebner if he would send me the virus after
publication of this exciting discovery. He immediately responded that prior publication was not necessary. He then called Dr. Rowe and told him to send me samples from his
adenoid cultures. (Can you imagine that occurring today?). On receipt of the sample, I demonstrated that it could replicate and be passaged in HeLa cells. Shortly, thereafter
Dr. Maurice Hilleman, who was then in the U.S. Army, visited Western Reserve... [Hilleman was working on acute respiratory diseases of recruits and had isolated a virus
from recruits who were thought to have influenza, but this was not influenza virus Ginsberg showed that Hillemans virus was related to Rowes virus by complement fixation
assays but distinct by neutralization assay. Since Rowe first isolated an adenovirus from human adenoid cells, more than 100 different viruses have been identified from humans
and various mammals, all related to each other by various methods, all distinct by neutralization assay.]
Rowe WP, Huebner RJ, Gilmore LK, Parrott RH, Ward, TG. Isolation of a cytopathogenic agent from human adenoids undergoing spontaneous degeneration in tissue culture.
Proc Soc Exp Biol. 1953;84:570-573.
Ginsberg HS. The life and times of adenoviruses. Adv Vir Res. 1999;54:1-13. page 240

1953 Wallace Rowe, Robert Huebner, others discovery of human adenoviruses


James Dewey Watson Francis Crick (1916-2004) Maurice Hugh Wilkins (1916-2004)
The lives of Francis Crick and James Watson changed when they met at Cambridge in 1951; neither was supposed to
be working on DNA, but they at once fell into often loud discussions on how the problem of its structure might be
approached. Jim and I hit it off immediately, Crick wrote, partly because our interests were astonishingly similar
and partly, I suspect, because a certain youthful arrogance, a ruthlessness and an impatience with sloppy thinking
came naturally to both of us. Their approach was to build an exact scale model of a DNA molecule that was driven
by the limited information available from X-ray crystallography. They constructed the backbone of a DNA molecule
in the form of a double anti-parallel spiral, or helix, with the two helices held together in the middle by ionic forces
the key change to this model came when Watson played with cardboard cutouts of the four bases, A, T, G, C,
and noticed that an A-T pair was identical in shape with a G-C pair. He immediately perceived how the bases could
point inward, holding the spiral together, provided that adenine always paired with thymine, guanine with cytosine
(Chargaff s rule). The pairing rule at once explained how one DNA chain could serve as the template for building
another, the essential requirement for any molecule that encoded hereditary information. Building the final model
took only a few days after this. It has not escaped our notice, they wrote in their paper in Nature, that the specific
Watson J, Crick F. Molecular structure of pairing we have postulated immediately suggests a possible copying mechanism for the genetic material. The
nucleic acids. A structure for deoxyribose personal/professional intrigues surrounding the work, involving Crick, Watson, Maurice Wilkins, Rosaland Franklin,
nucleic acid. Nature 1953;171:737-738. et al, which have been written about ever since, are not cited further here.
page 241

1953 James Watson, Francis Crick, Maurice Wilkins, Rosalind Franklin discovery of the structure of DNA
Rosalind Elise Franklin (1920-1958)

James Watson and Francis Crick with their structural model of DNA
Rosalind Franklins photo 51X-ray diffraction image of sodium salt of DNA, B form
This image provided critical evidence in identifying the structure of DNA. The photo was taken by Rosalind
Franklin while working at Kings College, London. James Watson was shown the photo by Maurice Wilkins.
Watson said, The instant I saw the picture my mouth fell open and my pulse began to race. Whether or not
Wilkins had Franklins approval to share the image has been a matter of endless controversy. The Nobel prize
is not awarded posthumously; Franklin, whose breakthrough data was used to formulate the
DNA structure, had died in 1958, and thus was not eligible for nomination. page 242

1953 James Watson, Francis Crick, Maurice Wilkins, Rosalind Franklin discovery of the structure of DNA
Morphology of Viruses!
by Frederik B. Bang
The Johns Hopkins University
[Annual Reviews of Microbiology, 1955]

The Microbe is so very small


You cannot make him out at all,
But many sanguine people hope
To see him through a microscope.
His jointed tongue that lies beneath
A hundred curious rows of teeth;
His seven tufted tails with lots
Of lovely pink and purple spots,
On each of which a pattern stands.
Composed of forty separate bands;
His eyebrows of a tender green;
All these have never yet been seen. But,
scientists, who ought to know.
Assure us that they must be so . . . .
Oh! let us never, never doubt
What nobody is sure about.
(CAUTIONARY VERSES
by Hilaire Belloc)

Murphy JS, Bang FB. Observations


with the electron microscope on cells
of the chick chorio-allantoic membrane
infected with influenza virus. J Exp Med.
1952;95:259-268.
James Bumgardner Murphy (1921-2007) Frederik B. Bang (1916-1981)

Influenza virus, budding from


the plasma membrane of an
infected cell in culture. Thin
section electron microscopy.

from: Noda T, Sagara H, Yen A,


Kawaoka Y, et al. Architecture
of ribonucleoprotein complexes
in influenza A virus particles.
Nature 2006;439:490-492.
page 243

1953 James Murphy, Frederik Bang discovery of virus release by budding at the cell surface
Sendai virus infection
adult mouse, 24 hours
after intranasal instillation
of virus, small bronchus
showing infection of nearly all
epithelial cells, frozen section
immunofluorescense

Nakao Ishida (1923-2009) Sendai virus murine parainfluenza virus 1


Negative contrast electron microscopy
In 1952, there was an outbreak of pnemonitis in newborn babies in the university hospital in Sendai, Japan. A virus was isolated from lung tissues from five fatal cases
by intranasal instillation of mice. All of the mice died between 5 to 7 days and showed a consolidation of the lungs, resembling that of influenza. The histopathological
findings in the mice were similar to that seen in the newborn babies. The virus was cultivated in embryonating eggs, and its hemagglutinin was compared with that of other
hemagglutinating viruses known at the time the hemagglutinin associated with the virus was unique. However, all this was smoke further characterization established the
virus as murine parainfluenza virus type 1, an important pathogen of laboratory mice but not of humans. Only later when methods improved and human parainfluenza virus
types 1, 2, 3, 4a/4b were discovered by Robert Chanock and his colleagues and shown to be important pathogens of humans did it become clear that the presence of the murine
virus, Sendai virus, was confounding the study of coincident human parainfluenza viruses. For years this served as an object lesson in the need for verification of virus identity.
Kuroya MK, Ishida N, Shiratori T. Newborn virus pneumonitis (type Sendai). 2. The isolation of a new virus possessing haemagglutinin activity. Yokohama Medical Bulletin
1953;4:217.
Ishida N, Homma M. Sendai virus. Adv Virus Res. 1978;23:349-383. page 244

1953 Nakao Ishida, colleagues discovery of Sendai virus (murine parainfluenza virus 1)
Rupert E. Billingham (1921-2002) Peter Brian Medawar (1915-1987) Leslie Baruch Brent
In 1945, Ray Owen, who was interested in bovine blood groups, discovered that most dizygotic-twin cattle fetuses had placental anastomoses and shared their blood supply
in utero. These cattle maintained a stable mixture of each others red cells throughout their lives. Unlike the situation with non-twin cattle, even when each had a different
blood group, transfusions between the twins did not cause any transfusion reaction. This mosaicism provided the first clue of immune tolerance. In 1949, Rupert Billingham
and Peter Medawar came across Owens work in a monograph by Frank Macfarlane Burnet and Frank Fenner, entitled The Function of Antibodies. This publication helped
them make sense of their work. The authors postulated that the age of the animal at the time of its first encounter with a foreign body was the critical factor in determining its
responsiveness and, hence, its recognition of nonself-antigens.
In a paper in Nature in 1953, Billingham, Leslie Brent, and Medawar reported the successful transplantation in mice of cells from normal mouse bone marrow, spleen, and
lymph nodes. The donor cells were obtained from adult mice and infused intravenously into normal newborn mouse recipients whose immune system was not yet sufficiently
developed to reject the cells. When the recipient mice grew up, they could freely accept skin and other tissue grafts from the original cell donor mouse strain, but from no
other donor. These first studies of acquired transplantation tolerance led to Medawars award (with F. Macfarlane Burnet) of the 1960 Nobel Prize. Medawar shared his
portion of the cash award with Billingham and Brent. [The mechanism of the phenomenon discovered by Billingham, Medawar and Brent is T-cell tolerance it is based on
the deletion of autoreactive T cells in the thymus so that the animal is rendered tolerant to self. T-cell tolerance has a very important role in advancing our understanding of
the pathogenesis, the immunopathogenesis of viral infections.]
Billingham RE, Brent L, Medawar PB. Activity acquired tolerance of foreign cells. Nature 1953;172:603-606.
page 245

1953 Rupert Billingham, Peter Medawar, Leslie Brent discovery of immunological tolerance
Thin section electron microscopy of retroviruses. (A) Intracisternal
type A particles. (B) Type B particles, mouse mammary tumor virus.
Wilhelm Bernhard (1920-1978) Albert J. Dalton (1905-1992) (C) Type C particles, murine leukemia virus. (D) Type C particles,
avian leukosis virus. (E) Type D particles, Mason-Pfizer monkey
Between 1953 and 1966, Wilhelm Bernhard, Albert Dalton, Franois virus. (F) Deltavirus, bovine leukemia virus. (G) Lentivirus, bovine
Haguenau, Etienne de Harven, Odile Croissant, Ludwik Gross and others immunodeficiency virus. (H) Spumavirus, bovine syncytial virus.
demonstrated the structure and intracellular development, first of Rous
sarcoma virus, then of other retroviruses including mouse mammary tumor
virus, avian endothelioma virus, avian erythroblastosis and myeloblastosis Family Retroviridae, Subfamily Orthoretrovirinae
viruses, murine leukemia viruses, and others. It was Bernhard who proposed Genus Betaretrovirus Mouse mammary tumor virus (the former type B particles)
the classification of types A, B, and C (and later D) retroviruses, now Genus Gammaretrovirus Murine leukemia virus (some of the former type C particles)
embedded in the definitions and names of the subfamilies and genera of Genus Alpharetrovirus Avian leukosis virus (some of the former type C particles)
the family Retroviridae. This book uses vernacular virus nomenclature and Genus Deltaretrovirus Bovine leukemia virus (some of the former type C particles)
taxonomy for formal taxonomy and nomenclature, the reader is referred Genus Epsilonretrovirus Walleye dermal sarcoma virus
to the current editions of the report of the International Committee on Genus Lentivirus Human immunodeficiency virus 1
Taxonomy of Viruses (ICTV) [http://talk.ictvonline.org/]. Nevertheless, for Family Retroviridae, Subfamily Spumaretrovirinae
clarity in this instance, it seems necessary to cite the formal family, subfamily Genus Spumavirus Simian foamy virus
and genus taxonomy and nomenclature:
Bernhard W. Electron microscopy of tumor cells and tumor viruses: A review. Cancer Res.
1958;18:491-509.
page 246

1953-1966 Wilhelm Bernhard, Albert Dalton, others morphology/morphogenesis/classification of retroviruses


Albert Bruce Sabin (1906-1993) Mammalian reovirus 3 (strain Dearing)
In 1959, Albert Sabin coined the acronym reovirus as a descriptive term for a new group of viruses. The acronym refers to the fact that although reoviruses can be isolated
from the respiratory and enteric tracts, there is no association of carriage with clinical disease, hence, they are orphan viruses (r-e-o). There are three common reovirus types,
based on their hemagglutination-inhibition activity. Prototypical laboratory strains of each were isolated from the respiratory and enteric tracts of children: Type 1 Lang, Type
2 Jones, Type 3 Dearing. All three virus types are found ubiquitously in the environment, including in such sources as water and sewage. This, combined with the fact that the
viruses are environmentally stable, explains why as many as 50% of adults carry antibodies to the viruses.
Sabin AB. Reoviruses. A new group of respiratory and enteric viruses formerly classified as ECHO type 10 is described. Science. 1959;130:1387-1389.
page 247

1953 Albert Sabin discovery of orthoreoviruses (the first reoviruses)


Dulbecco and Vogts growth curve of WEE virus on
a monolayer of chicken embryo cells

Renato Dulbecco (1914-2012) Marguerite Vogt (1913-2007)

Actually, the first one-step growth experiment was done in 1939 by Emory L. Ellis and Max Delbrck,
using bacteriophage proving that lysis of the host bacteria and release of the daughter burst of phage
indeed occur strictly on the dot [i.e. at a precise time interval.] But, it was considered an important advance
when Renato Dulbecco and Marguerite Vogt did this with a vertebrate virus (Western equine encephalitis
virus) in cell culture, since at that time few believed that discoveries made using bacteriophage pertained
directly to the viruses of vertebrates.

Dulbecco R, Vogt, M. One-step growth curve of western equine encephalomyelitis virus on chicken embryo
cells grown in vitro and analysis of virus yields from single cells. J Exp Med. 1954;99:183-199.

page 248

1954 Renato Dulbecco, Marguerite Vogt concept of the latent period, eclipse period, single cell burst size
Jonas Salk (1914-1995) and Julius Youngner Children lining up for polio vaccine, 1955
In 1949, John Enders, Thomas Weller, and Frederick Robbins grew poliovirus in monkey kidney cell culture. That same year, the virus
was separated into three immunological types. The development of an inactivated poliovirus vaccine moved forward quickly from this
point, under the leadership of Jonas Salk and Julius Youngner at the University of Pittsburgh and Pierre Lpine at the Institut Pasteur
in Paris. Salk used formalin to inactivate virus, Lpine used formalin followed by -propiolactone (the latter prevented the persistence
of residual virus that led to the Cutter Incident 200 cases of polio and 11 deaths in vacinees, all very soon after initial commercial
vaccine manufacture). In 1952, Salk vaccinated a group of children who had already had polio and recovered after vaccination,
their antibody titer increased. Next were volunteers who had not had polio, including Salk himself, his wife, and their children all
produced antibodies, and none developed polio. Next, in 1954, came field trials run by Thomas Francis of the University of Michigan
on behalf of the National Foundation for Infantile Paralysis; these trials were unprecedented in scope and size, involving 1.8 million
children in the U.S., Canada and Finland. These trials were among the first to use the
double-blind-placebo-controlled system that has since become standard. Scale up of
trivalent vaccine production involved six U.S. pharmaceutical companies, eventually
led by Connaught Laboratories in Montreal, where by 1955 most of the vaccine used in
North America was produced. Over 4 million doses had been given by August of 1955:
by 1957 polio cases in the U.S. had dropped by 85-90%.

Pierre Lpine
(1901-1989)
page 249

1954 Jonas Salk, Julius Youngner, Pierre Lpine, Thomas Francis development of inactivated polio vaccine
Owls Eye intranuclear inclusions of cytomegalovirus
Top: from Jesionek & Kiolemenoglov, 1904, then thought to be a
Margaret Gladys Smith (1896-1970) protozoan agent. Bottom: recent image.
Cytomegalovirus infections are characterized by the enlargement of infected cells (a process called cytomegaly by Ernest Goodpasture in 1921) and the presence of intranuclear
inclusions. The first proof of the viral nature of an agent responsible for cytomegaly was by Rufus Cole and Anne Kuttner in 1926. They ultrafiltered homogenates of
submaxillary gland tissue from infected adult pigs through a Berkfeld N filter and inoculated the filtrate into inclusion-free guinea pigs. Subsequently, inclusions were observed
in the inoculated animals guinea pig cytomegalovirus. Murine cytomegalovirus was first isolated in cell culture in 1954 by Margaret Smith from salivary gland tissues of
infected laboratory mice. This followed demonstration in 1934-36 by Anne Kuttner and Shao-Hsun Wang that the etiological agent of intranuclear inclusion bodies in murine
tissues could be transmitted by the inoculation of (unfiltered) tissue homogenates into healthy mice.
Smith MG. Propagation of salivary gland virus of mouse in tissue cultures. Proc Soc Exp Biol Med. 1954;86:435-440.
Jesionek A, Kiolemenoglou B. Uber einen befund von protozoenartigen gebilden in den organen eines hereditar-luetischen foetus. Muenchner Med Wochenschr.
1904;51:1905-1907. page 250

1954 Margaret Smith discovery of murine cytomegalovirus


Sigurdssons Institute for Experimental Pathology, Iceland

Bjrn Sigurdsson (1913-1959)


During the 1930s a flock of Karakul sheep was imported into Iceland from Germany. The intention was to start a lamb fleece industry. This flock, although apparently healthy
while kept in quarantine for several months, brought several diseases to the previously isolated Icelandic native breed of sheep. These included diseases caused by maedi-
visna virus and the scrapie prion. Maedi is a slowly progressive interstitial pneumonia (adenomatosis) of adult sheep, whereas visna is a slowly progressive encephalomyelitis
caused by the same lentivirus. Owing to Icelands sheep farming practices, using vast common summer pastures, these diseases spread widely causing enormous losses.
Annual mortality often reached 20-30%. In the face of these diseases, the Government, with financial aid from the Rockefeller Foundation, built the Institute for Experimental
Pathology, at Keldur, and made it a unit of the medical faculty of the University of Iceland. The director was Bjrn Sigurdsson, who had been a fellow at the Rockefeller
Institute, Princeton, New Jersey from 1941 until 1943. Sigurdsson isolated maedi-visna virus and worked on other diseases with long incubation periods and protracted
courses of illness, affecting largely the central nervous system and/or the lymphoreticular system, usually culminating in death. In 1954, Sigurdsson coined the term slow virus
infections to bring attention to this poorly defined group of diseases, which were eventually found to be caused by several quite different viruses and prions.
Sigurdsson B. Rida, a chronic encephalitis of sheep: with general remarks on infections which develop slowly and some of their special characteristics.
Br Vet J. 1954;110:341-354.
page 251

1954 Bjrn Sigurdsson development of the concept of slow viruses (e.g., maedi-visna virus, scrapie prion)
The first microtiter system was made in Budapest in 1951 by Gyula Taktsy; it consisted of (1) a plate with small wells drilled
into it, (2) micro-droppers, and (3) coiled wire loops for making dilutions. He machined by hand 6 rows of 12 drilled wells,
each ~5mm in diameter and 10mm deep, in a Lucite block he soon changed this to 8 x 12 wells the ubiquitous 96-well
plate. His original loop for making dilutions looked like the platinum loop used everywhere in bacteriology labs, but the
end of the wire was wound into a loose ball so that capillary action would draw up a constant volume into the interstices
of the coiled wire. Calibrated loops and droppers enabled him to perform dilution series in the wells of the block. Serial
dilutions were made by swizzling the loops in successive wells of the plates. Holding eight or twelve of the loops in one
hand, he was able to complete large numbers of serial dilutions at one time. The wire loops were quickly replaced by thin
steel knitting needles with a cut-out cup on the end that could be flame-sterilized. The system took years to make it to the
west, not because of patent protection, but because Taktsys publications went unnoticed. It wasnt until John Sever, at
the NIH, found Taktsys 1955 paper that the system was introduced internationally. Sever wanted to do large-scale viral
disease serologic surveys, but the method of transferring reagents and making dilutions, using small Cornwall syringes,
was very slow. He acquired a Taktsy device through a dealer in Budapest and Frank Cooke (Cooke Engineering, now
Dynex Technologies) improved it to serve Severs needs. They developed commercially improved loops and droppers and
injection-molded 96-well (polystyrene) plates. A tape dispenser was made to seal the plates, an especially valuable addition
for micro-neutralization tests. The plates were read from the bottom, using a concave shaving mirror mounted in a frame.
The microtiter system was a great success in virology and serology, for example for hemaglutination-inhibition assays,
complement-fixation assays, cell culture neutralization assays, etc., where very large numbers of tests were run as part of
large serosurveys. The first automated liquid handling device for automating serial dilutions, the Autotiter, was developed
in 1967 by Astec Inc. (now Tomtec Inc.). The first microplate readers evolved into the Multiskan photometer and later by
various spectrophotometric readers. By 1990 there were more than 15 companies producing a wide range of microplate
systems with different features. Today, it is estimated that 125 million microplates are used annually.
Taktsy G. The use of spiral loops in serological and virological micromethods. Acta Microbiologica et Immunologica
Hungarica 1955;3:191-202. [State Institute of Public Health, Budapest, Hungary]
Gyula Taktsy (1914-1980) Sever J. Major technological advances affecting clinical and diagnostic immunology. Clin Diag Lab Immunol. 1997;4:1-3.

Taktsys second dilution loop


Cookes modification of dilution loop
96-well plate reader
Holding 8 dilution loops in each hand!
Virus titration in 96-well plate page 252

1955 Gyula Taktsy invention of the microtiter system for virologic and serologic assays
1967

George K. Hirst (1910-1994) Peter Wildy (1920-1987)


1967

1955 Lloyd Kozloff (1924-2012), Robert Wagner (1923-2001), and Norman Salzman (1926-1997)
page 253

1955> George Hirst, Peter Wildy, Lloyd Kozloff, Robert Wagner, Norman Salzman three major virology journals
Telford H. Work (1921-1995) and Herbert
Richard Moreland Taylor (1887-1981) Hurlbut (1908-2001), bleeding a hooded
crow (Corvus corone), Egypt, 1955

Richard Moreland Taylor and young scientists Herbert


Hurlbutt and Telford H. Work, working under the auspices
of the Rockefeller Foundation at NAMRU-3, Cairo, Egypt
in 1954-55, were evaluating yellow fever endemicity
Richard Taylor bleeding Dinka following a severe outbreak which struck secluded Nubian
people, Sudan, 1955 populations in preceding years. In the course of this work
they discovered Sindbis virus.
Taylor RM, Hurlbut HS, Work TH, Kingston JR,
Frothingham TE. Sindbis virus: a newly recognized
arthropod-transmitted virus. Am J Trop Med Hyg.
1955;4:844-862. [Departments of Virology and Entomology,
U. S. Naval Medical Research Unit No. 3, Cairo, Egypt.]

Richard Moreland Taylor (1887-1981)


page 254

1955 Richard Taylor, Herbert Hurlbut, Telford Work, others discovery of Sindbis virus
Heinz Fraenkel-Conrat (1910-1999) Robley Cook Williams (1908-1995)

Fraenkel-Conrat H, Williams RC. Reconstitution of active tobacco mosaic virus from its inactive protein and nucleic acid
components. Proc Natl Acad Sci USA. 1955;41:690-698. [from the Virus Laboratory, University of California Berkeley.]
Summary. The preparation from TMV of protein and RNA fractions, which tend to recombine to form a nucleoprotein
carrying virus activity, is described. An electron microscopic search revealed no TMV rods in either of the two component
solutions at a concentration level thirty fold to three hundred fold greater than those at which the reconstituted virus was
assayed. In the reconstituted virus, on the other hand, up to about one-third of the material consisted of rods of the typical Tobacco mosaic virus
diameter and length of TMV, many, if not all, containing a nucleic acid core.The evidence thus seems reasonably complete from minimum-exposure study
that, under the conditions described, TMV nucleic acid enters into combination with TMV protein subunits and favors by Robley Williams, 1974
aggregation to rods, some of which are of sufficient length and structural integration to carry infectivity. negative contrast electron microscopy

page 255

1955 Heinz Fraenkel-Conrat, Robley Williams reconstitution of tobacco mosaic virus from its protein & nucleic acid
In 1954, Seymour Benzer began mapping the fine structure of a region of the genome of
bacteriophage T4; by the time he finished his work in 1961 he had developed the neoclassical
concept of the gene i.e., a discrete chromosomal region which (1) is responsible for a specific
cellular product, and (2) consists of a linear collection of potentially mutable units, each of which
can exist in several alternative forms and between which crossing over can occur. Benzer called
this unit a cistron, basing his definition on a criterion known as the cis-trans test for a functional
Seymour Benzer (1921-2007) unit of the genome. In the diagram of the cis-trans complementation test, m1 and m2 are
fictional alleles representing simple recessive mutations for years this was the way one told if
two recessive genes were part of the same gene or on two different genes. The test originated
with Edward B, Lewis who at Caltech in 1942 presented the concept as his PhD thesis. [Lewis
was awarded a Nobel Prize in 1995 for discovering the group of nearly identical master control
genes that orchestrate the development of body parts in all animals.]
Today, many different definitions of the gene are in use, all deriving from our evolving
understanding of molecular genetics. Maxine Singer and Paul Berg suggested the following
popular definition: A eukaryotic gene is a combination of DNA segments that together constitute
an expressible unit. Expression leads to the formation of one or more specific functional gene
products that may be either RNA molecules or polypeptides. Each gene includes one or more
DNA segments that regulate the transcription of the gene and thus its expression. Thus, a gene
includes: [1] a transcription unit, which includes the coding sequences, introns, leader and
trailer flanking sequences, and [2] regulatory sequences, which flank the transcription unit and
which are necessary for its specific function. As usual, virologists have been left to extend such
definitions to the RNA virus world.
Benzer S. The elementary units of heredity. In: McElroy WD, Glass B, editors. The chemical
basis of heredity. Baltimore: John Hopkins University Press; 1957.
Singer M, Berg P. Genes and genomes: a changing perspective. Mill Valley: University Science
Books; 1991.

Paul Berg and Maxine Singer


page 256

1955> Seymour Benzer, Paul Berg, Maxine Singer definition of a gene (cis-trans test, molecular genetic definition)
Zvonimir Dinter
(1914-1990)

Clinical signs of fowl plague include:


sudden death, labored breathing and
coughing, nasal and ocular discharge,
swollen face, cyanosis/necrosis of
Werner Schfer (1912-2000) comb and wattles, paralysis, paresis.

A number of orthomyxoviruses morphologically indistinguishable from influenza virus and possessing the
ribonucleoprotein antigen common to influenza A viruses were isolated early on from different species of birds. The first
virus to fall into this category was classical fowl plague virus, discovered in 1909 but not identified as an influenza A virus
until 1955, by Werner Schfer. Another virus, related to but not identical to, fowl plague virus was described by Zvonimir
Dinter in 1949 and called virus N it was identified as an influenza A virus by Rudolph Rott and Werner Schfer in 1960.
Schfer W. Vergleichende sero-immunologische untersuchungen ber die viren der influenza und klassischen geflgelpest.
Z Naturforsch. 1955;10b:81-91.
Dinter Z. Eine variante des virus der geflgelpest in Bayern. Tierrztl Umschau 1949;4:185-186.
Rott R, Schfer W. Physikalisch-chemische und biologische eigenschaften des virus N und seine beziehungen zur
influenza-a-untergruppe der myxoviren. Zbl Vet-Med. 1960;B7:237-248.
page 257

1955> Werner Schfer, Svonimir Dinter discovery that fowl plague virus is an influenza virus, likely zoonotic
Respiratory syncytial virus infection in cell culture
showing typical multinucleated syncytium formation
multiple fluorescent probes, confocal microscopy
Respiratory syncytial virus was first recovered in 1956 by J. Anthony Morris and colleagues
from chimpanzees during an outbreak of coryza in a colony at the Walter Reed Army
Institute of Research. In 1956, Robert Chanock and colleagues recovered the same virus
from an infant with pneumonia and from another infant with croup. Subsequently, the
virus, respiratory syncytial virus, has been found to be responsible for a considerable
proportion of the severe respiratory illness afflicting infants and small children. Chanock
wrote in 1961, It is clear that an RS virus vaccine, whether live attenuated or inactivated,
should receive a very high priority in any future plans for immunoprophylaxis of pediatric
respiratory disease.
Blount RE Jr, Morris JA, Savage RE. Recovery of cytopathogenic agent from chimpanzees
Robert Merritt Chanock (1924-2010) with coryza. Proc Soc Exp Biol Med. 1956;92:544-549.
Chanock R, Roizman B, Myers R. Recovery from infants with respiratory illness of a virus
related to chimpanzee coryza agent (CCA). I. Isolation, properties and characterization.
Am J Hyg. 1957;66:281-290.
Johnson KM, Chanock RM, Cook MK, Huebner RJ. Studies of a new human hemadsorption
virus. I. Isolation, properties and characterization. Am J Hyg. 1960;71:81-92.

Bovine respiratory syncytial virus, calf, 4 days postinfection,


infection of bronchial epithelium, immunohistochemistry page 258

1956 J. Anthony Morris, Robert Chanock, others discovery of respiratory syncytial virus (the first pneumovirus)
The first isolations of rhinoviruses were reported
in 1956 by William Pelon, William Mogabgab and
Winston Price, who had noted the growth in monkey
kidney cells of ultrafilterable agents associated
with mild respiratory disease. Their viruses had Phylogenetic
many similarities, but they were antigenically tree
distinct. Serological evidence indicated widespread depicting the
relationships
human infection, and a causal relationship with among 101
respiratory disease. Early studies were hampered by human
the insensitivity and variability of the tissue culture rhinovirus
systems used, but by 1958 the use of roller drums, serotypes
rather than stationary racks of cell culture tubes, and (P1-P2 region
of 5NCR)
of second generation monkey kidney cells had added
significantly to the sensitivity of the isolation system.
Sensitivity was soon improved further by the use of
WI-38 diploid human embryonic lung cells. By 1965,
an international workshop was held to organize the
classification and nomenclature of the very many
similar but distinct viruses being isolated around the
world, and in 1967 the numerical nomenclature system
was published in Nature. The signatories of this paper
were: A.Z. Kapikian (Chairman), R.M. Conant, V.V.
Hamparian, R.M. Chanock, P.J. Chapple, E.C. Dick,
J.D. Fenters, J.M. Gwaltney, Jr., D. Hamre, J.C. Holper,
W.S. Jordan, Jr., E.H. Lennette, J.L. Melnick, W.J.
Mogabgab, M.A. Mufson, C.A. Phillips, J.H. Schieble,
William J. Mogabgab (1921-2001) and D.A.J. Tyrrell.

Price WH. The isolation of


a new virus associated with
respiratory clinical disease
in humans. Proc Natl Acad
Sci USA. 1956;42:892-896.
Mogabgab WJ, Pelon W.
Problems in characterizing
and identifying an
apparently new virus
found in association with
mild respiratory disease in
recruits. Ann NY Acad Sci.
1957;67:403-412.
Consortium. Rhinoviruses: Model of human rhinovirus 3,
a numbering system. Nature William S. Pelon (1926-2005) from H. Adam Steinberg and
1967;213:761-763. Winston H. Price (1924-1981) and Albert Sabin Jean-Yves Sgro, University of
page 259 Wisconsin-Madison

1956 Winston Price, William Mogabgab, William Pelon discovery of human rhinoviruses, numerical nomenclature
Alfred Gierer Gerhard Schramm Heinz Fraenkel-Conrat Beatrice A. Singer
(1910-1969) (1910-1999) (1923-2005)
Until about 1948, most attention was focused on the proteins in viruses as the likely stuff of inheritance. Although it was
known that enzymes were proteins, the function of the nucleic acids was not known. Then, in 1949, Roy Markham and
Kenneth Smith showed that a plant virus, turnip yellow mosaic virus, contained two classes of particles, one infectious and
containing a nucleoprotein, the other non-infectious and containing an identical protein but no nucleic acid (in this case
RNA). This suggested that the viral RNA was essential for infectivity. This pioneering work was done just after Oswald Avery,
Colin MacLeod, and Maclyn McCarty demonstrated that DNA was the bacterial transforming factor (1944) and just before
the Hershey-Chase experiment (1952). However, doubts as to whether the RNA in viruses was really the genetic material
persisted. Heinz Fraenkel-Conrat and Robley Williams showed in 1955 that upon incubating a solution containing a mixture
of a non-infectious disassembled tobacco mosaic virus (TMV) protein and purified, apparently non-infectious, viral RNA,
normal-looking infectious TMV particles were formed. This experiment had a big impact because it appeared that a living
molecule had been produced in vitro from non-living constituents. The next year, 1956, the pioneering experiments of
Alfred Gierer and Gerhard Schramm showed that TMV RNA alone was capable of initiating virus replication; the same result
was obtained at the same time by Heinz Fraenkel-Conrat and his wife Beatrice Singer. Thus it became clear that the infectivity
observed a year before in Fraenkel-Conrats TMV reconstitution experiment was a result of the infectivity of TMV RNA.
Fraenkel-Conrat and Singer then did reconstitution experiments using different host range variants to provide protein and
RNA: reconstituted viruses had the host range specificity characteristic of the virus from which the RNA was obtained, not
the protein.
Gierer A, Schramm G. Infectivity of ribonucleic acid from tobacco mosaic virus. Nature 1956;177:702-703.
Gerhard Schramm (center)
observing sedimentation of a protein,
Schlieren optics camera, air-driven Fraenkel-Conrat H, Singer B. Virus reconstitution and the proof of the existence of genomic RNA. Phil Trans R Soc Lond B
ultracentrifuge 1999;354:583-586. (historical review)
page 260

1956 Alfred Gierer, Heinz Fraenkel-Conrat, others discovery of the infectivity of viral RNA (tobacco mosaic virus)
Stewart Harvey Madin Charles J. York Delbert Grant McKercher
(1918-2002) (1920-2000) (1914-1988)
The first report on infectious bovine rhinotracheitis
appeared in 1954; it was described as an apparently new
upper respiratory disease of dairy cattle in California. It
appeared suddenly and was characterized by high fever
and agalactia in addition to respiratory signs. The following
year, a similar disease was described in Colorado feedlots
and dairies. Delbert McKercher showed by comparative
serology that the respiratory disease found in California
dairy and beef cattle and in Colorado beef cattle were the
same. Stewart Madin, Charles York and Delbert McKercher
isolated the causative agent from nasal washing from
clinically affected cattle: infectious bovine rhinotracheitis
virus, now also called bovine herpesvirus 1 (BHV-1). In
1961, J.A. Armstrong, Helio Pereira and Christopher
Andrewes showed that the virus is a herpesvirus.

Madin SH, York CJ, McKercher DG. Isolation of


the infectious bovine rhinotracheitis virus. Science
School of Veterinary Medicine, University of California Davis, 1961 1956;124:721-722.
page 261

1956 Stewart Madin, Charles York, Delbert McKercher discovery of infectious bovine rhinotracheitis virus
Chikungunya disease was first described by Marion Robinson and William Hepburn Russell
Lumsden in 1955, following an outbreak in 1952 on the Makonde Plateau, along the border between
Mozambique and Tanganyika (today Tanzania). According to Marion Robinsons initial 1955 report
on the epidemiology of the disease, the term chikungunya is derived from the Makonde root verb
kungunyala, meaning to dry up or become contorted, in reference to the stooped posture developed
as a result of the arthritic clinical manifestations of the disease. Subsequent authors apparently
overlooked the references to the Makonde language and assumed that the term derived from
Swahili, the lingua franca of the region the result has been many erroneous interpretations of the
name.
From Lumsden: The epidemic to be described here was first recognized in 1952 by Dr. Marion
C. Robinson of the Lulindi and Newala Hospitals of the Universities Mission to Central Africa.
The occurrence of the epidemic was notified to the Virus Research Institute [Entebbe] by the
Director of Medical Services, Tanganyika, in February, 1953, and a team was sent by air to attempt
virus isolation and to carry out general epidemiological studies. Dr. R. W. Ross and I arrived in
Newala on 21 February, 1953, and began immediately the collection of sera from acute cases of the
disease and the catching of biting insects from which to attempt virus isolation The description
of the epidemic falls naturally into three parts, the clinical features of the disease (Dr. Marion C.
Robinson), the general description of the environment and the epidemiology (Dr. W.H.R. Lumsden),
and the characteristics of the viruses concerned (R. W. Ross) and it seems convenient to record the
results in this form.
Robinson MC. An epidemic of virus disease in Southern Province, Tanganyika territory, in 1952-
1953. I. Clinical features. Transactions of the Royal Society of Tropical Medicine and Hygiene
1955;49:28-32.
Lumsden WHR. An epidemic of virus disease in Southern Province, Tanganyika territory, in
1952-1953. II. General description and epidemiology. Transactions of the Royal Society of Tropical
Medicine and Hygiene 1955;49:33-57.
Ross RW. The Newala epidemic. III. The virus: isolation,
pathogenic properties and relationship to the epidemic. J
Hyg (Lond). 1956;54:177-191.
[R. William Ross, at the time a staff member of the
Rockefeller Foundation sponsored Virus Research Institute
at Entebbe, Uganda, later spent many years as a faculty
member of the Department of Pathology, University of
Cambridge. Despite much searching, no photo of him has
been found.]

William Hepburn Russell Lumsden


(1914-2002) page 262

1956 R.William Ross, Marion Robinson, W.H.R. (William) Lumsden discovery of chikungunya virus
Phylogenetic tree of the family Paramyxoviridae,
showing the human parainfluenza viruses (HPIVs)

Robert Merritt Chanock (1924-2010)


The human parainfluenza viruses (HPIVs) were discovered by Robert Chanock and his colleagues between
1956 and 1960. HPIV1, 2 and 3 were first isolated from infants and children with lower respiratory tract
disease and HPIV4a and b from children and young adults with minor upper respiratory tract infections.
Soon afterward, the viruses were shown to be major causes of croup (HPIV1, 2,3) and pneumonia and
bronchiolitis (HPIV3). As a group these viruses are second only to respiratory syncytial virus as the cause of
severe, sometimes fatal respiratory tract disease in infants and children.
Chanock RM, Parrott RH, Cook K, Andrews BE, Bell JA, Reichelderfer T, Kapikian AZ, Mastrota FM,
Huebner RJ. Newly recognized myxoviruses from children with respiratory disease. N Engl J Med.
1958;258:207-213. Parainfluenza virus 4a
Thin section and negative contrast electron microscopy
Chanock RM, Johnson KM. Infectious disease: respiratory viruses. Annu Rev Med. 1961;12:1-18.

page 263

1956 Robert Chanock, others discovery of human parainfluenza viruses


Margaret Gladys Smith Thomas Huckle Weller Wallace P. Rowe Human CMV, kidney, H&E
(1896-1970) (1915-2008) (1926-1983)
Human cytomegalovirus (CMV) was isolated in cell culture only after the development of the technology to culture human cells by
John Enders, Thomas Weller and Frederick Robbins. In 1955, Margaret Smith isolated human CMV from the salivary gland of a patient
at autopsy; the virus grew in human but not in mouse cell culture. Her paper was rejected because she was also working with murine
CMV and the editor thought she might have had a contaminant. It was only in 1956 when she reisolated the virus and isolated the same
virus from the kidney of another patient that her paper was accepted. In 1957, Weller, working on toxoplasma in human embryonic cell
culture, isolated a virus from an infant suspected of having congenital toxoplasmosis, and Wallace Rowe, working on respiratory viruses,
isolated a virus from adenoid tissue of a patient suspected of having varicella. Rowe took his material to Wellers lab for confirmation,
where it was found to be similar to the virus Weller had isolated. The three independently isolated viruses were exchanged among the
three groups, and their similarity was established in advance of publication indeed, a commendable example of cooperation. Weller
named the virus cytomegalovirus. [Ho M. The history of cytomegalovirus and its diseases. Med Microbiol Immunol. 2008;197:65-73.]
Smith MG. Propagation in tissue cultures of a cytopathogenic virus from human salivary gland virus (SVG) disease. Proc Soc Exp Biol
Med. 1956;92:424-430.
Rowe WP, Hartley JW, Waterman S, Turner HC, Huebner RJ. Cytopathogenic agent resembling human salivary gland virus recovered
from tissue cultures of human adenoids. Pro Soc Exp Biol Med. 1956;92:418-424.
Wellers image of human CMV Weller TH, Macauley JC, Craig JM, Wirth P. Isolation of intranuclear inclusion producing agents from infants with illnesses resembling
inclusions in cell culture, 1956 cytomegalic inclusion disease. Proc Soc Exp Biol Med. 1957;94:4-12.
page 264

1956 Margaret Smith, Thomas Weller, Wallace Rowe discovery of human cytomegalovirus
Aleutian disease: lung, mink, multisystemic immunologically
mediated lymphoplasmacytic perivascular and diffuse lesions
Aleutian disease virus of mink was first recognized in 1956 by John Gorham and
John R. Gorham (1922-2011) James B. Henson G. R. Hartsough in ranch-raised mink (Mustela vison) carrying the Aleutian coat
color gene, a gene which produces a highly desired gun-metal grey pelt. In the
1960s, it was common practice for mink ranchers to make their own vaccine against
canine distemper virus, an important pathogen of mink, by emulsifying tissue from
distemper-infected mink and formalizing it. This practice led to severe outbreaks
of Aleutian disease and extensive virus spread (Aleutian disease virus, a parvovirus,
is more resistant to formalin than canine distemper virus). In adult mink, Aleutian
disease is a persistent, slowly progressive lethal infection, marked by high levels
of antiviral antibodies and hypergammaglobulinemia. However, the virus is not
neutralized and severe immune complex glomerulonephritis and vasculitis develop.
Massive accumulations of macrophages and plasma cells occur in many organs and
tissues. Antibody-coated virions infect macrophages leading to antibody-dependent
enhanced disease. Because of this exceptional immunopathology, leaders in viral
disease pathogenesis research did extensive work on the disease throughout the 1960s
and 1970s. Included in this were research groups at the Rockefeller Institute led by
Robert Leader (1919-1996) and the Scripps Research Institute led by Frank Dixon
(1921-2008). At the same time, research continued at Washington State University led
by John Gorham and James Henson.
Hartsough GR, Gorham JR. Aleutian disease in mink. Nat Fur News 1956;28:10-11.
Henson JB, Gorham JR, Leader, RW, Wagner, BM. Experimental
hypergammaglobulinemia in mink. J Exp Med. 1962; 116:357-364.
Frank Dixon (1921-2008) Robert Leader (1919-1996)
page 265

1956 John Gorham, James Henson, Robert Leader, Frank Dixon discovery of Aleutian disease of mink virus
Early theories of immunity explained the large repertoire of
antibody specificities with the notion that antigen somehow acts
as a template to transfer information to the globulin-producing
machinery in certain cells. This was termed an instruction theory;
another theory, termed a selective theory, postulated that antigen
exposure induces the elaboration of a sample of antibodies from a
range of pre-existing specificities. In 1955, Niels Jerne suggested
that the information for all antibody specificities is inherent in the
host, and that the sole function of antigen is to select the matching
antibody and transport it to the appropriate cells where, somehow,
they would be stimulated to produce more of the same. Jernes
idea attracted little support at the time; its surviving claim-to-
fame lies in the fact that David Talmage and Frank Macfarlane
Burnet independently saw in its selectionist approach the seed of
an interesting idea. In 1957, Talmage suggested, it is tempting to
consider that one of the multiplying units in the antibody response
is the cell itself only those cells are selected for multiplication
whose synthesized product has affinity for the antigen injected.
Burnet, having also read Jernes paper, and pondering heavily on
it, asked why it was so attractive, though obviously wrong. By
replacing molecules with cell clones and their membrane receptors,
Burnet said the whole picture fell into shape. In his 1957 paper,
Niels Kaj Jerne (1911-1994) Burnet assumed that a large repertoire of lymphoid cell clones were
precommitted to producing a similarly large repertoire of antibody
specificities. As he put it, ... antibody production is a function not
only of the cells originally stimulated, but of their descendants.
When an antigen enters the body, a number of cell clones that
happen to produce antibodies against that antigen are selected for
and multiply into much larger clones of cells, producing antibody
Frank Macfarlane Burnet (1899-1985) molecules specific against the intruding antigen. Although it
encountered some skepticism in the beginning, the clonal selection
theory rapidly gathered support, and within less than a decade the
immunological community switched from believing in templates
to believing in selection. Since then, clonal selection has been the
explicit theoretical foundation of immunology.
Jerne NK. The natural-selection theory of antibody formation. Proc
Nat Acad Sci USA. 1955;41:849-857.
Talmage DW. Allergy and immunology. Annu Rev Med.
1957;8:239-256.
Burnet FM. A modification of Jernes theory of antibody production
using the concept of clonal selection. Aust J Sci. 1957;20:67-69.

David W. Talmage Burnets clonal selection diagram page 266

1957 Niels Jerne, David Talmage, Frank Macfarlane Burnet clonal selection as the central mechanism in immunity
Confocal microscopy offers advantages over conventional optical microscopy, including the ability to control
depth of field, to eliminate background information away from the focal plane, and to collect serial optical sections
(tomographic sections) from thick specimens. It uses filtering techniques to eliminate out-of-focus light or glare
in specimens whose thickness exceeds the immediate plane of focus. Current instruments are highly evolved from
the earliest versions, but the principle of confocal imaging advanced by Marvin Minsky, and patented in 1957, is
employed in all modern confocal microscopes. Today, image formation in a confocal microscope is achieved by
scanning one or more focused beams of light, usually from a laser, across the specimen, which is usually labeled
with one or more fluorescent probes. The pinhole point of illumination is brought to focus in the specimen by the
objective lens and a sequence of points of light from the specimen are built up by scanning the specimen and sending
the light signal to a photomultiplier tube and from there to a computer.
Marvin Minsky, from the Massachusetts Institute of Technology, was a leader in the development of artificial
intelligence concepts and technologies, and only worked on confocal microscopy on the side: he only published his
invention as a patent. Yet he not only built a prototype microscope and made it work, but he also tried to see that it
was applied however, as he said, I demonstrated the confocal microscope to many visitors, but they never seemed
very much impressed with what they saw on that radar screen [he used a surplus radar screen as the photomultiplier
tube]. Only later did I realize that it is not enough for an instrument merely to have a high resolving power; one must
also make the image look sharp. Perhaps the human brain requires a certain degree of foveal compression in order to
engage its foremost visual abilities. In any case, I should have used film - or at least have installed a smaller screen! It
took many years for confocal microscopy to catch on, but when it did there was an explosion in its use, due in part to
the relative ease with which extremely high-quality images can be obtained from specimens usually prepared in the
same way as they would be for conventional fluorescence microscopy. In fact, confocal technology is proving to be
one of the most important advances ever achieved in optical microscopy.
Marvin Lee Minsky

Sagittal section of rat cerebellum, confocal microscopy, multiple fluorescent probes


page 267

1957 Marvin Minsky invention of the confocal microscope


In 1956, Alick Isaacs was investigating influenza
viruses at the National Institute for Medical Research
(NIMR), Mill Hill, when he had a cup of tea with Jean
Lindenmann, who had just arrived as a postdoc from
Switzerland. They began investigating the mechanism
of viral interference, that is, the capacity of cells
infected with one virus to resist infection by another.
To study the matter they set up a simple system:
pieces of chick embyro chorio-allantoic membrane
were placed in culture medium in test tubes, treated
with heated virus (later ultraviolet light inactivated
virus) and rotated in a roller drum for 24 hours. The
membranes were then removed, washed, and put
into tubes and challenged with fresh virus. The
fluids in these tubes were tested for the presence
of viral hemagglutinin as proof whether virus was
replicating or not. True to form, the first virus
seemed to be thwarting the growth of the second.
They wrote, It was surprising to find that after 24
hours considerable interfering activity remained in
the surrounding fluid after the treated membranes
were removed and new membranes added. So the
possibility was considered that fresh interfering
activity was produced by the membranes. The active
substance was christened interferon (a hybrid of
interference and the suffix on, which was in vogue
at the time), and it was concluded that establishment
of interference was accompanied by the liberation
of interferon. The initial discovery was recorded in
Isaacs lab notebook under the entry: In search of an
interferon. Lindenmann took it all in stride, saying,
Alick Isaacs (1921-1967) Jean Lindenmann I thought it quite natural that when you did research
you discovered things. As stated by Christopher
Andrewes, who was director of the NIMR at the time,
The paper, in the Proceedings of the Royal Society
reporting this discovery must be accounted one of the
outstanding landmarks in biological science The
term breakthrough is one of those most abused and
overworked especially in popular accounts of science,
but it can justly be used concerning the discovery of
interferon.

Isaacs A, Lindenmann J. Virus interference. I. The


interferon. Proc Roy Soc London Set B 1957;147:258-
273.
page 268

1957 Alick Isaacs, Jean Lindenmann discovery of interferons


Telford Hindley Work (1921-1995) Telford Work and Pushpa
Kyasanur Forest disease virus, the etiologic agent of a hemorrhagic disease in
humans (mortality rate ~10%), was first isolated in the Shimoga district of Karnataka
state (then Mysore) of India in 1957. The virus commonly infects the black faced
langur (Presbytis entellus, from which the first virus isolate was made) and red faced
bonnet monkey (Macaca radiata); it is transmitted between monkeys by the bite
of the tick, Haemaphysalis spinigera, especially in its nymphal stage. The virus also
circulates in rodents, shrews and birds. The cause of the sudden emergence of the
virus in India in the late 1950s and its subsequent localization to the Karnataka state
alone are not clear. It has been argued that the virus and other hemorrhagic and
encephalitic tick-borne viruses almost certainly exist in other parts of India and in
the region in general, but their presence has not yet been recognized. Phylogenetic
analyses of the virus and other tick-borne flaviviruses suggest that they evolve
gradually, whereas mosquito borne viruses evolve more rapidly and become more
genetically and ecologically divergent in the course of their evolution. The original
paper on the virus and disease by Telford Work, Pravin Bhatt, F.R. Roderiguez and
their colleagues reminds us of the classic terse style of ecologic and epidemiologic
description of the era:
Work TH, Roderiguez FR, Bhatt PN. Virological epidemiology of the 1958 epidemic
of Kyasanur Forest disease. Am J Public Health Nations Health 1959;49:869-874. Pravin Bhatt, bleeding a subject, 1959
page 269

1957 Telford Work, F.R. Rodriguez, Pravin Bhatt discovery of Kyasanur Forest disease virus
Equine viral arteritis is a contagious disease of horses caused by equine arteritis
virus (EAV). The disease was first described about 200 years ago. Horsemen also
recognized long ago that otherwise healthy stallions could transmit the disease to
susceptible mares at breeding, and that these carrier stallions could be a source of
infection for many years. The virus was fist isolated by Elvis Doll, John Bryans, William
McCollum and M.E.W. Crow during an outbreak of respiratory disease and abortion
on a breeding farm in Bucyrus, Ohio in 1953. The infection of equine species (horses,
donkeys, and mules) occurs throughout the world, although its incidence varies
markedly between countries and among horses of different breeds. The vast majority
of infections are inapparent or subclinical, but outbreaks occur that are characterized
by any combination of influenza-like illness in adult horses, abortion in pregnant
mares, and fatal interstitial pneumonia in young foals. The global dissemination of
the virus and the rising incidence of the disease likely reflect the incredible amount of
international movement of horses for competition and breeding,

Doll ER, Bryans JT, McCollum WH, Crowe MEW. Isolation of a filterable agent
causing arteritis of horses and abortion by mares. Its differentiation from the equine
abortion (influenza) virus. Cornell Vet. 1957;47:3-41.
Elvis Roger Doll John T. Bryans
(1912-1967) (1924-2004)

page 270

1957 Elvis Doll, John Bryans, William McCollum discovery of equine arteritis virus (the first arterivirus)
James R. Gillespie
Alan O. Betts (1927-2005)
Kyu M. Lee

Cell culture clean room

Veterinary Virus
Research Institute
(now Baker Institute
for Animal Health)
Cornell University
1956

James R. Gillespie (1918-2011) Bovine virus diarrhea is a very important disease of cattle, worldwide. When it was first identified and studied,
however, by James Gillespie and Kyu M. Lee at Cornell University, A.B. Hoerlein and his colleagues at Colorado State
University, and Albert L. Fernelius at what is now the National Animal Disease Center in Ames Iowa, only the most
severe forms of the disease were recognized most dramatically, fatal mucosal disease. As is now known, bovine
virus diarrhea (BVD) virus infection may be inapparent or may cause severe disease, with abortion, infertility, and
immunosuppression that underlies calf respiratory and enteric diseases. BVD virus causes two patterns of infection: (1)
infection of a seronegative animal usually causes no clinical illness the virus is eliminated by the humoral and cellular
immune response and the animal is protected against further infection. (2) In contrast, infection of the dam between
the second and fourth month of gestation often leads to persistent infection of the fetus the fetus is immunotolerant
to the infecting BVD virus strain and at birth is antibody negative. The calf sheds large amounts of virus during
its entire lifetime [Infection later in gestation may lead to abortion, malformation, or to the birth of a normal,
immunocompetent calf ]. If the persisting virus mutates into a cytopathogenic virus biotype or if the persistently
infected animal is superinfected with an antigenically related biotype, it will succumb to lethal mucosal disease. The
virus is maintained in nature by the small population of animals that become immunotolerant and persistently infected;
these animals often die prematurely but some live a relatively normal life for several years, all the time shedding large
amounts of virus and serving as a very efficient virus reservoir.
The complex pathogenesis of bovine Lee KM, Gillespie JH. Propagation of virus diarrhea virus of cattle in tissue culture. Am J Vet Res. 1957;18:952-953.
virus diarrhea virus infection and
Underdahl NR, Grace OD, Hoerlein AB. Cultivation in tissue culture of cytopathogenic agent from bovine mucosal
mucosal disease
disease. Proc Soc Biol Med. 1957;94:795.
page 271

1957 James Gillespie, Kyu M. Lee, Norman Underdahl, others discovery of bovine virus diarrhea virus
Matthew Meselson Frank W. Stahl Jerome Vinograd (1913-1976)
In 1957, Matthew Meselson and Frank Stahl were seeking a means to implement their idea for a bold experiment to verify the hypothesis that
DNA replication involved the separation of the two parental strands, one going into each of the two daughter DNA molecules see following
pages for this, what has been called the most beautiful experiment in history. They initially wanted to make the parental strands heavier than
normal by incorporation of 5-bromouracil and sought Jerome Vinograds advice as to whether 5-bromouracil-containing DNA could be separated
from normal DNA by velocity ultracentrifugation. Vinograd indicated that this seemed unlikely unless the difference could be magnified by
approximately matching the density of the DNA to the density of the salt solution in which the DNA was suspended. From this germinated
the concept of equilibrium sedimentation of macromolecules in density gradients. In equilibrium sedimentation methods the macromolecule
(e.g., DNA) is dissolved in a salt solution (e.g., CsCl) of the appropriate density and centrifuged to equilibrium (approximately 24 hours). At
equilibrium, the CsCl builds its own stable concentration (density) gradient, increasing in density with the radius. At equilibrium the suspended
DNA molecules form a narrow band at the CsCl density that is equivalent to their own density. Vinograds notion worked beautifully. In several
later papers they refined the theory and extended the application of their density gradient technique to other macromolecules: hemoglobin,
MS2 bacteriophage RNA, T7 bacteriophage DNA, X174 bacteriophage, and southern bean mosaic virus. They also found that the addition of
dimethylsulfoxide enhanced the ability to band RNA in CsCl without aggregation.

Meselson M, Stahl FW, Vinograd J. Equilibrium sedimentation of macromolecules in density gradients. Proc Natl Acad Sci USA. 1957;43:581-588.
page 272

1957 Matthew Meselson, Frank Stahl, Jerome Vinograd invention of isopycnic density gradient ultracentrifugation
Meselson M, Stahl FW. The replication of DNA
in Escherichia coli. Proc Natl Acad Sci USA.
1958;44:671-682. [original figure 4 legend below and
analytic ultracentrifuge Schlieren optics photographs
at right]
Ultraviolet absorption photographs showing DNA
bands resulting from density gradient centrifugation
of lysates of bacteria sampled at various times after
the addition of an excess of 14N substrates to a
growing 15N-labeled culture. Each photograph was
taken after 20 hours of centrifugation at 44,770 rpm.
The density of the CsCl solution increases to the
right. The degree of labeling of a species of DNA
corresponds to the relative position of its band
fully labeled and unlabeled DNA are shown in the
lowermost frame, which serves as a density reference.
The DNA in the band of intermediate density is just
half-labeled proof is provided by the frame showing
the mixture of generations 0 and 1.9 the peak of
intermediate density is centered at 50 (2) percent of
the distance between the 14N and 15N peaks.

For those who learned their molecular biology after the


Meselson-Stahl experiment (the vast majority of scientists living
today), it will be surprising to learn of the grave doubts about
the semiconservative mechanism for the replication of DNA
implied by the Watson-Crick paper of 1953. Even Max Delbrck
worried about the means of unwinding the strands of DNA
during replication. The result of the Meselson-Stahl experiment
more or less resolved the doubts about all this, leading to broad
support from other scientists for the Watson-Crick structure
and the semiconservative mechanism of the replication of DNA.
page 273

1958 Matthew Meselson, Frank Stahl discovery of the semi-conservative mode of replication of DNA
Throughout the 1940s and 1950s everyone
dismissed the notion that viruses could cause cancer
that is, except for Sarah Stewart. She began her
career at NIH in 1936. When Bernice Eddy arrived
the following year the two became friends and
developed a long-running collaboration.
In 1944, Stewart asked for support for research
on the possible link between animal tumors and
viruses, but she was told she was not qualified. So,
she enrolled in Georgetown University School of
Medicine, graduating in 1949 (as the first woman to
receive an MD from that institution).
After an internship, she returned to the NIH, to the
National Cancer Institute, just as Ludwik Gross at
the Rockefeller Institute reported that some mouse
leukemias and parotid gland tumors were caused by
viruses.
In 1956, in Bernice Eddys laboratory, Stewart
injected tumor extracts from mice into other
laboratory animals and into cell cultures. Mouse
tumor material passaged in rhesus monkey cell
cultures and then inoculated into newborn mice and
mouse embryos caused 20 distinct types of tumors.
Tumors were also produced in hamsters, rabbits,
guinea pigs and rats. The virus, at Eddys suggestion,
Bernice Eddy (1903-1989) was called polyoma virus.
The findings caused an international sensation
there was even a Time magazine cover story the
director of the NCI, John Heller, said, Right now,
the hottest thing in cancer research is research on
Sarah Elizabeth Stewart (1906-1976) viruses as possible causes. Thus, the field of viral
oncology was born.
Stewart SE. Eddy BE, Borgese N. Neoplasms in
mice inoculated with a tumor agent carried in tissue
culture. J Nat Cancer Inst. 1958;20:1223-1243.
Eddy BE, Stewart SE, Young R, Mider GB.
Neoplasms in hamsters induced by mouse tumor
agent passed in tissue culture. J Nat Cancer Inst.
Murine polyoma virus 1958;20:747-760.
model built from X-ray
crystallographic data
page 274

1958 Sarah Stewart, Bernice Eddy discovery of murine polyoma virus (the first polyomavirus)
After Albert Coons (1912-1978) first developed
immunofluorescence technology in 1942, its first practical
application was in 1956 by William Cherry, who used it to
identify several bacterial pathogens. Then in 1958, Robert
Kissling and a guest in his lab at CDC, Robert Goldwasser,
developed an immunofluorescence (fluorescent antibody)
test for rabies. Impression smears of brain tissue of
animals suspected of having exposed humans were fixed
and overlayed with fluorescin isothiocyanate-conjugated
hyperimmune rabies globulin. Positives clearly showed
apple-green stippling and large aggregates of rabies viral
antigen (Negri bodies) when examined in a microscope
equipped with an appropriate light source and UV
filters. Thousands of diagnostic specimens were tested
to compare the new technique with other diagnostic
methods of the day, especially intracerebral inoculation of
mice (taking 21 days), as first developed by Leslie Webster
in the 1930s. The massive comparative testing was done in
several labs, including those of James McQueen (Florida),
Donald Dean and Melvin Abelseth (New York) and the
Fourth Army Medical Lab, Ft. Sam Houston, Texas, where
the author was a fledgling veterinary corps officer, and
where many of the specimens submitted to him from
the area were rabies positive. The comparative testing Rabies infection, fox, brain,
proved that the fluorescent antibody test was at least as impression smear, diagnostic
good as mouse inoculation and results could be reported Robert E. Kissling immunofluorescence, ~1965
in hours of specimen receipt. The test was as good from
the beginning as today conjugates made according
to the CDC recipe were excellent and microscopes
and filters were good enough (but were improved over
the years). For the first time, laboratory results greatly
influenced post-exposure human treatment (vaccine, later
supplemented with human immune globulin). In recent
years, despite success with several other rapid diagnostic
test methodologies, the fluorescent antibody test is still
the standard for rabies diagnosis globally. The first rapid
diagnostic test in virology has become the oldest!
Goldwasser RA, Kissling RE. Fluorescent antibody
staining of street and fixed rabies virus antigens. Proc Soc
Exp Biol Med. 1958;98:219-223.
Goldwasser RA, Kissling RE, Carski TR, Hosty TS.
Fluorescent antibody staining of rabies virus antigens in
the salivary glands of rabid animals. Bull World Health Robert Goldwasser CDC Rabies Advisory Committee, 1959: Front (L-R): W.G. Winkler, E.S. Tierkel,
Organ. 1959;20:579-588. (1918-2012) R.K. Sikes, H.N. Johnson, T.F. Sellers. Back: R.L. Parker, R.E. Kissling,
page 275 D. Dean, J.H. Steele, J.P. Fox, K. Habel, R. Courter

1958 Robert Kissling, Robert Goldwasser development of rabies immunofluorescence diagnostic method
Mercedes Weissenbacher

Celia Coto and Armando S. Parodi (1909-1969) Ignacio Pirosky (1901-1987)


The viral etiology of Argentine hemorrhagic
fever was established in 1958 by two independent
groups. Each isolated the virus from the blood of
patients and organs obtained at autopsies at the
Junn Regional Hospital in the province of Buenos
Aires, Argentina.
Parodi AS, Greenway DJ, Rugiero HR, Frigerio M,
De La Barrera JM, Mettler N, Garzon F, Boxaca
M, Guerrero L, Nota N. Sobre la etiologa del
brote epidmico de Junn. Dia Med. 1958;30:2300-
2301.
Pirosky IJ, Zuccarini EA, Molinelli A, Di Pietro
P, Martini B, Ferreyra LF, Gutman F, Vazquez T.
Virosis hemorrgica del noroeste bonaerense.
Endemoepidmica, febril, enantemtica
y leucopnica. I. La primera inoculacin
experimental al hombre. Orientacion Med.
Calomys musculinus 1959;8:144-148.
Marta Sabattini
page 276

1958 Armando Parodi, Ignacio Pirosky, colleagues discovery of Junin virus (Argentine hemorrhagic fever)
Denis Burkitt was trained as a surgeon at Trinity College, Dublin, and went to Kampala, Uganda first
during WWII. In 1958, he described common jaw tumors in African children. Shortly thereafter,
the tumors were identified as a form of malignant B cell lymphoma, now called Burkitts lymphoma.
Histologically, the lymphomas are composed of morphologically uniform medium sized cells with round
nuclei and abundant basophilic cytoplasm. These cells have an extremely high rate of proliferation.
Burkitts lymphoma is most commonly associated with a complex chromosomal translocation of the c-myc
gene and overexpression of myc protein, but it also is epidemiologically associated with Epstein-Barr virus
(EBV) infection and with malaria in children in equatorial Africa (endemic Burkitts lymphoma). It occurs
sporadically in other geographical areas, where it also occurs among adults. Questions remain about
the role of particular EBV variants rather than ubiquitous virus strains, and the evidence for association
with malaria is weak. Burkitts lymphoma has become the basis of many lessons: (1) The lymphoma is
not amenable to surgery or radiation therapy but the success of chemotherapy (~90% of cases cured) has
influenced all cancer chemotherapy. (2) The involvement of c-myc and the chromosomal translocation
has been a model for understanding the molecular pathogenesis of other lymphomas and leukemias.
(3) The lymphoma was the first human tumor shown to be associated with a virus this inspired the
search for others, some preventable by vaccination (HBV, HPV). (4) EBV was shown to be associated
with nasopharyngeal carcinoma, T cell lymphomas, and other tumors in immunodeficient patients, again
pointing to pathogenetic research themes. (5) The finding that EBV is able to induce proliferation in B cells
via latent genes and probably drives proliferation of Burkitt lymphoma cells has led to yet other discoveries
of neoplastic mechanistics. (6) The finding of the unique geographic distribution Burkitts lymphoma
(often in areas of high incidence of intense malaria infection) has led to the development of a new kind
of geographic medicine. (7) The association of HIV/AIDS and Burkitts lymphoma in Africa has led to
another set of pathogenetic mechanistic questions, all still under investigation.
Burkitt D. A sarcoma involving the jaws in African children. Brit J Surg. 1958;46:218-223.

Denis Parsons Burkitt (1911-1993)


Global distribution of Burkitts lymphoma cases

page 277

1958 Denis Burkitt description of Burkitts lymphoma in African children


CT tomography, brain, PML

Gabriele M. Zu Rhein Billie L. Padgett


and Duard Lee Walker (1921-2010)
In 1958, at the Massachusetts General Hospital, Karl-Erik strm, Edward Peirson Richardson, Jr. (1918-
1998) and their colleagues described progressive multifocal leukoencephalopathy (PML) as a specific
disease entity after thorough clinical and histopathological observation of two patients with chronic
lymphocytic leukemia and one patient with Hodgkins disease who presented with multiple demyelinating
lesions of the central nervous system and a rapid, fatal outcome. The first clue of a possible viral cause JC virus, oligodendrocyte nucleus, PML
stemmed from the detection of inclusion bodies in the nuclei of damaged oligodendrocytes; this was
confirmed in 1965 by demonstration of papovavirus-like particles in lesions. The virus was isolated in
1971 from the brain of a patient with Hodgkins disease who died of PML, and it was named JC virus
(JCV) after the patients initials.
strm K-E, Mancall EL, Richardson EP Jr. Progressive multifocal leuko-encephalopathy; a hitherto
unrecognized complication of chronic lymphatic leukaemia and Hodgkins disease. Brain 1958;81:93-111.
Zu Rhein GM, Chou S-M. Particles resembling papovaviruses in human cerebral demyelinating disease.
Science 1965;148:1477-1479.
Padgett BL, Walker DL, ZuRhein GM, Eckroade RJ, Dessel BH. Cultivation of papova-like virus from
human brain with progressive multifocal leucoencephalopathy. Lancet 1971;1:1257-1260.
Demyelination, brainstem, PML
page 278

1958 Gabriele Zu Rhein, Duard Walker, Billie Padgett, others progressive multifocal leukoencephalopathy, JC virus
By the late 1960s, it was known that hematopoietic stem cells from the bone
marrow are disseminated into the bloodstream as T cell precursor cells or as B
cells. The T cell precursors migrate to the thymus, where they differentiate into T
cells (blue). They are then exported to the periphery, where they recirculate from
the blood through thymus-dependent areas of lymphoid tissues (e.g., paracortical
areas of lymph nodes), into the lymph and back to the blood via the thoracic
duct. B cells (red) populate the thymus-independent areas of lymphoid tissues
(e.g., follicles); some recirculate in the same way as T cells. From: Miller JF. The
Peter Medawar (1915-1987) and James Learmonth Gowans
golden anniversary of the thymus. Nat Rev Immunol. 2011;11:489-495.

When James Gowans published his landmark paper on the recirculation of lymphocytes in the rat from blood to lymph and back
again, the field of immunology was still focused on humoral immunity cellular immunology was ill defined. Gowans had become
interested in lymphocytes at the suggestion of Howard Florey and Peter Medawar. Today we know that lymphocyte recirculation
is the main mechanism for dispersing the immunologic response repertoire, for directing lymphocyte subsets to specialized
microenvironments (especially in the thymus) that control their differentiation, and for targeting immune effector cells to the sites of
viral (and microbial) invasion and damage. The continual mixing of cells is particularly important for proper function of the immune
system because cellular activation events often require direct cell-to-cell contact. Inappropriate migration and activation, however, can
contribute to immunopathology and autoimmunity. Gowans and E. Julie Knight found that the output of lymphocytes is about 109/day,
and is kept in homeostatic status by recirculation. Their work was made possible by mastering the difficult technique of thoracic duct
cannulation. The thoracic duct, the terminal lymphatic draining conduit, empties into the superior vena cava. Tracing the fate of RNA-
labeled small lymphocytes collected from the thoracic duct and transfused into normal rats, Gowans and Knight found that the main
channel from blood to lymph was within lymph nodes, the lymphoid follicles of the spleen and Peyers Patches of the intestine.
Gowans JL. The recirculation of lymphocytes from blood to lymph in the rat. J Physiol. 1959;146:54-169.
Gowans JL, McGregor DD, Cowen DM. Initiation of immune responses by small lymphocytes. Nature 1962;196:651-655.
Gowans JL, Knight EJ. The route of re-circulation of lymphocytes in the rat. Proc R Soc Lond B Biol Sci. 1964;159:257-282.
page 279

1959 James Gowans discovery of lymphocyte recirculation


People lined up for blocks
for Sabin polio vaccine,
San Antonio, Texas, 1962

Albert Bruce Sabin Herald Rea Cox


(1906-1993) (1907-1986)

Oral polio vaccine is an attenuated live-virus vaccine, which was developed by passage of
the viruses through cells in culture and animals, thereby producing attenuating mutations.
Such vaccines were developed by Albert Sabin and coworkers and separately by Hilary
Koprowski and Herald Cox. In 1958, an NIH committee evaluated these vaccines for their
ability to induce immunity to polio, while retaining a low incidence of neuropathogenicity
in monkeys. Based on these results, the Sabin strains were chosen for worldwide
distribution. In 1961-1962, monovalent and in 1963 trivalent vaccines were licensed the
latter became the vaccine of choice in the U.S. and most other countries of the world it
was easy to administer, making it more suitable for mass vaccination campaigns. There
was second wave of mass immunizations between 1962 and 1965 about 100 million
Hilary Koprowski Americans (~56% of the population) received Sabin vaccine. The result was a substantial
(1916-2013) reduction in the number of polio cases, even from the much reduced levels that followed
Derivation of Sabins vaccine viruses the earlier introduction of the Salk vaccine.
page 280

1959 Albert Sabin, Herald Cox, Hilary Koprowski, others development of attenuated live-virus polio vaccine
Kilham rat virus
infection ataxia
(splayed feet) from
virus-induced cerebellar
hypoplasia

Kilhams H-1 virus


Negative contrast electron microscopy
Parvoviruses of rats and mice have been shown to cause (1) fetal death and resorption, (2) neonatal hepatitis,
(3) cerebellar hypoplasia, and (4) severe hemorrhagic encephalopathy, all varying with the species, strain,
age and immune competence of the host animal. Most early experiments, led by Lawrence Kilham, involved
parenteral inoculation of virus into pregnant or neonatal animals, seeking models for virus-induced birth
defects. This work was extended to cats and hamsters, which when infected before or at birth also exhibited
Lawrence Kilham (1910-2000) cerebellar hypoplasia. Productive replication of murine parvoviruses (Kilham rat virus, H-1 virus, minute
virus of mice, and perhaps all the autonomously replicating parvoviruses of other species) requires cellular
Lawrence Kilham: When I factors that are expressed only during cell differentiation and division this accounts for the predilection of
started out as a microbiologist or these viruses for mitotically active cells and this in turn accounts for the tropism and damage seen in infected
virus hunter, I wondered how animals. In recent years, the interest in these viruses has changed: they are of major interest because of their
my other love, studying birds on adverse effects on all biomedical research involving rodents. Infection, often not recognized in colonized
early mornings, evenings, and rodents, causes immunomodulation (inhibition of lymphocyte proliferation and the generation of cytolytic
weekends, would fare. Would my T cell activity, etc.), inhibition of the antibody response, hematopoetic disruption, tumor suppression, and
enthusiasm for birds and animals contamination of cultured cell lines any and all confounding much research involving laboratory rodents.
cut in on my pursuit of viruses? I
need not have worried. Indeed, Kilham L, Olivier L. A latent virus of rats isolated in tissue culture. Virology 1959;7:428-437.
Kilhams book became a key Kilham L. Rat virus (RV) in hamsters. Proc Soc Exp Biol Med. 1961;106:825-829.
resource during the West Nile
virus invasion of the U.S. Kilham L, Margolis G. Fetal infection of hamsters, rats and mice induced with minute virus of mice (MVM).
starting in 1999. Teratology 1971;4:43-62.
page 281

1959 Lawrence Kilham, others discovery of murine parvoviruses and their role in congenital diseases
Sydney Brenner Robert W. Horne
Negative staining revolutionized our view of the viruses, for the first time allowing details of
virion surfaces [capsomeres, envelopes, projections (spikes, peplomers)] and some internal
features] to be seen. Where would virology be today if the many variations on the negative
staining method had not been available?
Brenner S, Horne RW. A negative staining method for high resolution electron microscopy of
viruses. Biochimica et Biophysica Acta 1959;34:103-110. [The first viruses imaged by the new
method were tobacco mosaic virus and turnip yellow mosaic virus.]
Horne RW, Brenner S. A negative staining technique for high resolution visualization of viruses.
In: Proc Fourth International Conference on Electron Microscopy, vol. 2. Berlin: Springer Verlag;
1960. p. 625. [from the Medical Research Council Unit for Molecular Biology and Electron
Microscopy, Cavendish Laboratory, Cambridge, Great Britain]
Horne RW, Wildy P. Virus structure revealed by negative staining. Adv Virus Res. 1963;10:101-70.
Horne RW, Wildy P. An historical account of the development and applications of the negative
staining technique to the electron microscopy of viruses. J Microsc. 1979;117:103-122. Human adenovirus 5, negative contrast electron microscopy
page 282

1959 Sydney Brenner Robert Horne invention of negative contrast electron microscopy
In researching the virologists cited in this book, many talents were found other than
those related to their scientific careers. This page, a placeholder for this subject, is
exemplary.
From Alasdair Steven and Wolfgang Baumeister: Graphical representation of
molecular and cellular features is a key form of communication in structural biology,
in which abstract symbols of an economical and visually appropriate kind, pseudo-
color coding, and dynamic animations all play their parts. Accordingly, it should
not be surprising that many structural biologistslike traditional biologists before
themare talented artists who also express themselves on non-scientific topics.
[Here we] illustrate the approaches of and pictures by several practicing scientist-
artistsin this sampling, electron microscopists.
The Swedish training schooners Gladan and Falkon off Bornholm. From an original
oil painting by Robert W. Horne. Robert Hornes interest in marine artwork started
with studies relating to the form, construction and development of early sailing
vessels. His color work began with the use of inks, pens and fine brushes, followed
by water-colors, and finally oil painting. He embarked seriously on marine painting
during the 1970s at the Norwich School of Art and Design. His work includes
detailed research into the plans of historic sailing vessels and their reconstruction in
paintings.

The two-mast cutter/schooner HMS Pickle bringing news of the naval victory
at Trafalgar in 1805. Water-color painting by Robert W. Horne. Pickle is depicted
being driven hard in a freshening wind as she approaches Falmouth on the south
coast of England. Completing delivery to London required an additional two-and-
a-half days land journey by the messenger, Lieut. J. Lapotiere, in a post-chaise,
involving 23 staging points.
Steven AC, Baumeister W. The state of the art. J Struct Biol. 2008;163:201-207.

page 283

1959 Sydney Brenner Robert Horne invention of negative contrast electron microscopy
Single-stranded
circular DNA of
X174 bacteriophage.
This was the first DNA
viral genome to be
sequenced, by Fred
Sanger, in 1977.

Albrecht Karl Kleinschmidt


(1916-2000)

Albrecht Kleinschmidt invented the technique for preparing monolayers


of DNA for electron microscopy, in which the DNA molecules are bound
to a film of denatured basic protein (e.g., cytochrome c) on the surface of
an aqueous solution. The DNA molecules are consequently brought from
a three dimensional conformation to a two dimensional one by adsorption
to the protein layer. The DNA-protein complex is then transferred to a
solid surface (e.g., an electron microscope grid containing a collodion film),
dried, and contrasted with heavy metals by shadowing or staining. The
method has also been applied to RNA.
Kleinschmidt, AK, Lang D, Jachters D, Zahn RK. Darstellung und
lngenmessung des gesamten desoxyribonucleinsure-inhaltes von T2-
bakteriophagen. Biochimica et Biophyisca Acta 1962;61:857-864.
[This paper contains the famous electron micrograph of the DNA of T2
bacteriophage, which was used in many publications, even on the cover of
several molecular biology and microbiology books. It is used here as well.] Electron micrograph of the DNA of T2 bacteriophage
Original magnification, x100,000 page 284

1959 Albrecht Kleinschmidt invention of method for electron microscopic visualization of viral DNA
Gerald Maurice Edelman Rodney R. Porter (1915-1985) Alfred Nisonoff (1923-2001)
Up to the year 1959, our knowledge of the nature and mode of action of antibody
molecules was vague and incomplete, in spite of a century of research. Gerald
Edelmans and Rodney Porters experimental approaches were quite different:
both first separated the molecule now called IgG (molecular weight 150 kD),
from other serum proteins. Porter then subjected IgG to brief digestion with
the enzyme papain and separated the fragments he obtained two identical
fragments, now called Fab, and one fragment, now called Fc. Edelman dissociated
IgG by reducing its interchain disulfide bonds with mercaptoethanol he
obtained two fractions, one called light chain, the other heavy chain. Alfred
Nisonoff s approach was similar to Porters, only he used the enzyme pepsin he
obtained a single large fragment composed of two Fab-like fragments and one
Fc fragment. Porter finally answered how the fragments went together using
antibodies against each fragment. Porter was then able to build a model of the
molecule, which has since been generally accepted and successively fitted with
Porters model of 7S antibody (IgG) 1959 Contemporary model of IgG additional molecular details. The structure of the other classes of antibody
molecules, IgM, IgA, IgE and the subclasses of IgG were resolved subsequently.
These discoveries incited a major burst of research in immunology, which
immediately spilled over into virological research.
page 285

1959 Gerald Edelman, Rodney Porter, Alfred Nisonoff discovery of the molecular structure of antibody molecules
DNA of
bacteriophage
X174

From Robert Sinsheimer, Oral History, 1990. California Institute of Technology Archives. (Interview by
Shelley Erwin):
[1957] we got Phi X DNA purified, did the light-scattering measurements on it, established its
molecular weight it wasnt Watson and Crick, and therefore could not be double-stranded. And that
fit with the sedimentation coefficient and the light scattering, which gave now a molecular weight of
1.7 million daltons. All that said it was single-stranded. And then the only question really was, Was this
somehow something we had done in extracting it from the virus? I was able to show spectroscopically and
in other ways that it was a single-strand in the virus itself. So it clearly was single-stranded DNA, and we
wrote this up and published it in the first issue of the Journal of Molecular Biology. And that, of course,
was for a time a sensation. We had never seen a single-stranded DNA. Of course, that immediately also
Robert L. Sinsheimer raised a number of obvious questions of great importance. We had this model for double-stranded DNA,
but how did the single-stranded DNA reproduce? Indeed, how would it even get transcribed? So we
developed ways of labeling the DNA and following it into the cell. And to make a long story short, we
were able to show that the first thing that happened to the single-stranded DNA after it went into the cell
was that it became a complementary-stranded DNA it became a double-stranded molecule. And then,
obviously that could be transcribed. And then, that double-stranded form multiplied as such we call
that a replicative form it multiplied as such for a time, and then later in the infection it started to make
the single strands of DNA. It went back to the single-stranded form to put into the progeny. Not only was
it really novel in itself, but what was really novel were the techniques to follow it into the cell.
Computer model of
Sinsheimer RL. A single-stranded deoxyribonucleic acid from bacteriophage X174.
bacteriophage X174
J Mol Biol. 1959;1:43-53. page 286

1959 Robert Sinsheimer discovery that a virus may have a single stranded DNA genome (bacteriophage X174)
Yalow RS, Berson SA. Assay of plasma insulin in
human subjects by immunological methods. Nature
1959;184:1648-1649.
Yalow RS, Berson SA. Immunoassay of endogenous
plasma insulin in man. J Clin Invest.
1960;39:1157-1175.

Rosalyn Sussman Yalow (1921-2011) Solomon A. Berson (1918-1972)


Rosalyn Yalow spent many years at the Veterans Administration Hospital in the Bronx, New York, where with
her collaborator, Solomon Berson, she used radioisotopes in novel ways, first to obtain more accurate estimates
of blood volume, then to study how the thyroid gland and kidneys remove iodine from the blood. They expanded
these studies to globins and other serum proteins, and then to hormones, especially insulin. They discovered that
globulins bind radioactive insulin in the blood of insulin-treated diabetics, and concluded that treatment with
insulin injections immunized patients so that they developed insulin-binding antibodies, which kept the insulin
molecules in the bloodstream. This was the first proof that so small a protein as insulin could stimulate an immune
response. The scientific establishment was reluctant to accept this finding, but when their observations were
confirmed by others the significance of the discovery became clear. Not only had they proven that the immune
system could recognize and respond to smaller molecules than previously thought possible, but they had done so
using a breakthrough technology radioimmunoassay. The development of radioimmunoassays, the first method
for measuring antigen-antibody complexes directly, stimulated a revolution in immunology and also in virology. By
the end of the 20th century, many different radioimmunoassays were in use in viral diagnostics and research. Bersons
death at age 54 precluded his nomination for the Nobel Prize in Physiology or Medicine, which Yalow shared with
Roger Guillemin and Andrew Schally in 1977.
page 287

1959 Rosalyn Yalow, Solomon Berson development of radioimmunoassays (RIAs)


In 1955, Arthur Kornberg started to focus on the synthesis
of DNA after learning that Severo Ochoa and his colleagues
had apparently created a synthetic RNA from adenosine
diphosphate (ADP) (the product turned out to be not RNA
polymerase, but polynucleotide phosphorylase). Working with
cell extracts of E. coli, Kornberg found which combinations of
nucleotides and other ingredients resulted in the most rapid
synthesis of DNA. From this, in 1958 he found and purified
DNA polymerase, and with it was able to synthesize DNA
in the lab. Subsequently, Kornberg and his students found
enzymes responsible for DNA repair and others responsible for
the initiation and elongation of DNA molecules. The enzymes
they discovered helped make possible the development of
recombinant DNA technology, i.e., genetic engineering. [In
1967, Kornberg became the first person to synthesize infectious
viral DNA in vitro (bacteriophage X174).] A chance discovery
in 1955 by Marianne Grunberg-Manago, a postdoctoral fellow
in Ochoas lab, was the key to the eventual discovery of RNA
polymerase: she observed that a bacterial extract converted
phosphate substrate into ADP. The responsible enzyme,
purified with Ochoas urging and direction, astonishingly
converted ADP and other nucleoside diphosphates into
RNA-like polymers. The initial hope that this enzyme, named
polynucleotide phosphorylase, might be responsible for the
biosynthesis of RNA was soon dispelled the discovery of
true RNA polymerases, which copy DNA templates with
great specificity using nucleoside triphosphates rather than
Arthur Kornberg Severo Ochoa de Albornoz diphosphates, settled the matter. Nevertheless, polynucleotide
(1918-2007) (1905-1993) phosphorylase proved to be of great value, particularly in
deciphering the genetic code. Ochoa (with help from Leon A.
Heppel) employed the enzyme to synthesize a variety of RNA-
like polymers, which were then used to identify many of the
nucleotide triplets that encode the amino acids in the synthesis
of proteins. The 1959 award of the Nobel Prize in Physiology or
Medicine to Kornberg and Ochoa was for their discovery of
the mechanisms in the biological synthesis of ribonucleic acid
and deoxyribonucleic acid.
Grunberg-Manago M, Ortiz PJ, Ochoa S. Enzymic synthesis of
polynucleotides. I. Polynucleotide phosphorylase of Azotobacter
vinelandii. Biochim Biophys Acta 1956;20:269-285.
Lehman IR, Bessman MJ, Simms ES, Kornberg A. Enzymatic
synthesis of deoxyribonucleic acid. I. Preparation of substrates
and partial purification of an enzyme from Escherichia coli. J
Biol Chem. 1958;233:163-70.
page 288

1959 Arthur Kornberg, Severo Ochoa discovery of mechanisms in the replication of DNA and RNA
William H. Prusoff (1920-2011)
William H. Prusoff developed the first antiviral drug ever to be (and still) licensed and marketed, first for the topical treatment of herpetic keratitis, and then for fatal or
disabling illnesses such as adult herpetic encephalitis, keratitis in immunocompromised (transplant) patients, and disseminated herpes zoster. This is 5-iodo-2-deoxyuridine
(IUdR), an analog of thymidine. Prusoff first was looking for anti-cancer activity, but a physician, H.E. Kaufman, working in a Boston Hospital, conceived the idea of using IUdR
to treat herpetic keratitis. He first reported animal experiments, showing that the drug was truly therapeutic; it promoted healing of the cornea when applied 2 or even 5 days
after infection the virus load in rabbit corneas after 4 days of treatment was much less than in controls. This led to clinical trials and licensing.
Prusoff is also known for research conducted during the 1980s with his colleague Tai-Shun Lin, on d4T, a potent treatment for HIV/AIDS. Under the trade name Zerit (Bristol-
Myers Squibb), d4T (2-3-didehydro-2-3-dideoxythymidine, a nucleoside analog reverse transcriptase inhibitor) forms part of many drug cocktails used to treat AIDS
worldwide.
Prusoff WH. Synthesis and biological activities of iododeoxyuridine, an analog of thymidine. Biochim Biophys Acta 1959;32:295-296.
Kaufman HE. Clinical cure of herpes simplex keratitis by 5-iodo-2-deoxyuridine. Proc Soc Exptl Biol Med. 1962;109:251-252.
Kaufman HE, Nesburn AB, Maloney ED. IDU therapy of herpes simplex. Arch Ophthalmol. 1962;67:583-591.
page 289

1959 William Prusoff development and licensing of the first antiviral drug, 5-iodo-2-deoxyuridine (IUdR)
William Reeves (1916-2004) Wilbur Downs (1913-1991) William Scherer (1925-1982)
The founding committee,
American Committee on
Arthropod-Borne Viruses
1959

The Committee still provides a forum for


exchange of information among people
interested in arbovirus research. It meets
annually in association with the American
Society of Tropical Medicine and Hygiene.
An executive council is its governing body,
and its work is carried out by subcommittees:
Subcommittee on Arbovirus Laboratory
Safety, Subcommittee for Evaluation of
Arthropod-Borne Status, Subcommittee
on Information Exchange, Subcommittee
on Inter-relationships Among Catalogued
Arboviruses, and Subcommittee on Low
Passage Viruses.

Alexis Shelokov Richard Taylor (1887-1981) Telford Work (1921-1995) page 290

1959 William Reeves, others founding of the American Committee on Arthropod-Borne Viruses (ACAV)
World Food Prize

Walter Plowright and his team at the East African Veterinary


Research Organization at Muguga in Kenya, ~1970. Sitting on
Plowrights left is the Head Laboratory Assistant, Francis Ngugi
Plowright W. The application of monolayer tissue
culture techniques in rinderpest research. II. The use of
attenuated culture virus as a vaccine for cattle. Bulletin
de lOffice International des Epizooties 1962;57:253-257.
Plowright W, Ferris RD. Studies with rinderpest virus in
tissue culture. The use of attenuated culture virus as a
vaccine for cattle. Res Vet Sci. 1962;3:172-182.
Plowright W, Taylor WP. Long-term studies of the
immunity in East African cattle following inoculation
Walter Plowright (1923-2010) with rinderpest cell culture vaccine. Res Vet Sci.
1967;8:118-128.

Walter Plowright, as a young veterinary pathologist, left his native Great Britain to carry out research in Kenya and
Nigeria, starting in 1950. Working at the East African Veterinary Research Organization at Muguga in Kenya from
1956 to 1971, he adopted the then new cell-culture techniques of John Enders, et al., that had been used to develop
measles vaccine, to produce a live-attenuated rinderpest virus vaccine. Unlike its predecessors, Plowrights tissue
culture rinderpest vaccine (TCRV) could be used safely in all breeds of cattle of any age or health status. It could
be produced very economically and conferred lifelong immunity. Initial field trials (1956 to 1963) showed that the
vaccine was genetically stable, produced no clinical side effects and was easily standardized. This vaccine was used
as the centerpoint in the global rinderpest eradication program (GREP) of the FAO, which succeeded in its goal in
2010. Plowright was recognized with the 1999 World Food Prize for his development of the vaccine that led to the Walter Plowright demonstrating the growth
eradication of the second disease from the world, the first being smallpox. of rinderpest virus in cell culture to the
Governor of Kenya, ~1970
page 291

1960> Walter Plowright development of the attenuated-live virus rinderpest vaccine used in global eradication
Maurice Hilleman (1919-2005) SV40 virus
at Walter Reed Army Institute of Research, 1957 Negative contrast electron microscopy
from minimum-exposure study by Robley Williams, 1974

In 1957, after two years of widespread distribution of Salk inactivated polio vaccine, Maurice Hilleman and Benjamin Sweet, then at the Walter Reed Army Institute of
Research, identified the existence of a virus in the rhesus monkey kidney cells being used for vaccine production (the 1:4,000 dilution of formaldehyde used to inactivate
poliovirus did not inactivate this other virus). By 1960, they had characterized the virus, calling it SV40, and had found that it contaminated Sabin attenuated live-virus vaccine
as well. The virus was named for its effect on African green monkey cells, which developed an unusual number of vacuoles, and for the fact that it was the 40th virus they
found contaminating rhesus monkey kidney cells. Soon, SV40 virus antibody was found in the sera of more than half of all individuals that had received Salk vaccine. The virus
became famous when Bernice Eddy, working at the NIH, tested extracts from the monkey kidney cells used to make polio vaccine for possible cancer causing agents the
newborn hamsters she inoculated developed several kinds of tumors similar to those induced by polyoma virus, which she and Sarah Stewart had described earlier. A scandal
arose when she was silenced and prohibited from working on polio vaccine by supervisors who saw political fallout from her findings. Her findings did leak out and Sweet
and Hilleman, now at Merck Research Laboratories, replicated her work. To his credit, Hilleman repeatedly sounded warnings about the risks inherent in the use of monkey
cells for vaccine production. After years of procrastination by industry, polio vaccine production was switched to African green monkey cells, which were free of the virus
(but which were contaminated by other viruses, such as a simian cytomegalovirus). By 1962, it was confirmed that SV40 virus can transform human cells, but the question of
whether the virus does cause cancer in humans has only been answered in the negative by the most recent statistical analyses of epidemiologic/serologic data meanwhile, the
issue has become part of the argument used by activist groups that wish to stop all vaccinations.
Sweet BH, Hilleman MR. The vacuolating virus, SV40. Proc Soc Exp Biol Med. 1960;105:420-427.
Hilleman MR. Discovery of simian virus 40 (SV40) and its relationship to poliomyelitis virus vaccines. Dev Biol Stand. 1998;94:183-190.
page 292

1960 Benjamin Sweet, Maurice Hilleman discovery of SV40 virus (simian virus 40) (polyomavirus)
Vernon Todd Riley Kenneth Edmund Keith Rowson
(1914-1982) (1924-2004)
Lactate dehydrogenase-elevating virus (LDH virus) was discovered by Vernon Riley and his colleagues in
1960 during work on plasma enzyme levels in tumor-bearing mice. They found that transplantable tumors
of many types caused a 5-10-fold increase in plasma lactate dehydrogenase (LDH) activity within 3 days of
transplantation and before the tumors were clinically obvious. To produce this dramatic increase in plasma
enzyme level it was not necessary to transplant cells; cell-free plasma from tumor-bearing mice was equally
effective. The raised enzyme levels could be serially transmitted from mouse to mouse and proved to be The discovery of LDH virus had a profound effect on our
caused by a virus which replicated very rapidly, mostly from infection of macrophages. Very high titers of viral understanding of the subtleties of the effects of some
infectivity (109 ID50/ml) are present in the plasma within 24 hours after infection, and a stable viremia at a lower infections, especially on immunological responses and
level (104 ID50/ml) is established after 7 to 10 days. This persists for the remainder of the animals life but does tumor growth in experimental animals. Its discovery
not cause any obvious disease or reduction in life expectancy. The persistent viremia clearly provides a source also had a great effect on how we use experimental
of virus for transmission by bloodsucking ectoparasites. It seems likely that this is the method of cross-infection animals in research and how we interpret data from such
by which the virus is maintained in feral mice, a number of which have been found to be infected in Europe, experimentation, such that we now expect experimental
Australia and the U.S. Kenneth Rowson and Brian Mahy did much to call attention to the confounding, often animals to be free of all adventitious viruses (even if this is
inexplicable effect of the presence of contaminating LDH virus in various kinds of experiments employing mice, not always the case).
in effect giving rise to the universal demand for specific-pathogen-free mice [from Brian W.J. Mahy]
Riley VT, Lilly F, Huerto E, Bardell D. Transmissible agent
Vernon Riley was an innocent victim of the McCarthy era. He was accused of being a communist, and although associated with 26 types of experimental mouse neoplasms.
he eventually received restitution and acknowledgment of his innocence, he was forced to leave the National Science 1960; 132:545-547.
Cancer Institute. Thanks to the invitation of C.P. Rhodes. then President of the Memorial Sloan-Kettering
Cancer Center, he joined the Center and eventually became head of a Section in the Division of Experimental Rowson KEK, Mahy BWJ. Lactate dehydrogenase-elevating
Chemotherapy. It is at Sloan-Kettering where Riley did his seminal work on LDH virus. virus. J Gen Virol. 1985; 66: 2297-2312.
page 293

1960 Vernon Riley, Kenneth Rowson discovery of lactate dehydrogenase-elevating virus (arterivirus)
In 1960, a 4-year-old girl died of rural encephalitis in a
hospital in La Crosse County, Wisconsin. Four years later,
Wayne H. Thompson, Bernard Kalfayan and Ralph O.
Anslow isolated a novel virus from frozen homogenates
of the childs brain tissue by intracerebral inoculation of
suckling mice. The virus, La Crosse virus, is maintained in
nature through vertical and horizontal transmission cycles
involving the tree-hole breeding mosquito vector, Aedes
triseriatus and primary amplifying hosts (the eastern
chipmunk, Tamias striatus, the gray squirrel, Sciurus
carolinensis, and the fox squirrel, Sciurus niger). Habitat to
support this natural history pattern is found in deciduous
forests widely distributed throughout the central and
eastern United States, with cases reported from more
than 20 states. During an average year, about 75 cases of
encephalitis caused by La Crosse virus are reported to
the CDC. Because the virus is vertically transmitted from
the female mosquito to her eggs, the virus overwinters
in infected mosquito eggs with the high likelihood of
recurrent human disease each summer in highly endemic
areas. Preventive efforts focus primarily on removal of
Wayne H. Thompson Ralph Owen Anslow potential breeding sites such as tree stumps, old tires and
other objects that hold small, stagnant pools of water.
(1922-2000) (1915-1998)
Thompson WH, Kalfayan B, Anslow RO.
Isolation of California encephalitis group
virus from a fatal human illness. Am J
Epidemiol. 1965;81:245-253.

La Crosse virus
suckling mouse brain, thin section electron microscopy
page 294

1960 Wayne Thompson, Bernard Kalfayan, Ralph Anslow discovery of La Crosse virus
Frank Dixon (1921-2008) William Weigle (1927-2001) Joseph Feldman (1916-1995)

page 295 Michael Oldstone Richard Lerner LCM virus immune complex glomerulonephritis
1960> Frank Dixon, others founding of immunopathology, and the role of viruses in immune complex diseases
Cedric Mims Neal Nathanson Richard Johnson Bernard Fields Gerald Cole

Robert Blanden Ashley Haase Diane Griffin Michael Buchmeier Rafi Ahmed

Francisco Gonzlez-
Scarano Volker ter Meulen Kathryn Holmes Geoffrey Smith Harriet Robinson
page 296

1960> Cedric Mims, Neal Nathanson, others founding of the modern era of viral pathogenesis research
Robert Merrifield received his PhD in chemistry
from the University of California Los Angeles in
1949. He then moved on to the Rockefeller Institute
for Medical Research, where he remained for the
rest of his life. In 1963, he published a seminal paper
that launched the method of solid-state synthesis, in
which reactions are carried out on a solid support
rather than in solution. The centerpiece of his
insight was that important biological molecules
such as peptides and proteins were linear polymers
and that one could thus attach one end to a matrix
during their synthesis. The method was initially used
for the synthesis of peptides and proteins; more
recently, it has found wide use in the synthesis of
oligonucleotides, carbohydrates, and even for the
construction of complicated organic molecules.
Initially, his method of solid-state synthesis received
less-than-enthusiastic support from chemists who
were used to a rigorous analysis of the products from
each step in the long series of reactions used to build
up the molecule of interest. By contrast, the solid-
state method did not allow analysis at each step; only
the finished product could be analyzed after it was
cleaved from the support. The success that led to the
acceptance of Merrifields process was his synthesis
in 1969, together with Bernd Gutte, of the 124-amino
acid-long enzyme ribonuclease A. The recovery of
active enzyme with a 78% nuclease-specific activity
left little doubt about the fidelity of his method.
In 1984, Merrifield received the Nobel Prize in
chemistry for his development of methodology
for chemical synthesis on a solid matrix. While
Robert Bruce Merrifield (1921-2006) his methods were simple, his grasp of the organic
reaction mechanisms employed in the various
synthesis procedures was deep and innovative.
Merrifield RB. Solid phase peptide synthesis. I.
The synthesis of a tetrapeptide J Am Chem Soc.
1963;85:2149-2154.
Merrifield RB, Stewart JM. Automated peptide
synthesis. Nature 1965;207:522-523.
Gutte B, Merrifield RB. The synthesis of ribonuclease
Robert Merrifield adjusting an early version A. J Biol Chem. 1971;246:1922-1941.
of his peptide synthesizer, 1969
page 297

1960s Robert Merrifield development of solid phase synthesis technology (polypeptides, oligonucleotides, etc.)
Franois Jacob Jacques Monod Andr Lwoff Jacques Monod, Franois Jacob, in the lab, 1960
(1920-2013) (1910-1976) (1902-1994)
Franois Jacob and Jacques Monod built upon concepts derived by Andr Lwoff from studies of bacteriophage lysogeny and conceived the mechanisms responsible for the
transfer of genetic information from the genome to the macromolecule synthetic machinery of the cell. They, as well, conceived the regulatory pathways which regulate the
timing and amount of synthesis of these macromolecules. They developed the operon concept, using as their model the lactose-operon (lac-operon) in which the inducible
enzyme -galactosidase in Escherichia coli is activated and regulated to metabolize lactose. In their experiments the inducer, lactose, served to inhibit the gene that was
regulating the synthesis of -galactosidase. According to their operon model, a set of structural genes in the bacterial genome encode a messenger RNA that is delivered to
ribosomes, which then translate the RNA message and synthesize protein accordingly. Each set of structural genes has its own operator and regulator genes lying upstream to
it the three together represent the operon. The operator gene is the switch that turns on or turns off its set of structural genes, and thus regulates the synthesis of the protein.
In an inducible system, like the lac-operon, the regulator gene encodes a repressor protein. The repressor protein does one of two things: when no lactose is present, the
repressor protein attaches to the operator and inactivates it, in turn, halting structural gene activity and protein synthesis. When lactose is present, the repressor protein binds
to the regulator gene instead of the operator, and thus frees up the operator and permits protein synthesis to occur. With a system such as this, a cell can adapt to changing
environmental conditions, and produce the proteins it needs when it needs them. Extending from the work of Lwoff, bacteriophages were found to contain genetic control
circuits as well, complete with operators, repressors, and structural genes. In its lysogenic state, the bacteriophage remains inactive. When repressor signals are interrupted,
the bacteriophage is activated, starts to grow rapidly and soon lyses the host bacterium and is released to continue its life cycle. When, in 1961, the now-famous Jacob-Monod
operon model was published, Gunther Stent described the paper as one of the monuments in the literature of molecular biology.
Jacob F, Monod J. Genetic regulatory mechanisms in the synthesis of proteins. J Mol Biol. 1961;3: 318-356.
page 298

1961> Franois Jacob, Jacques Monod, Andr Lwoff discoveries of genetic control of enzyme and virus synthesis
(Above) In the lac-operon, the operator (structural) gene is blocked by a repressor protein when
there is no lactose present (a). When lactose is present, it acts as an inducer by binding to the
repressor protein, preventing it from attaching to the operator. RNA polymerase can then bind to
the operator and transcribe the mRNA molecule (b). Three proteins are synthesized. When all of the
lactose has been catabolized, the repressor protein binds to the operator shutting down the operon.
(Top, left) Micrograph of ongoing transcription of ribosomal RNA, illustrating growing primary
transcripts. Begin indicates the 3 end of the DNA template strand, where new RNA synthesis
begins; end indicates the 5 end, where primary transcription is almost complete.
(Bottom, left) Micrograph of ongoing translation, synthesis of polypeptides from mRNA running
through ribosomes.
The molecule which later became known as ribosomal RNA was first hypothesized by Franois
Jacob and Jacques Monod. RNA synthesis by RNA polymerase was established in vitro by several
laboratories by 1965; however, the RNA synthesized by these enzymes had properties that suggested
the existence of an additional factor needed to terminate transcription. In 1972, Walter Fiers proved
the existence of the terminating enzyme.
Roger Kornberg won the 2006 Nobel Prize in Chemistry for his studies of the molecular basis of
page 299
transcription.

1961> Franois Jacob, Jacques Monod, Andr Lwoff discoveries of genetic control of enzyme and virus synthesis
Jacques Miller
Miller JFAP. Immunological function
of the thymus. Lancet 1961;2:748-749.
Miller JFAP. The discovery of thymus function and
of thymus-derived lymphocytes. Immunol Rev.
2002;185:7-14.
The thymus is divided into two histologically defined regions, the cortex and the medulla, each of
which contains several different thymic epithelial cell subtypes. In adults, T-cell precursors enter
the thymus at the cortico-medullary junction, and then begin a highly ordered differentiation
program, which is linked to migration through the thymic stroma. Different thymocyte subsets are
therefore found in the four spatially restricted regions of the thymus: Region 1, the cortico-medullary
junction, the site of entry into the thymus and contains uncommitted progenitors, CD4-CD8- double-
negative 1 (DN1) cells; Region 2, where cells differentiate to the DN2 stage, undergo a proliferative
clonal expansion, and lose B- and natural killer (NK) cell potential; Region 3, where T-cell lineage
commitment and the onset of T-cell receptor (TCR) chain rearrangement occurs; Region 4, where
the transition from DN to CD4+CD8+ double-positive (DP) status occurs. DP cells then migrate back
through the cortex and, having differentiated into either CD4+ or CD8+ single-positive (SP) cells, into
the medulla. Positive selection occurs mainly in the cortex, whereas negative selection occurs mainly
in the medulla. SP cells that have completed the differentiation program egress from the medulla to the
periphery. CEC, cortical epithelial cell; MEC, medullary epithelial cell.
page 300

1961 Jacques Miller discovery of the role of the thymus in cellular immunity
Stoker M, Macpherson I.
Syrian hamster fibroblast
cell line BHK-21 and
its derivatives. Nature
1964;203:1355-1357.

Michael George Parke Stoker


Michael Stoker was the first professor of virology at the University of Glasgow (the first
professorship of virology to be established at a British university), his tenure extending
from 1958 to 1968. It was during this time that he and Ian Macpherson developed the
BHK-21 cell line, one of the most widely used in virology. He also did seminal research on
tumor viruses and rickettsiae. He later became Director of the Imperial Cancer Research
Fund Laboratories, and subsequently President of Clare College, Cambridge. He was made
a CBE in 1974 and was knighted in 1980.
Ian Macpherson also worked with Stoker in the Department of Tumour Virology at the
Imperial Cancer Research Fund, London [now: Cancer Research UK] from 1963 to 1965.
While a faculty member of the Institute of Virology in Glasgow, he worked with Luc
Montagnier among other things, they developed the soft agar method for selectively BHK-21 cells in culture, stained with multiple fluorophors,
growing cancer cells. especially showing the cytoskeleton (actin filaments,
microtubules, etc.)
page 301

1961 Michael Stoker, Ian Macpherson development of BHK-21 cell line


Sydney Brenner
In the early 1960s, it was thought that the genetic code consisted of units of 4 nucleotides, so as to code for
20 amino acids. Sydney Brenner postulated a triplet code based upon theoretical grounds. Francis Crick and
Sydney Brenner later wrote: In a long series of complex experiments, we induced mutations in the DNA
of bacteriophage T4. The mutations changed individual bases in the DNA, knocking out the function of a
crucial phage gene (the B cistron of the rII region). When two or four mutations were together, the gene was
Francis Crick (1916-2004) still inactive, but when three mutations were put together in the same gene, the gene started to work again.
We concluded that the genetic code is a triplet code (three bases code for one amino-acid), and that the
code is degenerate (an amino-acid may be coded by more than one triplet of bases). Separately, we showed
Crick FH, Barnett L, Brenner S, Watts-Tobin RJ. General nature that the code is non-overlapping. As indicated in the diagram, (a) single nucleotide deletions or insertions
of the genetic code for proteins. Nature 1961;192:1227-1232. cause frameshifts and single nucleotide insertions compensate for single nucleotide deletions. Two or four
single nucleotide insertions still give frameshifts, but (b) when there are a total of three nucleotide deletions
or insertions the code is left in frame. This work was done with John Griffith, Leslie Orgel, Margaret Leslie
Barnett, and Richard Watts-Tobin.
page 302

1961 Francis Crick, Sydney Brenner, others discovery of the triplet coding strategy of DNA (bacteriophage)
Many accounts of the history of molecular biology give the
impression that following the discovery of the double helix
structure of DNA, discovery-followed-discovery in short order.
In fact, as John Cairns stated later, many people found it hard to
imagine that biology could be reduced to something so simple. For
some ten years after the discovery of the DNA double helix much
research was done to link its structure to its functional activities
and its replication. In this setting, John Cairns set out to determine
the nature of the DNA of T2 bacteriophage. Two techniques were
available electron microscopy and autoradiography. Cairns
rejected the high resolution of the electron microscopic approach
because he presumed that intact DNA molecules would not fit
on a specimen carrier (grid). So, he turned to autoradiography,
a technique he had used previously. He grew bacteriophage in
the presence of tritiated thymidine, thereby labeling its DNA. He
affixed the bacteriophage DNA to glass microscope slides and
covered the spread out monolayer with stripping film gelatin-
based photographic emulsion sensitive to the beta particles
released by the decay of tritium. The emulsion would record the
image of the DNA molecules after a sufficiently long exposure
(about 2 months). As Cairns admitted, the success of his method,
achieving full extension of DNA molecules was just good luck
(and many replicate experiments). Nevertheless, he found a
sufficient number of DNA molecules to indicate that its fully
extended length was 52m. From this, he calculated that the mass
of T2 DNA as 110x106 daltons. He postulated an uncomplicated
double helix, and a few years later showed that it was circular.
Cairns went on: the Watson-Crick model posed the problem of
how DNA strands could unwind and separate for replication,
without everything getting tangled. At the time, theoretical
considerations of unwinding exercised some of the best minds in
Autoradiograph of replicating DNA the science. Using the same approach, he showed that the DNA
of bacteriophage T2 of the bacteriophage T2 is circular, 700-900m long, and that its
Bacteria infected with bacteriophage T2 were
John Cairns and Salvador Luria labeled with tritiated thymidine (3H-TdR) and later
replication produces a fork as the helix separates and each strand is
at Cold Spring Harbor Laboratory, 1968 duplicated unwinding did not seem a problem. From: Witkowski
lysed. Insert: the circular phage DNA has two forks.
J. Illuminating life: selected papers from Cold Spring Harbor, Vol 1
The segment represented by the double black line
(1903-1969). Cold Spring Harbor: Cold Spring Harbor Laboratory
had completed two replications in the presence of
Press; 1999.
3
H-TdR, whereas the segment represented by the red/
black line had replicated only once in the presence Cairns, JP. An estimate of the length of the DNA molecule of T2
of the 3H-TdR. The density of grains in the original bacteriophage by autoradiography. J Mol Biol. 1961;3:756-761.
autoradiogram was twice as great in the segment
that had completed two cycles of replication in the Cairns, JP. The chromosome of Escherichia coli. Cold Spring
presence of 3H-TdR versus one cycle. Harbor Symposia on Quantitative Biology 1963;28:43-46.

page 303

1961 John Cairns discovery of the nature and length of the circular DNA of the bacteriophage T2
The first illustration ever published in MMWR, 1985
page 304

1961 U.S. Communicable Disease Center publication of CDCs Morbidity and Mortality Weekly Report (MMWR)
J. Heinrich Matthaei Har Gobind Khorana (1922-2011) Robert W. Holley (1922-1993)
and Marshall W. Nirenberg (1927-2010)
The genetic code is the set of rules by which the information encoded in DNA or mRNA is translated into
proteins by living cells. The code defines the relationship between tri-nucleotide sequences (codons), and
amino acids. The vast majority of genes contain exactly the same code/codons, although there are some
variants (e.g., mitochondria employ a genetic code that differs a bit from the standard code). The first
elucidation of a codon was done by Marshall Nirenberg and Heinrich J. Matthaei in 1961. They used a cell-
free system to translate a poly-uracil RNA sequence (i.e., UUUUU...) and discovered that the polypeptide
that they had synthesized consisted of only the amino acid phenylalanine. They deduced that the codon
UUU specifies the amino acid phenylalanine. Nirenberg presented his results at the International
Biochemistry Congress in Moscow in 1961; his findings electrified the audience and led to great activity to
work out the full code. Severo Ochoa showed that the poly-adenine RNA sequence (AAAAA...) coded for
the polypeptide poly-lysine and the poly-cytosine RNA sequence (CCCCC...) coded for poly-proline: that
is, the codon AAA specifies the amino acid lysine, and the codon CCC specifies proline. Using different
copolymers and other kinds of translation experiments the remaining codons were then determined,
mostly be the work of Har Gobind Khorana. Shortly afterward, Robert Holley determined the structure
of transfer RNA (tRNA), the adapter molecule that facilitates the translation of mRNA into protein.
Nirenberg devised the simple but elegant diagram included here it shows the relationship between the
four nucleotides of DNA, taken three at a time, and the 20 amino acids commonly found in proteins.
page 305

1961> Marshall Nirenberg, Har Khorana, Robert Holley, colleagues deciphering of the genetic code
Jordi Casals Richard Taylor James Porterfield Robert Shope Charles Calisher
(1911-2004) (1887-1981) (1924-2010) (1929-2004)
Until the 1980s, all the arboviruses obtained from clinical or field-collected samples that ended up in reference collections derived from rather standardized procedures: (1)
isolation of the virus in mice (infant mice) or cell culture; (2) production of a virus stock; (3) production of antigen(s); (4) preparation of antibody(ies); and (5) deposition of
the virus in reference collections. In the heyday of this activity (~1960-1975), accumulation of such reagents allowed reference centers to identify new viruses, and as well to
determine broad serologic/antigenic relationships and from these data build a classification system. In the course of this work, much information was gathered on phenotypic/
pathotypic characteristics and vector relationships of the viruses and clinical and epidemiologic information on the diseases they caused. All data were linked in a large
catalogue (the latest being Karabatsos N. International catalogue of arboviruses including certain other viruses of vertebrates, 3rd ed. San Antonio: American Society of Tropical
Medicine and Hygiene; 1985). As newer techniques (in particular, polymerase chain reaction and nucleic acid sequencing) were introduced in the 1980s it was remarkable how
well the serologic/antigenic classification system held up the basic structure of what became the taxa of today did not change, but were refined, made more precise. In fact,
the greater emphasis of the earlier arbovirus classification system on the most important human and animal pathogens made it less likely that any would disappear among the
huge numbers of viruses discovered by more modern means, most of which have turned out to be less important in infectious disease practice.
Casals J. Procedures for identification of arthropod-borne viruses. Bull World Health Organ. 1961;24:723-734.
Taylor RM. Purpose and progress in cataloguing and exchanging information on arthropod-borne viruses. Am J Trop Med Hyg. 1962;11:169-174.
Porterfield JS. The basis of arbovirus classification. Med Biol. 1975;53:400-405.
Calisher CH, Shope RE, Brandt W, Casals J, Karabatsos N, Murphy FA, Tesh RB, Wiebe ME. Proposed antigenic classification of registered arboviruses I. Togaviridae,
Alphavirus. Intervirology 1980;14:229-232.
Calisher CH, Karabatsos N. Arbovirus serogroups: definition and geographic distribution. In: Monath TP, editor. The arboviruses: epidemiology and ecology, vol. 1. Boca Raton
(FL): CRC Press; 1988. p. 19-58.
Calisher CH, Karabatsos N, Dalrymple JM, Shope RE, Porterfield JS, Westaway EG, Brandt WE. Antigenic relationships between flaviviruses as determined by cross-
neutralization tests with polyclonal antisera. J Gen Virol. 1989;70:37-43.
American Committee on Arthropod-borne Viruses, Subcommittee on InterRelationships Among Catalogued Arboviruses. Identification of arboviruses and certain rodent-
borne viruses: reevaluation of the paradigm. Emerg Infect Dis. 2001;7:756-758. page 306

1961> Jordi Casals, Richard Taylor, Robert Shope, Charles Calisher, others antigenic classification of arboviruses
The Vero cell line was derived from the kidney of a
normal adult African green monkey (Chlorocebus
aethiops) in 1962, by Yosihiro Yasumura and Yosio
Kawakita, at Chiba University School of Medicine,
Japan. Yasumura wanted a cell line susceptible to
SV40 virus for virus-cancer studies the kidneys
of African green monkeys were commonly and
severely affected by SV40, so this seemed like
a logical starting point. Indeed, the cell line
supported SV40 virus growth to very high titer. The
cell line was supplied to the Chiba Serum Institute,
where it was investigated as a substrate for polio
vaccine production. Yasumura and Kawakitas
paper was not immediately translated into English
and so this incredibly valuable cell line remained
unknown in the west until 1964, when it was taken
to the laboratory of Ned Wiebenga at the NIH
by Bunsiti Simizu, and from there to the Middle
America Research Unit in the Panama Canal
Zone. Its first success there was in the growth of
Machupo virus, the etiologic agent of Bolivian
hemorrhagic fever. Subsequently, the cell line
proved valuable for the cultivation of many viruses:
adenoviruses, herpesviruses, influenza viruses,
paramyxoviruses, measles virus, respiratory
syncytial virus, picornaviruses, poxviruses,
reoviruses, rubella virus, and especially many
arboviruses. The name Vero comes from Verda
Reno meaning green (African green monkey)
and truth in Esperanto.
Yasumura Y, Kawakita Y. Studies of SV40 in tissue
culture. Preliminary steps for cancer research in
vitro. Nihon Rinsho (in Japanese) 1963;21:1201-
1215. Translation in: Simizu B, Terasima T, editors.
Vero Cells Origins, Properties and Biomedical
Applications. Chiba, Japan: Chiba University
School of Medicine; 1988.
Earley EM, Johnson KM. The lineage of the Vero,
Vero 76 and its clone C1008 in the United States.
In Simizu B, Terasima T, editors. Vero Cells
Origins, Properties and Biomedical Applications.
Chiba, Japan: Chiba University School of Medicine;
Vero cell monolayer, normal (top) and infected with herpes simplex virus, 1988.
page 307 resulting in a large syncytium (bottom). Phase-contrast green light microscopy

1962 Yosihiro Yasumura, Yosio Kawakita establishment of the Vero cell line
Lizbeth M. Kraft (1920-2002) Mouse hepatitis virus
Lizbeth Kraft was a true pioneer, specializing in veterinary microbiology and the diagnosis and prevention of mouse intestine, thin section electron microscopy
murine viral diseases. In 1958, she published a seminal paper on the natural history and control of epidemic
diarrhea of infant mice (EDIM, caused by a rotavirus), a disease then prevalent in mouse colonies. The control
concepts contained in this publication led to the invention of the filter-top for mouse cages (early-on referred
to as Kraft tops) that ever since have contributed significantly to the control of murine viral diseases.
In 1962, she described lethal intestinal virus of infant mice (LIVIM); she determined that the agent was
<300m in diameter and was sensitive to heat and ether, but she did not characterize it further. In following
years, several theories were advanced about the nature of the etiologic agent, but it was only in 1979 that
John Hierholzer, Roger Broderson and Frederick Murphy showed that the agent is an enteric strain of mouse
hepatitis virus. Mouse hepatitis virus strains/pathotypes are remarkably diverse the first to be discovered
caused encephalomyelitis in mice it was discovered by F. Sargent Cheever and his colleagues in 1949. Mouse
hepatitis virus represents the most important viral pathogen affecting contemporary mouse-based research.
Kraft LM. An apparently new lethal virus disease of infant mice. Science 1962;137:282-283.
Cheever FS, Daniels JB, Pappenheimer AM, Bailey OT. A murine virus (JHM) causing disseminated
encephalomyelitis with extensive destruction of myelin. 1. Isolation and biological properties of the virus. J
Exp Med 1949;90:181-194.
Mouse hepatitis virus
Hierholzer JC, Broderson JR, Murphy FA. New strain of mouse hepatitis virus as the cause of lethal enteritis club-shaped epithelial syncytia at tips of intestinal villi,
in infant mice. Infect Immun. 1979;24:508-522. H&E
page 308

1962 Lizbeth Kraft discovery of mouse hepatitis virus (coronavirus)


Frontispiece, Cold
Spring Harbor
Symposium on
Quantitative Biology:
Basic Mechanisms in
Animal Virus Biology

Top row (left-to-right): I. Tamm, A. Lwoff, M. G.P. Stoker, R. Top row: G.K. Hirst, H. Koprowski, H. Noll; Y. Ito; F.K. Sanders, C.
Dulbecco, A. Isaacs; F. Fenner, R. Franklin, D. Baltimore. Morgan, P. Cooper, R.W. Schlesinger.
Second row: A. Granoff, L. Levintow, B. Roizman; D.L.D. Caspar, C. Second row: F.K. Sanders; J.S. Colter, T.S. Work; E. Winocour.
Hotchkiss; F.L. Schaffer, B.C. Backus. Third row: J.E. Hotchin; H.M. Temin, B. Ephrussi, G. Attardi; W.
Third row: A. lsaacs, G.K. Hirst, H. Noll, P. Wildy; G. Klein, F. Fenner, Levinson, F. Fenner, A. Chovnick.
L. Frisch; B.R. Burmester, T. Hanafusa, B.L. Seecof. Bottom row: R. Franklin, P.I. Marcus, B.S. Spendlove, E. Wecker, D.
Bottom row: P. Tournier, R.W. Simpson, W. Bernhard; H. Rubin; L. Baltimore, A.F. Graham; T. Hanafusa, D. Baltimore, W.K. Joklik, N.
Sachs, B. Ephrussi. Ledinko, J. Cairns.
The rather prosaic title of the 1962 Symposium belied the extraordinary nature of this meeting. The ability to grow viruses in animal cells in culture
had transformed the field [from the time of the 1953 meeting, which dealt mostly with bacteriophages], producing large amounts of virus for physical
and chemical analysis, as well as providing dynamic systems for the analysis of virus life cycles. Renato Dulbecco marveled that not only was it possible
to devote a whole symposium to viruses, but also that the experiments reported are as sophisticated as those obtained in any other field of biology...
Most of the papers provided descriptions of the various virus-cell combinations being studied. The authors of these papers read like a roll call of those
who would lay the foundations of virology in the coming years There were two papers added to the original program for the meeting. One was a short,
one-page document by Caspar, Dulbecco, Klug, Lwoff, Stoker, Tournier and Wildy proposing and defining a set of terms for use in virology [virion,
capsid, capsomere, nucleocapsid, peplomer, envelope, etc.]. The second was an attempt by Lwoff to devise a standardized and rational classification of
viruses We can see, with the wisdom that comes with hindsight, that this Symposium leaves us with tantalizing glimpses of how research on viruses
was going to progress. From Jan Witkowski, in the prospectus for the volume, Cold Spring Harbor Laboratory Press.
page 309

1962 Cold Spring Harbor Symposia on Quantitative Biology: Basic Mechanisms in Animal Virus Biology
Cold Spring Harbor Symposium on Quantitative Biology: Andre Lwoff: My ambition is to show that the word
Basic Mechanisms in Animal Virus Biology, 1962
virus has meaning Frenchmen, said Paul Valry, deem
possible that a prodigiously diverse ensemble of highly
complex phenomena can be and must be condensed
and finally reduced into a few plain formulae, at the
same time necessary and sufficient. Belonging to an
hyperlogical extrovert nation, I have coined numerous
definitions as if I had really penetrated the essence of
things. [So,] I shall defend a paradoxical viewpoint,
namely that viruses are viruses.

The qualities of the viruses have been used to argue the


question, Are viruses alive? One adroit answer is to
envision viruses, at the edge of life (i.e., in some ways
fulfilling the criteria we use to define life, in some ways
not) note that this notion does not indicate which side
of the edge the viruses sit on!
Lwoff A. The concept of virus. J Gen Microbiol.
1957;17:239-253.
Wilson EB. At the Edge of Life: An Introduction to
Viruses. Report from the National Institute of Allergy
and Infectious Diseases. NIH publication 80-433; 1980.

Andr Lwoff (1902-1994)


Andr Lwoff, Robert Horne, Paul Tournier:
The world, said Paul Valry, is equally threatened with two
catastrophes: order and disorder. So is virology. Whether the
proposed system will decrease or increase the entropy of virology
is not yet clear. The system has not, however, reached the degree of
rigidity or perfection corresponding to a real catastrophe.
page 310

1962 Andr Lwoff, Robert Horne, Paul Tournier first classification of viruses based upon virion characteristics
Aaron Klug Donald Caspar Francis Crick, Donald Caspar, Aaron Klug
holding Keplers platonic model of the solar and Rosalind Franklin, 1955
system from his Mysterium Cosmigraphicum
Donald Caspar: In 1956, Crick and Watson, at Cambridge, predicted that isometric virus particles should be
constructed from identical subunits arranged with cubic symmetry. From the same laboratory, I reported X-ray
crystallographic data providing the first evidence for icosahedral virus symmetry [tomato bushy stunt virus]. Aaron
Klug and his colleagues in London soon showed that other isometric viruses were icosahedral, and suggested
that some general principle might explain a preference for icosahedral symmetry. I began a collaboration with
Klug in 1958 The problem was to explain how to build the virus shells from a large number of identical units
by repeating the same pattern of contact without the constraint of strict equivalence. A key to our solution was
Klugs recognition of an analogy with Buckminster Fullers geodesic dome designs. In 1962, Klug asked me to
join him in writing a paper for a Cold Spring Harbor Symposium that was intended as an introduction to our
major collaborative paper [We] ended up incorporating much of the theory that the major paper was to have
introduced. They never completed the major paper. We introduced three terms in our Citation Classic that
have been useful for describing a variety of structures: triangulation number, to define the possible icosahedral
surface lattice designs; quasi-equivalence, to describe nearly equivalent bonding of identical units; and self-
assembly, to identify assembly processes controlled by the specific bonding of the parts Our quasi-equivalence
Caspar DLD, Klug A. Physical principles in the theory explained icosahedral symmetry and enumerated the possible designs. The beauty of this theory and the
construction of regular viruses. Cold Spring Harbor success of the predictions made it appear that conservation of bonding specificity was a necessity in icosahedral
Symposium on Quantitative Biology 1962;27:1-24. virus architecture. [An exception was found much later, in Caspars lab polyoma virus assembly does not follow
page 311 the original rules.]

1962 Aaron Klug, Donald Caspar discovery of the principles of the icosahedral structure of viruses
John J. Trentin (1918-2005) Human adenovirus 5 in paracrystalline array
nucleus of a cell in culture, thin section electron microscopy

From the 1962 National Cancer Institute Annual Report: The finding of John J. Trentin, Yoshiro Yabe and Grant Taylor at Baylor College
of Medicine, that human adenovirus type 12 induces tumors in hamsters (confirmed by Robert Huebner and Wallace Rowe along with the
demonstration that adenovirus type 18 also is oncogenic in hamsters), together with the confirmation of the oncogenic nature of SV40 virus
and the further characterization of its properties in tissue culture (e.g., growth of virus and transformation with chromosomal changes in
human cells in culture), and the delineation of the nature of the papovaviruses (especially by Joseph Melnick, Hilary Koprowski, and Maurice
Hilleman) represent important highlights of ongoing cancer virus research. In fact, these findings were the hottest news in oncology in the
early 1960s. It was thought that the quest for the cause of human cancers was within reach and that prevention and treatment would follow
directly.
Trentin JJ, Yabe Y, Taylor G. The quest for human cancer viruses. A new approach to an old problem reveals cancer induction in hamsters by
human adenovirus. Science 1962;137:835-841.
page 312

1962 John Trentin, colleagues discovery of the induction of tumors in hamsters by human adenoviruses
Albert S. Cosgrove (1922-2002) & Hiram N. Lasher (1920-2012) Infectious bursal disease virus
negative contrast electron microscopy
In 1957, Albert Cosgrove, while working with Hiram Lasher at the Delaware State Poultry Laboratories, recognized a syndrome
later termed avian nephrosis on a broiler farm near the community of Gumboro, Delaware. The syndrome became known as
Gumboro disease and was characterized by kidney pathology and 1-10% mortality infection also caused immunosuppression
associated with chronic infection. Within three years the disease was recognized throughout the southern United States and
then around the world. The etiologic agent was thought to be a variant of infectious bronchitis virus (coronavirus) and in other
instances it was thought to be a picornavirus or an adenovirus or a reovirus-orbivirus. Studies to characterize the etiologic agent
began in 1962 at the University of Delaware and Auburn University, but it was not until 1976 that H. Nick, D. Cursiefen, and H.
Becht of the Institut fr Virologie, Justus Liebig-Universitt, Giessen, Germany characterized the virus well enough to conclude
that it could not be placed into any recognized taxon. They found that virions were nonenveloped icosahedra, 60nm in diameter,
and had two segments of double-stranded RNA. The virus became the prototype of a new family, the Birnaviridae. The family
now contains many viruses, including several from fish, molluscs, insects and birds.
Cosgrove AS. An apparently new disease of chickens avian nephrosis. Avian Dis. 6:385-389. 1962.
Lasher HN, Davis VS. History of infectious bursal disease in the USA The first two decades. Avian Dis. 1997;41:11-19.
Nick H, Cursiefen D, Becht H. Structural and growth characteristics of infectious bursal disease virus. J Virol. 1976;18:227-234.

page 313

1962 Albert Cosgrove, Hiram Lasher discovery of infectious bursal disease virus (the first birnavirus)
(Right) Rate-zonal vs. density
gradient ultracentrifugation:
1. soluble proteins
2. rough membranes
3. smooth membranes
4. mitochondria
5. nuclei
6. ribosomes and polysomes
7. glycogen
8. RNA
9. poliovirus
10. T3 bacteriophage
11. adenovirus
Norman Anderson developed a
combined rate-zonal / density
gradient ultracentrifugation
system that took advantage of
the unique windows in which
various cell organelles fell and
which could be employed in
separation schemes. He found
that viruses fall in a rather clear
space, the virus window.

Top and side


Anderson NG. Virus isolation in section views
the zonal ultracentrifuge. Nature of a zonal rotor
1963;199:1166-1168. showing
Anderson NG. Zonal centrifuges loading,
and other separation systems. separation
Science 1966;154:103-112. of virus from
Norman G. Anderson (1919-2007) contaminants,
and unloading

Norman Anderson invented the zonal ultracentrifuge at Oak Ridge National Laboratory. His rotors were large, hollow, bowl-shaped chambers. Density gradient forming media
and virus-containing samples were caused to flow into a rotor spinning at low speed (3,000 rpm) and then it was accelerated to maximum speed to effect separation based on
either sedimentation rate or density (or both). This was followed by deceleration to low speed and recovery by displacement of the gradient as isolated fractions. Anderson
took advantage of the expertise at Oak Ridge derived from uranium enrichment in huge gas centrifuges: We built (and sometimes blew up) a lot of centrifuges, but our
rotors became progressively better at separating biological materials. In the early 1960s, Anderson turned to purifying viruses with his ultracentrifuges in collaboration with
Robert Huebner and his human cancer virus program. Anderson thought that the same approach could be used for the purification of human vaccines: Testing these systems
required large quantities of virus [we obtained] a batch of polio vaccine that did not meet FDA standards As it became clear that no human cancer viruses were being
found around which to design a vaccine purification system, I decided that we should work on an existing vaccine that required better purification. At that time, egg-grown
influenza vaccines contained appreciable amounts of egg proteins We approached Eli Lilly about designing a centrifugal system specifically to purify influenza vaccine. The
result was the K-II continuous-sample-flow-with-banding ultracentrifuge. Use of this centrifuge essentially eliminated vaccination deaths from anaphylactic shock and allowed
vaccination under minimal medical supervision. Almost 40 years later, the K-II is still in use for vaccine manufacture around the world, with minimal modifications.
page 314

1962 Norman Anderson invention of rate-zonal ultracentrifugation


After the discovery of herpes simplex
virus in 1919, a number of studies
on genital herpes were carried out
by several European virologists.
On the basis of both clinical and
laboratory studies, it was suggested
that what was called herpes febrilis
was caused by a different virus than
herpes genitalis. However, this work
was largely ignored for many years.
It was only in the 1960s that it was
demonstrated conclusively that
most isolates from genitalia differ
antigenically and biologically from
most nongenital strains. Today, the
viruses can be distinguished by a
variety of laboratory techniques.
Viral isolates commonly recovered
from nongenital sites are now
designated herpes simplex virus1
(HSV1) (5 to 10% of genital
infections are caused by HSV1).
This virus causes fever blisters
(cold sores), gingivostomatitis,
keratoconjunctivitis, encephalitis,
Karl-Eduard Schneweis Andr J. Nahmias Walter Reed Dowdle and skin eruptions above the waist.
The virus associated primarily with
infections of the urogenital organs
is designated herpes simplex virus
2 (HSV2). The recovery of HSV1
mainly from upper body sites and
HSV2 from lower body sites is most
likely a reflection of the principal
mode of transmission of each virus;
however, recent studies suggest that
the two viruses have also evolved to
match the microenvironment of each
target organ/tissue.
Schneweis KE. Serologische untersuchungen zur typendifferen zierung des
herpesvirus hominis. Z Immunitaetsforsch. 1962;124:24-48.
Dowdle WR, Nahmias AJ, Harwell RW, Pauls FP. Association of antigenic types of
herpesvirus hominis with site of viral recovery. J Immunol. 1967;99:974-980.
Nahmias AJ, Dowdle WR. Antigenic and biological differences in herpesvirus
page 315 Herpes simplex virus, cell culture, thin section electron microscopy hominis. Prog Med Virol. 1968;10: 110-159.

1962> Karl-Eduard Schneweis, Walter Dowdle, Andr Nahmias differentiation of herpes simplex viruses 1 and 2
William Graeme Laver (1929-2008) Robert G. (Rob) Webster
Graeme Laver and Rob Websters work with avian influenza viruses can be said to have sprung from a
beach walk, during which they noticed a large number of dead birds along the shoreline. They wondered
whether it was possible that the birds had died from avian flu, and subsequently traveled to a Great Barrier
Reef atoll to collect samples from hundreds of birds. This led to more trips, trips that became famous in
Australian virology circles, and eventually they discovered the genetic link between avian flu and human
flu viruses. They saw the link as also suggesting the possibility that the viruses might reassort and yield
new viruses to which human populations had no immunity. Years later, Webster noted that when they
first found that there was a link, few virologists paid any attention to what they saw as a great danger.
They theorized that pigs and ducks would be fine mixing vessels and then showed that important
reassortment events do occur this way in nature. In recent years the complexity of the reassortment
events, over time and place, has been proven over and over.
Webster RG, Laver WG. Studies on the origin of pandemic influenza. I. Antigenic analysis of A2 influenza
viruses isolated before and after the appearance of Hong Kong influenza using antisera to the isolated
hemagglutinin subunits. Virology 1972;48:433-444; and Laver WG, Webster RG. II. Peptide maps of the
light and heavy polypeptide chains from the hemagglutinin subunits of A2 influenza viruses isolated
before and after the appearance of Hong Kong influenza. Virology 1972;48:445-455. page 316

1962> Robert Webster, Graeme Laver discovery of the link between avian and human influenza viruses
Peter J. Gomatos (1929-2012) Igor Tamm (1922-1995) Reovirus 3, double-stranded RNA

In the early 1960s, Peter Gomatos and Igor Tamm were studying reoviruses, about which little was known other
than that they were found in the respiratory tracts of humans and animals. In 1963, they made the discovery that the
genetic material of these viruses consists of double-stranded RNA. This was the first description of double-stranded
RNA in any biological system, and Gomatos and Tamm soon found double-stranded RNA in another virus, wound
tumor virus of plants. Shortly thereafter, the laboratories of Albrecht Kleinschmidt and of Aaron Shatkin presented
evidence that the reovirus genome comprises several genome segments. Subsequently, many other viruses causing
diseases in humans, animals, insects, and plants were found by various investigators to have segmented double-
stranded RNA as their genetic material and the several genera of the family Reoviridae were thus created.
Gomatos PJ, Tamm I. Animal and plant viruses with double-helical RNA. Proc Natl Acad Sci USA. 1963;50:878-885.
Langridge R, Gomatos PJ. The structure of RNA. Science 1963;141:694-698.
page 317

1963 Peter Gomatos, Igor Tamm discovery of double-stranded RNA in a virus (reovirus)
The 2008 Nobel Prize
in Chemistry was
awarded to
Osamu Shimomura,
Martin Chalfie and
Roger Y. Tsien
for their earlier
discovery and
development
of green fluorescent
protein, GFP

(Below) Neurons, mouse


hippocampus, fluorescing
in many colors via three
GFP-tagged proteins,
allowing tracing of
individual neuronal
processes.Jean Livet, Ryan
Draft, et al. Osamu Shimomura Martin Chalfie Roger Y. Tsien

Intact brain of mouse infected with recombinant Semliki Forest virus Rabies virus carrying GFP gene used to trace the connections of a
carrying a GFP marker protein. John Fazakerley, et al. single neuron in mouse brain. K-K Conzelmann. page 318

1962> Osamu Shimomura, Martin Chalfie, Roger Tsien green fluorescent protein and its use as a tagging tool
First International Conference on
Measles Immunization, 1961.
L-R: Samuel Katz, Ann Holloway,
Kevin McCarthy, Anna Mitus,
John Franklin Enders Thomas Chalmers Peebles Milan Milovanovic, John Enders,
(1897-1985) (1921-2010) Gisele Ruckle, Frederick Robbins,
Ikuyu Nagata.
Following their initial isolation of measles virus in cell culture in 1954, investigators in the laboratory of John Enders at Harvard
conducted the research, development, and initial clinical studies responsible for the licensure in 1963 of a successful attenuated
live-virus measles vaccine. The team included Thomas C. Peebles, Samuel Katz (who spent 12 years in the Enders lab), Kevin
McCarthy, Milan Milovanovic, Anna Mitus, Gisele Ruckle and Ann Holloway. Also in the lab during this period were Carleton
Gadjusek, Ian Gresser and George Miller.
The virus was isolated from blood and throat washings of David Edmonston, age 11, son of a mathematician in Bethesda,
Maryland. Propagation of the virus successively in human kidney cells, human amnion cells, embryonating hens eggs, and
finally chick embryo cell cultures selected for an attenuated variant virus that when inoculated into susceptible monkeys by
Kevin McCarthy proved immunogenic without causing viremia or overt disease (in contrast to an early kidney cell-passaged
virus, which in similar monkeys produced viremia with illness mimicking human measles).
Clinical studies in children by the Enders group and then by collaborating investigators in many sites established the safety,
immunogenicity and efficacy of the vaccine. The Edmonston strain measles virus became the progenitor of vaccines used
throughout the United States and many other countries.
Katz SL. John F. Enders and measles virus vaccinea reminiscence. Current Topics in Microbiology and Immunology
2009;329:3-11.
page 319
Samuel L. Katz
1963 John Enders, Thomas Peebles, Samuel Katz, Kevin McCarthy, colleagues development of measles vaccine
Maurice Hilleman Harry Meyer Jr. (1928-2001) Stanley Plotkin Michiaki Takahashi
(1919-2005) and Paul Parkman

Vaccine coverage, preschool age children, U.S.

Anne Gershon Ann Arvin Walter A. Orenstein


When the U.S. Department of Health and Human Services highlighted the top 10 achievements in public health in the
20th century, vaccination was listed first. This success was the result of major scientific discoveries, collaboration between
the public and private sectors of medicine and public health, initiatives of pharmaceutical companies, and the dedication
of numerous people in the immunization community. The global situation is also worthy of much praise. This subject is
too large to be covered here, but as a foundation subject in the history of virology it deserves much prominence. page 320

1963> Maurice Hilleman, others development, promulgation and commercialization of pediatric viral vaccines
The Trinidad Regional Virus Laboratory was established in Port of Spain in 1953 by
the Rockefeller Foundation. The labs first director was Wilbur Downs who served
until 1961; he was succeeded by Leslie Spence. Large-scale surveys were conducted,
searching for viruses in humans as well as domestic and wild animals and arthropods.
At the time the lab was established there were a number of common but unidentified
infectious diseases seen in Trinidad; over the years the etiologic agents of many of
these were found. A semi-permanent bush camp was set up at Bush Bush Wildlife
Sanctuary in the Nariva Swamp in southeastern Trinidad and a large tower was built
in the Vega de Oropouche rainforest near Sangre Grande, with platforms at 60, 90
and 120 feet, to the top of the forest canopy. From early on there was a great interest
in bats H. Metivier and J. L. Pawan were early investigators of bat rabies, especially
vampire bat rabies. A government program to control vampire bats by shooting,
netting and trapping started in 1931. This program provided the lab with a steady
supply of correctly identified bats belonging to many different genera and species 58
bat species were recognized in Trinidad at the time. In the course of the bat rabies
research program in the lab, Tacaribe virus was discovered in 1956 and described in the
literature in 1963. It was isolated from Artibeus spp. bats (the genus comprises the fruit
bats of the Americas) and from mosquitoes. It was at first thought to be an arbovirus,
but in an addendum to the first paper it was noted that Robert Shope had just shown
the virus to be antigenically related to Junin virus, the etiologic agent of Argentine
Wilbur George Downs Thomas H. G. Aitken hemorrhagic fever. Tacaribe virus became a charter member of the family Arenaviridae
(1913-1991) (1913-2007) when it was created in 1970.
Downs WG, Anderson TR, Spence L,
Aitken THG, Greenhall AH. Tacaribe
virus, a new agent isolated from Artibeus
bats and mosquitoes in Trinidad, West
Indies. Am J Trop Med Hyg.
1963;12:640-646.
Tacaribe virus
Vero cells
1960s

Charles R. Anderson Leslie Spence Wilbur Downs, field work,


page 321 (1915-1984)
Trinidad, 1950s

1963 Wilbur Downs, Charles Anderson, Leslie Spence Tacaribe virus (the first western hemisphere arenavirus)
Seiichi Matsumoto (1918-1983) Rabies virus, mouse neuron, thin section electron microscopy, 1961
After their first description by Adelchi Negri in 1903, the nature of the prominent intracytoplasmic inclusion bodies (Negri bodies) seen in rabies virus infected neurons was
intensely questioned, early on based upon some quite bizarre notions. That they are virus-specific was first shown using immunofluorescence microscopy in 1959. Some of
the first attempts to resolve the question of their nature using electron microscopy also led to bizarre, incorrect notions. This was all settled by the beautiful work of Seiichi
Matsumoto and Kaneatsu Miyamoto, who in the early 1960s studied infections in mice caused by street (Pasteurs term for wild type virus) and fixed (laboratory adapted) virus
strains. They showed that the eosinophilic (pink/magenta) matrix of Negri bodies is made up of masses of excess viral nucleocapsid, at first rather loosely aggregated (hardly
visible by light microscopy) and later densely packed (the classic inclusions seen by pathologists). The basophilic (blue) large and small granules seen within matrices were seen
to be entrapped masses of virus and degenerated cellular organelles. They also studied the ultrastructural pathology of street vs. fixed virus strain infections and showed that
the main difference was in the cellular damage that occurs with fixed viruses, rather than anything fundamentally different in viral morphogenesis. Everyone who has followed
in the study of rabies pathogenesis using the magnification afforded by the electron microscope has marveled at the terrible beauty of the infection, especially in the largest
neurons in the nervous system.
Matsumoto S. Electron microscope studies of rabies virus in mouse brain. J Cell Biol. 1963;19:565-591.
Miyamoto K, Matsumoto S. The nature of the Negri body. J Cell Biol. 1965;27:677-682;
and Comparative studies between the pathogenesis of street and fixed rabies virus infection. J Exp Med. 1967;125:447-456. page 322

1963 Seiichi Matsumoto, Kaneatsu Miyamoto morphology/morphogenesis of rabies virus, nature of the Negri body
Anthony Epstein (abridged): In 1961, Denis Burkitt came to London
and gave his first talk outside Africa at Middlesex Hospital on the new
lymphoma, entitled The commonest childrens cancer in tropical Africa. A
hitherto unrecognized syndrome. I saw the title on a notice board and for
reasons I am to this day unsure about, I attended and was electrified from
the start by the strange nature and rapid progression of the tumour, and
its epidemiology which seemed to show that its distribution depended on
temperature and rainfall. I postulated that a climate-dependent arthropod
vector might be involved and resolved to investigate the tumour for unusual
viruses. I began with discussions with Denis Burkitt; it was agreed he would
make biopsy samples available to me. For almost two years the standard
techniques for virus detection current at the time were applied to the
lymphoma samples with uniformly negative results. I recruited Yvonne
Barr and Bert Achong to join the project in 1963; that year, a biopsy sample
arrived as usual, floating in guinea pig serum transit fluid, but unusual in
that the fluid was cloudy. At this point there was a feeling that we could
leave the lab for the weekend since it was assumed that the specimen was
contaminated. But instead of going home I put a drop of the transit fluid on
a slide and examined it as a wet preparation under the microscope. Rather
than seeing bacteria I was astonished to find that the cloudiness was due
to large numbers of round, viable looking, free-floating tumour cells. The
cells were set up in suspension culture: shortly afterwards the first BL-
derived continuous cell line designated EB1 grew out. As soon as material
could be spared from the BL-derived cell line, it was processed for electron
microscopy. I was exhilarated to observe unequivocal virus particles in the
very first grid square to be searched. I was extremely agitated in case the
specimen might burn up in the electron beam; I therefore switched off,
walked round the block in the snow wearing only my lab coat, and returned
only when somewhat calmer to take electron micrographs of what I had
seen. I recognized the virus at once as having the morphology of the herpes
group with which I was familiar. There was no means of knowing which
Michael Anthony Epstein Bert Achong herpesvirus it was, although it did seem extraordinary that a member of the
(1928-1996)
herpes family was producing virions in a cell line but was so biologically
Yvonne Barr inert as not to be causing destruction of the cultures. It was indeed fortunate
that the work on BL cells and the search for a virus was going forward in
Epstein MA, Achong BG, Barr YM. Virus
a laboratory where the electron microscope, a rather rare tool at the time,
particles in cultured lymphoblasts from Burkitts
was in daily use, as otherwise its biological inertness could well have left the
lymphoma. Lancet 1964;1:702-703.
agent undiscovered. In this connection it is worth noting that EBV appears
Epstein MA. The origins of EBV research: to be the first virus to have been found solely by electron microscopy.
discovery and characterization of the virus. In: I rapidly set about reporting the discovery with Dr. Achong and Miss
Robertson ES, editor. Epstein-Barr Virus. Saffron Barr the paper became a Citation Classic in 1979. It became imperative
Walden, UK: Horizon Scientific Press; 2005. to have our work repeated in some other laboratory; two leading British
herpes virologists were approached but neither was interested. I therefore
Human lymphoid cells, EBV capsid
contacted Werner and Gertrude Henle in Philadelphia, thereby initiating
antigen immunofluorescence,
their long-term contributions to our understanding of EBV.
page 323 from Paul Feorino, CDC
1964 Michael Epstein, Bert Achong, Yvonne Barr Epstein-Barr virus and its association with Burkitts lymphoma
Citation Classic (Current Contents 1990;33:24-25): Feline leukaemia was shown to
be transmissible experimentally in cats using cell-free extracts of lymphosarcoma
tissue from a spontaneous field case This was the first transmission of a spontaneous
mammalian leukaemia. William Jarrett: From the start of this century, it had been
known that fowl leukaemia was transmissible and caused by a virus. Somehow this
finding was not thought to be germane to mammals... I had worked for some years
in human pathology and had become interested in haematopoietic tumours. When I
returned to veterinary hospital work, I was surprised to find so many neoplasms of this
type, and I was prompted to carry out a survey in Glasgow. Surprisingly, this showed
that leukaemia appeared to be about five times more common in cats than in humans.
I therefore cooperated closely with a local veterinarian, Harry Pfaff [We] started to
find as many of these as possible in order to get fresh material for passage, electron
microscopy, and culture. Here we had a stroke of luck. One of his clients was a dear,
but slightly crazy, old lady who lived alone in a large house with 100 cats. She had
taken these in, off the streets, as strays. Her feline philanthropy ran to bed and board,
but not to sex. So she had my friend neuter them all, toms and tabbies alike. This was
tough for the cats but excellent for us as it meant that all of these animals were outbred
and unrelated. In the course of a year or so, he submitted to me eight cases of different
kinds of leukaemia from this household. This was very striking evidence for horizontal
transmission of an infection, and so I decided to set up a transmission experiment
from this source material We had no grant and no other money. Mary F. Stewart, a
veterinary surgeon from Cornell University, had recently settled in Scotland (because of
a man and mountaineering) and had an isolated cottage in the middle of a golf course.
She was keen to look after the cats (for free) As the first year was drawing to a close,
we wondered if we had failed, when suddenly a cat died, but the material was unsuitable
for passage. Shortly afterwards, another cat became ill and this time we diagnosed
leukaemia on a routine blood count. Material was taken from this animal and passaged
again. I was nail-biting for several days as I had recently been given access for the first
William F. H. Jarrett (1928-2011) time to an electron microscope. Instructed by Elizabeth M. Crawford, my technician
and I prepared the cat material, cut sections, and popped it in the microscope. We
found the virus almost at once, and it was one of the great moments of our lives. Despite
the relative crudity of the techniques at that time, there it was, an incontrovertible
leukaemia virus of perfect morphology, two virions in the first picture showing as much
detail as they ever have since. It is one of my prized possessions still. Soon after that my
colleague Bill Martin and I grew tumour cells in culture and isolated virus from that
After this, I was joined by my brother Oswald, who has since become one of the main
figures in this field. The field study that we immediately carried out showed for the first
time that feline leukaemia-virus infection was widespread in a large city and low on
country farms and that the lower the socioeconomic group the cats were in, the higher
the incidence. But that is another story....
Jarrett WFH, Martin WB, Crighton GW, Dalton RG, Stewart MF. Leukaemia in the cat:
transmission experiments with leukaemia (lymphosarcoma). Nature 1964;202:566-567.
Jarrett WFH, Crawford EM, Martin WB, Davie F. Leukaemia in the cat: a virus-like
particle associated with leukaemia (lymphosarcoma). Nature 1964;202:567-568.
page 324

1964 William Jarrett, colleagues discovery of feline leukemia virus


Karl M. Johnson Patricia Webb (1925-2005) Machupo virus
In 1962, Karl Johnson and Ron MacKenzie of the Middle America Research
Unit (a unit of the NIH), located in the Panama Canal Zone, traveled to Bolivia,
and ended up investigating a disease killing people in the village of Magdalena.
They immediately found ten patients in hospital suffering from a hemorrhagic
fever. They returned with a larger research team and with biocontainment
equipment and soon determined that the location of the outbreak was actually
the village of San Jouquin. It was later estimated that there were 470 cases and
192 deaths in the area between 1959 and 1965. The work was cut short when
MacKenzie and Johnson developed hemorrhagic fever; they were treated in the
Canal Zone by a physician who had earlier treated Korean hemorrhagic fever
cases. Upon their recovery and presumption that they were now immune, they
returned and working with Merle Kuns unraveled the source of the virus (food
contaminated by rodent droppings and urine), the reservoir host the rodent
Calomys callosus and the natural history of the virus. Mouse traps proved
to be an effective control measure. Back in the Canal zone, Patricia Webb,
characterized the virus, named Machupo virus, showing that it is a member of
the family Arenaviridae.
Johnson KM. Epidemiology of Machupo virus infection. III. Significance of
virological observations in man and animals. Am J Trop Med Hyg.
1965;14:816-818.
Ronald MacKenzie Webb PA. Properties of Machupo virus. Am J Trop Med Hyg. 1965;14:799-802.

page 325
Scenes from Bolivia, 1963, from Karl Johnson

1964 Karl Johnson, Patricia Webb, others Machupo virus, the etiologic agent of Bolivian hemorrhagic fever
Michel Bouteille John L. Sever
Subacute sclerosing panencephalitis
(massed measles virus nucleocapsids in the nucleus of a neuron)
Subacute sclerosing panencephalitis (SSPE), a slowly progressive usually fatal disease, was suspected of having a viral etiology from the outset because the microscopic lesions
seen are rather like those produced by several viral infections. Over the years there were many reports of the isolation of various viruses and several unidentified agents. Then,
in 1965, measles or a measles-like virus was implicated not by virus isolation or immunologic means, but by electron-microscopy: Michel Bouteille and his colleagues, of the
Hpitaux de Paris, Institut National de la Recherche Scientifique, described in the brain of a patient particles that resembled the helical nucleocapsids of morbilliviruses. This
finding was confirmed quickly. In 1967, John Connolly and his colleagues reported the finding of astonishingly high levels of measles virus antibody in the sera of patients
with SSPE, as well as the presence of specific immunofluorescent staining for measles virus antigen in brain. Incontrovertible evidence for the role of measles virus came when
Joseph Baublis and Francis Payne maintained brain cells from a patient in culture and demonstrated the development of syncytia and inclusion bodies, and the presence of
measles antigen. However, antigen and cytopathological changes could be passed only with intact cells, and extra-cellular measles virus could not be recovered until Horta-
Barbosa and his colleagues passaged the cells multiple times followed by their co-culture with HeLa cells the basis for this result was later found to be a complex pattern of
defectiveness of the virus. PCR techniques and in situ RNA hybridization have in recent years further substantiated the etiologic role of measles virus in SSPE. Genetic studies
have supported epidemiologic evidence that measles vaccine virus does not cause SSPE. When diagnosed early, SSPE is treated with some success with interferon- and
inosine pranobex (mimics immune-stimulating hormones).
Bouteille MC, Fontaine C, Vedrenne CI, Delarue J. Sur un cas dencephalite subaigue a inclusions, fitude anatomo-clinique et ultrastructurale. Rev Neurol. 1965;113:454-458.
Connolly JH, Allen IV, Hurwitz LJ, Millar JHD. Measles-virus antibody and antigen in subacute sclerosing panencephalitis. Lancet 1967;1:542-544.
Sever JL, Zeman W. Measles virus and subacute sclerosing panencephalitis. Neurology 1968;18(Part 2):1-192. page 326

1965 Michel Bouteille, John Sever, others etiology of subacute sclerosing panencephalitis (measles virus)
Harvey J. Alter

Baruch Blumberg Hepatitis B virus


(1925-2011) Negative contrast electron microscopy
In 1963, while at NIH working with Harvey Alter, Baruch Blumberg discovered Australia antigen (Au), that is an antigen that precipitated in gel when reacted with antibody
in sera from some transfused patients and hemophiliacs. The antigen was named because the first reaction was with the serum of an Australian aborigine. Blumberg went
to Western Australia to collect and test a large number of additional sera, at the time not knowing why the precipitin band had developed between the sera of a hemophiliac
from New York and an antigen from an aborigine from Australia. He had no set views on where this path might lead and later admitted that he had no reason to think of
an association of Au with hepatitis. Using large numbers of stored serum samples from around the world, epidemiological surveys were done to determine the worldwide
distribution of Au. It was very rare in apparently normal populations from the United States, but common in some tropical and Asian populations. The presence or absence
of Au seemed to be a consistent feature of an individual; if it was present on initial testing, then it was present on subsequent testing however, in a Downs syndrome patient
who had originally been negative the Au antigen was present in a later blood sample. This suggested that Aa might be linked to an infection of some sort. The patient was
followed and found to have developed chronic hepatitis between the first and second test. Blumbergs clinical colleague, Alton Sutnick, wrote in the patients chart: SGOT
slightly elevated! Prothrombin time low! We may have an indication of (the reason for) his conversion to Au+. His prediction proved correct. The diagnosis of hepatitis was
clinically confirmed by liver biopsy, and the team now began to test the hypothesis that Au was associated with hepatitis. By 1967, Blumberg, Kazuo Okochi, Alfred Prince,
Alberto Vierrucci, and colleagues reported that Au is involved in the development of hepatitis B, and by 1968-1970 David S. Dane discovered that the Au antigen is actually a
22nm subunit particle of the virus, HBsAg, and is present in the blood of chronic hepatitis patients in very high titer; the virus itself is present also, but in much lower titer.
Blumberg BS, Alter HJ, Visnich S. A new antigen in leukemia sera. JAMA. 191:541-6, 1965.
Prince AM. An antigen detected in the blood during the incubation period of serum hepatitis. Proc Natl Acad Sci USA. 1968;60:814-821.
Dane DS, Cameron CH, Briggs M. Virus-like particles in serum of patients with Australia-antigen-associated hepatitis. Lancet 1970;1: 695-698.
page 327

1965 Baruch Blumberg, Harvey Alter, others discovery of Australia antigen, key to discovery of hepatitis B virus
David Arthur John Tyrrell (1925-2005) June Almeida (1930-2007) Human coronavirus 229E
In 1965, David Tyrrell and M. L. Bynoe isolated a virus, B814, from a person showing typical symptoms of the common cold. This was done by inoculating nasal washings into
human embryonic tracheal organ cultures (no virus growth was seen in usual cell cultures). The presence of an infectious agent was demonstrated by inoculating the medium
from the organ cultures intranasally in human volunteers; colds were produced in most of the subjects. At about the same time, D. Hamre and J. J. Procknow propagated in
tissue culture a virus with unusual properties from throat washings obtained from medical students with colds it was called 229E. And, while working in the laboratory of
Robert Chanock at the NIH, Kenneth McIntosh and his colleagues recovered several strains of virus, the series called OC and OC43 chosen as representative, from human
respiratory tract washings by using a technique similar to that of Tyrrell and Bynoe. Then, June Almeida and David Tyrrell did electron microscopy on samples from organ
cultures infected with B814, 229E and OC43 viruses and found particles in each that resembled infectious bronchitis virus of chickens, mouse hepatitis virus and transmissible
gastroenteritis virus of swine. This new group of viruses was named coronavirus. It was June Almeidas suggestion: corona denoting the crown-like appearance of the virion
surface projections the group is now the family Coronaviridae.
Tyrrell DA, Bynoe ML. Cultivation of viruses from a high proportion of patients with colds. Lancet 1966;1:76-77.
Almeida JD, Tyrrell DA. The morphology of three previously uncharacterized human respiratory viruses that grow in organ culture. J Gen Virol. 1967;1:175178.
Kahn JS, McIntosh K. History and recent advances in coronavirus discovery. Ped Inf Dis J. 2005;24:223-227.
page 328

1965 David Tyrrell, June Almeida, others discovery of human coronaviruses (B814, 229E, OC43)
Joseph L. Melnick (1914-2001) Wallace P. Rowe (1926-1983)
The discovery of adeno-associated viruses (AAVs) is yet another example of, chance favoring the prepared
mind. In 1965, R. Wayne Atchison at the University of Pittsburgh was researching the viral etiology of
human cancers and while checking by electron microscopy an adenovirus stock before injecting it into
hamsters noticed large numbers of small particles intermixed with typical adenovirus virions. Since the small
particles were uniform in appearance and size (as small as 18nm), and even had a hexagonal shape indicative
of icosahedral structure, Atchison presumed they were a virus. Since the particles were smaller than any
previously known virus, he knew his discovery would be novel if it were a virus. Soon, Atchison and research
groups led by Wallace Rowe at NIH and Joseph Melnick at Baylor recognized that the small particles, which
they initially thought to be subunits of adenoviruses, were actually parvoviruses, now members of the genus
Dependovirus, family Parvoviridae. AAVs are replication-defective and dependent upon helper functions
provided by adenoviruses or herpesviruses for productive replication in mammalian cells. Immediately after
their discovery, AAVs were also isolated from humans, but no association with disease has ever been found.
Atchison RW, Casto BC, Hammon M. Adenovirus-associated defective virus particles. Science 1965;49:754-
756.
Melnick JL, Mayor HD, Smith KO, Rapp F. Association of 20-millimicron particles with adenoviruses. J Bact.
1965;90:271-274.
Hoggan MD, Blacklow NR, Rowe WP. Studies of small DNA viruses found in various adenovirus preparations:
Adeno-associated virus virions and two adenovirus virions
negative contrast electron microscopy physical, biological and immunological characteristics. Proc Natl Acad Sci USA. 1966;55:1467-1472.
page 329

1965 Wayne Atchison, Joseph Melnick, colleagues discovery of adeno-associated viruses (parvoviruses)
Robin C. Valentine (1928-1968) Helio Pereira (1918-1994) Erling Norrby
In the early 1960s investigation of viruses was based primarily on their immunological properties. Helio Pereira
at the National Institute of Medical Research at Mill Hill, London, Erling Norrby at the Karolinska Institute in
Stockholm and Harry Ginsberg at Columbia University in New York City were at the forefront of research on
adenoviruses and their antigens. The relationship of each antigen to the icosahedral structure of the viruses
was not apparent and it was only when the isolated antigens were examined by negative contrast electron
microscopy that their nature was understood. At Mill Hill, this work was done by the great microscopist, Robin
C. Valentine. Similar work was soon done by Norrby at the Karolinska Institute. One of the antigens, the group
specific antigen, was tied to the major subunit of the virion capsid, the hexon; another was tied to fibers with
one small and one large knob at either end; and the third was tied to fibers with a small knob at one end only.
At first, it was not apparent whether these fibrous structures were derived from virions or not. The key to the
puzzle lay in the visualization for the first time of adenovirus virions at such resolution as to show fibers with
knobs protruding from each of the twelve vertices of the icosahedral structure. The three antigens were renamed
hexon, penton and fiber, respectively. Tragically, Robin Valentine (1928-1968) died a few years later. Pereira and
his colleagues crystallized the hexon this was the first animal virus protein to be crystallized and studied it
by x-ray crystallography. Today, the adenovirus virion is seen as a very complex structure, built from about 40 Adenovirus virions
different polypeptides, but there are still mysteries, especially concerning structures in the interior of the virion. negative contrast
Valentine RC, Pereira HG. Antigens and structure of the adenovirus. J Mol Biol. 1965;13:13-20. electron microscopy
Norrby E. Structural and functional diversity of adenovirus capsid components. J Gen Virol. 1969;5, 221-236.
page 330

1965> Robin Valentine, Helio Pereira, Erling Norrby discovery of adenovirus structural / functional elements
Graphic art from Russell Knightly; used with permission
Bernard Roizman Herpes simplex virus infection of a cell
Bernard Roizman, of the University of Chicago, has been a leader in virology research since the 1960s. At first, he pioneered in herpes simplex virus (HSV) biology, and as time
went by he came to be recognized as the leading authority in nearly every area of HSV research. One of Roizmans most important early contributions was the identification of
a viral gene that is responsible for HSV neurovirulence. He was one of the first investigators to apply molecular tools to epidemiological studies of viral disease; in doing so he
became responsible for the widely used (and sometimes abused) term, molecular epidemiology. He did this in a study of an outbreak of HSV infection in a newborn nursery, a
setting where infection is often associated with severe disease. This was done using restriction endonuclease cleavage profiles (fingerprinting) of isolates to trace the spread of
virus from one infant to another. This was based on the observation that no two epidemiologically unrelated isolates of HSV had identical restriction enzyme profiles. Roizman
and his colleagues showed that the nursery outbreak was caused by two different virus strains derived from two different adults. Roizmans work then turned to molecular
virology, per se; he determined the structure of the viral DNA and defined the basic molecular mechanisms of HSV replication. His laboratory also conducted extensive
investigation into the identification of many of the more than 90 HSV genes and the functions of the viral proteins encoded by them. As this information mounted, he was able
to study the mechanisms by which HSV causes disease and the mechanisms involved in the host response to acute infection and chronic recrudescent infection.
Buchman TG, Roizman B, Adams G, Stover BH. Restriction endonuclease fingerprinting of herpes simplex virus DNA: a novel epidemiological tool applied to a nosocomial
outbreak. J Infect Dis. 1978;138:488-498.
Roizman B, Buchman TG. The molecular epidemiology of herpes simplex viruses. Hosp Pract. 1979;14:95-104.
page 331

1965> Bernard Roizman seminal studies of the molecular biology and replication of herpes simplex virus
In 1960, Peter Nowell, created a new field, that of
cellular immunology; he made the startling discovery
that an extract from the kidney bean containing a
molecule termed phytohemagglutinin (PHA) could
stimulate lymphocytes to divide. He would never
have made his discovery had he not absentmindedly
left PHA-separated plasma enriched for leukocytes
in the incubator over a weekend. PHA transformed
small lymphocytes into large blastic cells and
stimulated their division. The mechanism responsible
for this remained obscure until 1965, when there
were two reports of an activity found in the culture
media of stimulated lymphocytes that promoted their
proliferation. Termed blastogenic factor by Shinpei
Kasakura and Louis Lowenstein, and Julius Gordon
and Lloyd D. McLean, this activity was due to IL-2, but
it would be another 15 years before this activity was
ascribed to a single molecule. Although IL-2 was the
first soluble, endogenous cytokine to be discovered it
was not named IL-1; but that is a long story, too long
for here. Subsequent to the discovery of IL-2, numerous
reports confirmed the basic observation; however,
further progress awaited Steven Gillis and his colleagues
and their first success in growing T cells in the lab in
1978. They found that T cells use up the IL-2 in the
medium and as they run out they rapidly die. After
being purified by Kendall Smith and his colleagues, the
IL-2 molecule was found to be a variably glycosylated
Kendall Smith MW15,500 protein. IL-2 was also the first cytokine to be
cloned and expressed from a cDNA library. Today, there
are more than 50 human cytokines, and many more in
other animals.
Nowell PC. Phytohemagglutinin: an initiator of mitosis
in cultures of normal human leukocytes. Cancer
Research 1960;20:462-468.
Kasakura S, Lowenstein L. A factor stimulating DNA
synthesis derived from the medium of leukocyte
cultures. Nature 1965;208:794-795.
Gordon J, Maclean LD. A lymphocyte-stimulating factor
produced in vitro. Nature 1965;208:795-796.
IL-2 Gillis S, Ferm MM, Ou W, Smith KA. T cell growth
factor: parameters of production and a quantitative
microassay for activity. J Immunol. 1978;120:2027-2032.
page 332

1965> Shinpei Kasakura, Kendall Smith, others discovery and characterization of the first cytokine, IL-2
Leonard Hayflick began his career in cell culture science with a
post-doctoral fellowship at the University of Texas Medical Branch at
Galveston under the distinguished cell biologist Charles M. Pomerat;
he then spent several years at the Wistar Institute in Philadelphia
with his colleague, Paul Moorehead, where in the early 1960s they
developed the first human diploid cell strains, including the most
widely used of these, WI-38. In 1962, Hayflick discovered that,
contrary to the belief of the day, cultured normal human and animal
cells have a limited capacity for replication. This discovery, known
as the Hayflick limit, overturned a dogma that existed since Alexis
Carrels work early in the 20th century that claimed that normal
cells were immortal and would proliferate endlessly in culture.
Hayflick demonstrated for the first time that mortal (normal) and
immortal (malignant) mammalian cells were different. Hayflick:
A cell strain is a population of cells derived from animal tissue,
subcultivated more than once in vitro, and lacking the property
of indefinite serial passage while preserving the chromosomal
karyotype characterizing the tissue of origin. Conversely, a cell line
is a population of cells derived from animal tissue and grown in
vitro by serial subcultivations for indefinite periods of time with a
departure from the chromosome number characterizing its source.
Hayflick produced the first oral polio vaccine made in a continuously
propagated cell strain. WI-38 is now also used for the production
of rubella, measles, varicella-zoster, mumps, rabies, adenovirus and
hepatitis A virus vaccines. Over one billion people have received
vaccines produced in WI-38 or derivative cells. Hayflick: I received
phone calls from the White House during the Reagan and first Bush
Leonard Hayflick administrations from staffers saying, We finally tracked you down.
We understanding that you are the one who developed the cell
strain derived from an aborted human fetus, and we want to confirm
something we simply cannot believe is going on. And that is that
vaccines have been produced in those cells and are available for sale
in the United States. And my reply was, Where the hell have you
WI-38 cell: Fluorescence been for the past twenty years?
image of a cell stained
Hayflick L, Moorhead PS. The serial cultivation of human diploid
with multi-spectral cell strains. Exp Cell Res. 1961;25:585-621. [This paper describes the
probes (tubulin, nucleic isolation and characterization of 25 strains of human fetal diploid
acid and Golgi markers) fibroblasts, WI-1 to WI-25.]
[ATCC CCL-75]
Hayflick L, Plotkin SA, Nobton TW, Koprowski H. Preparation of
poliovirus vaccines in a human fetal diploid cell strain. Am J Hyg.
1962;75:240-258.
Hayflick L. The limited in vitro lifetime of human diploid cell strains.
Exp Cell Res. 1965;37:614-636. [This paper describes studies with
page 333
WI-26, WI-38 and WI-44.]

1965 Leonard Hayflick development of human diploid cell strains (WI-1 through WI-44)
Lac repressor protein tetramer

Mark Ptashne Walter Gilbert

When Walter Gilbert and Mark Ptashne began their careers, Franois Jacob and Jacques Monod had just advanced the theory of gene regulation by repressors. Identifying
repressors became a Holy Grail of molecular biology. An understanding of repressors would reveal how genes are turned on and off in response to hormones, growth
factors, drugs, and other environmental signals. Many scientists searched for repressors, including Jacob and Monod, but in the end Walter Gilbert, working on the lac
operon repressor of Escherichia coli, and Mark Ptashne working on the repressor of bacteriophage , found the first repressors. They showed that repressors are proteins that
specifically bind to a small stretch of DNA and control the expression of the related upstream structural gene(s). Ptashne continued to work on gene regulation and soon made
his most notable discovery: he unraveled the molecular basis of the lambda switch, explaining how the repressor and another regulatory protein interact with the DNA of the
bacteriophage to switch genes on or off in response to environmental signals. Upon infecting its host bacterial cell, bacteriophage decides between two pathways, a lytic
pathway or a lysogenic pathway. When the switch is ON, the virus multiplies exponentially and kills the bacterial cell. When the switch is OFF, lysogeny occurs: the DNA of
the bacteriophage inserts itself into the chromosome of the host bacterium and remains quiescent until the switch is reversed by signals from the environment. Elucidating
the lambda switch opened the field of transcriptional regulation as it is known today and provided the first general model to explain the switching between developmental
pathways that occurs not only in viral and bacterial infections, but also during embryogenesis and cancers in higher organisms, including humans. Shortly after his work on
repressors. Gilbert left the field of gene regulation and went on to develop a new technique for sequencing DNA, for which he received a Nobel Prize in 1980.
Gilbert W, Muller-Hill B. Isolation of the Lac repressor. Proc Natl Acad Sci USA. 1966;56:1891-1898.
Ptashne M, Hopkins N. The operators controlled by the Lambda phage repressor. Proc Natl Acad Sci USA. 1968;60:1282-1287.
Meyer BJ, Kleid DG, Ptashne M. Lambda repressor turns off transcription of its own gene. Proc Natl Acad Sci USA. 1975;72:4785-4789.

page 334

1966> Mark Ptashne, Walter Gilbert identification of repressor genes (bacteriophage, lac operon of E. coli)
Peter Wildy Frank Fenner Richard Ellis Ford Matthews
(1920-1987) (1914-2010) (1921-1995)

page 335

1966 Peter Wildy, Frank Fenner, Richard Matthews, others International Committee on Taxonomy of Viruses
The History of Virus Taxonomy
The earliest experiments involving viruses were designed to separate them from microbes that could be seen in the light microscope and that usually could be cultivated
on rather simple media. In the experiments that led to the first discoveries of viruses at the turn of the 20th century a single physicochemical characteristic was measured,
ultrafilterability. No other physicochemical or other measurements were made at that time, and most studies of viruses centered on their ability to cause infections and
diseases. The earliest efforts to classify viruses, therefore, were based upon perceived common pathogenic properties, common organ tropisms, and common ecological and
transmission characteristics. It was not until the 1930s that evidence of the structure and composition of virions began to emerge. This prompted Frederick Bawden (1941) to
propose for the first time that viruses be grouped on the basis of shared virion properties. Among the first taxonomic groups constructed on this basis were the herpesvirus
group (1954), the myxovirus group (1955), the poxvirus group (1957), and several groups of plant viruses with rod-shaped or filamentous virions (1959). In the 1960s, there was
an explosion in the discovery of new viruses. Prompted by a rapidly growing mass of data, several individuals and committees independently advanced classification schemes.
The result was confusion over competing, conflicting schemes, and for the first but not the last time it became clear that virus classification and nomenclature are topics that
give rise to very strongly held opinions. Against this background, in 1966 the International Committee on Nomenclature of Viruses (ICNV) was established at the International
Congress of Microbiology in Moscow. The committee became the International Committee on Taxonomy of Viruses (ICTV) in 1973. At that time, virologists already sensed
a need for a single, universal taxonomic scheme, that is that all viruses should be classified in a single system. Nevertheless, there was much dispute over the taxonomic system
to be used. Andr Lwoff, Robert Horne and Paul Tournier (1962) argued for the adoption of an all-embracing scheme for the classification of viruses into subphyla, classes,
orders, suborders, and families. Descending hierarchical divisions were to be based, arbitrarily and monothetically, based upon nucleic acid type, capsid symmetry, presence
or absence of an envelope, etc. Opposition to this scheme was based upon its arbitrariness in deciding the relative importance of virion characteristics to be used and upon
the argument that not enough was
known about the characteristics
of most viruses to warrant an
elaborate hierarchy. Nevertheless,
except for its fancy nomenclature,
the Lwoff-Horne-Tournier scheme
was accepted and evolved in the
1970s into todays system. As
genomic sequencing has replaced
many earlier criteria for placing
viruses in taxa, little change
has been necessary this may be
taken as a tribute to the wisdom
of the founders in basing taxa on
very carefully considered data.
As Frank Fenner said in 1974, Taxa
are often built on relationships
that we would like to believe (from
an evolutionary standpoint), but
are unable to prove. At its meeting
in Mexico City in 1970, the ICTV
approved the first two families, 24
floating genera and 16 plant virus
groups. Today, the ICTV keeps
track of more than 6,000 viruses, 1991 Executive Committee of the International Committee on the Taxonomy of Viruses (ICTV)
in about 2,000 taxa (species, genus, Seated (L to R): C. Calisher, S. Ghabrial, A. Jarvis, F. Murphy, C. Fauquet.
family, and order), all based on Standing : M. van Regenmortel, R. Goldbach, B. Mahy, C. Pringle, H. Pereira, L. Berthiaume, G. Martelli, E. Carstens, J Maniloff,
evolutionary relationships. J. Strauss, M. Summers, M. Mayo, A. Gibbs, H. Ackermann, D. McGeoch, A. Della Porta, M. Horzinek.
page 336

1966 Peter Wildy, Frank Fenner, Richard Matthews, others International Committee on Taxonomy of Viruses
William Hadlow Michael Alpers
Photo: Rocky Mountain Laboratories/NIAID/NIH Photo from 1972, New Guinea
In 1957, Carleton Gajdusek went to New Guinea from Melbourne, Australia where he was working with Macfarlane
Burnet (1899-1985); he was to set up a study on child development and disease. Gajdusek met Vincent Zigas, district
medical officer, who introduced him to a strange neurological disorder, known as kuru, which was found only
among the Fore people, who lived in isolation in the eastern highlands (kuru is the Fore word for shiver or shake).
D. Carleton Gajdusek (1923-2008) People with kuru stumbled and twitched and were belligerent, prone to mirth, grinning, and shrieking. They died
Photo from 1957, New Guinea within months. Later studies of the Fore people indicated that the disease was transmitted by ritual cannibalism, in
which women and children ate the brains of those who had died of kuru. After the ritual was abandoned, the disease
slowly disappeared. Gajdusek spent almost a year living with the Fore, recording 200 cases of kuru and collecting
tissue samples for study when he returned to the United States in 1958. At the NIH, he found that victims had many
central nervous system lesions similar to the spongiform lesions seen in patients with Creutzfeldt-Jakob disease.
William Hadlow, a veterinary pathologist, by chance saw an exhibition on kuru at the Wellcome Museum and wrote
to Gajdusek, saying how similar the lesions were to those in sheep and goats with scrapie. Hadlow wrote: In my
letter to the editor of Lancet, I pointed out the striking similarity of kuru and scrapie and suggested that kuru, like
scrapie, might be a transmissible disease expressed after a long incubation period. Gajdusek, Clarence (Joe) Gibbs
and Michael Alpers built a primate facility at Patuxent, Maryland and inoculated nonhuman primates, especially
chimpanzees, with brain tissue. After an incubation period of two years, the chimpanzees became ill and died,
revealing the same spongiform lesions as seen in the human disease. Eventually, other species of nonhuman primates
also developed the same disease, confirming the infectious nature of the etiologic agent the kuru prion.
Hadlow WJ. Scrapie and kuru. Lancet 1959; 2:289-290.
Gajdusek DC, Gibbs CJ Jr, Alpers M. Experimental transmission of a kuru-like syndrome to chimpanzees. Nature
1966;209:794-796.
Frank Macfarlane Burnet (1899-1985)
Gajdusek DC, Gibbs CJ Jr, Alpers M. Transmission and passage of experimental kuru to chimpanzees. Science
Clarence Joseph Gibbs (1924-2001) 1967;155:212-214.
Photo from 1971, Patuxent Primate Center
page 337

1966 Carleton Gajdusek, Clarence Gibbs, William Hadlow transmission of the kuru prion to non-human primates
Rudolf Siegert (1914-1988) Werner Slenczka Gustav Martini (1916-2007)

Marburg virus, rhesus macaque liver


Siegert R, Shu HL, Slenczka W, Peters D,
Mueller G. Zur tiologie einer unbekannten,
von affen ausgegangenen menschlichen
Infektionskrankheit. Dtsch Med Wochenschr.
1967;92:2343-2370.
Gordon Smith CE, Simpson DIH, Bowen ETW,
Zlotnik I. Fatal human disease from vervet
monkeys. Lancet 1967; 2:1119-1121.
Kissling RE, Robinson RQ, Murphy FA, Whitfield
SG. Agent of disease contracted from green
Robert E. Kissling Roslyn Q. Robinson Frederick A. Murphy monkeys. Science 1968;160:888-890.
(~1931-1981) with Senator Ted Kennedy page 338

1967 Rudolf Siegert, Werner Slenczka, Robert Kissling, Frederick Murphy, others discovery of Marburg virus
Marburg Virus and the Beginnings of Civilian High-Containment Laboratories
In 1967, 31 cases of hemorrhagic fever, with seven deaths, occurred among laboratory workers in Germany and Yugoslavia
who were processing kidneys from African green monkeys [Cercopithecus (Chlorocebus) aethiops] that had been imported
from Uganda. A new virus was isolated from patients and monkeys in three laboratories: that of Rudolf Siegert and colleagues
in Marburg, Charles E. Gordon Smith and colleagues at the Microbiological Research Establishment at Porton Down, and
Robert Kissling and colleagues at the U.S. Centers for Disease Control and Prevention (CDC). It was named Marburg virus.
Zoonotic transmission occurred in circumstances where there was close contact between humans and Wmonkeys or monkey
tissues (kidneys, cell cultures derived from kidneys) in the absense of personal protective equipment or safe practices.
From the beginning, it was realized that Marburg virus was dangerous, calling for the best biocontainment of the day. At
CDC, a mobile containment lab was borrowed from the National Institutes of Health; it was housed in a truck trailer (an
18-wheeler), which was set up in the parking lot at CDC. The containment level provided was about what would be called
BSL-2+ today. The main safety feature was that only three people would be at risk, Robert Kissling, Roslyn Robinson and
Frederick Murphy, and each was experienced in safe practices for working with dangerous pathogens. Specimens were
obtained from David Simpson and Gordon Smith (originating from G. May, Frankfurt and W. Hennessen, Marburg); these
were used to develop diagnostic reagents (antigens, antisera, virus stocks) and for morphology, pathology and ultrastructural
pathology studies. Papers were published, complementing the excellent work done by the German and British virologists.
This Marburg virus episode of 1967 was the impetus for the development of permanent biocontainment facilities at CDC and
elsewhere. The model was the first glove port cabinet line, built in the late 1960s at the U.S. Army Medical Research Institute
for Infectious Diseases (USAMRIID) at Fort Detrick, Maryland. It was for military biological warfare defense purposes. At
CDC, a glove port cabinet line lab was constructed in a tin building; it was opened in 1968. Completion of the lab coincided Karl M. Johnson
with the emergence of Lassa fever in West Africa. Work on Lassa virus at
Yale University (Yale Arbovirus Research Unit) was terminated after one
person died and another nearly died. Thomas Monath and others at CDC
set to work to develop diagnostic systems and unravel the natural history
of Lassa virus. Work expanded from an emphasis on diagnostics to the
pathogenesis of several viral hemorrhagic fevers this even involved the
use of small monkeys. All this put a great strain on the glove port lab .
A second lab was completed at CDC in 1979 it housed both glove port
cabinet lines and the first space suit (positive pressure biocontainment
suit) laboratory, invented by Karl Johnson. He stated in an interview: As
technology developed, and the number of instruments grew, it became
prohibitive to build huge stainless steel glove port lines. His inspiration
came from a colleague, Peter Jahrling. Whereas all the dangerous lab work
at USAMRI1D was done in glove port cabinet lines, animals were held in a
negative pressure room serviced by technicians wearing rubber suits with
masks connected to an air hose. One day, Jahrling needed to make some
frozen section tissue slides and could not get the freeze-microtome into a
glove port cabinet. So, he put on one of the animal room suits and brought
the tissues and microtome into the animal room. Johnson saw this: I Above: James Lange and Herta Wulff working
watched him doing that, and when I got back to the CDC, I sat down with through glove ports in CDC Building 8, 1970s
the engineers and said, We need to make a suit lab well wear suits,
and well have coiled hoses in the ceiling for air. All BSL-4 laboratory Right: LuAnne Elliott in the first positive
development since then has followed upon this insight. pressure biocontainment suit, invented by
page 339 Karl Johnson at CDC, 1977

1967> U.S. Communicable Disease Center development of civilian high-containment virology laboratories
Kates JR, McAuslan BR. Messenger RNA
synthesis by a coated viral genome. Proc Natl
Acad Sci USA. 1966;57:314-320.
Kates JR, McAuslan BR. Poxvirus DNA-
dependent RNA polymerase. Proc Natl Acad Sci
Joseph R. Kates Brian R. McAuslan USA. 1967;58:134-141.
Early on, enzymes required for viral DNA synthesis were found to be induced in vaccinia virus-infected cells. DNA polymerase activity was identified in 1962, and in the
following year Brian McAuslan and others reported the presence of thymidine kinase and deoxyribonuclease. These enzymes are present in uninfected cells but they are
located in the nucleus. Because vaccinia virus DNA synthesis occurs in the cytoplasm, the question arose as to whether the virus-induced enzymes are different. McAuslan
showed that the thymidine kinases in uninfected and infected cells are different. These observations were compatible with the idea that the enzymes required for viral
DNA synthesis are encoded by the virus, but it was difficult at the time to see how their transcription from viral genes could take place in the cytoplasm of infected cells.
In eukaryotic cells, synthesis of mRNA, which is transcribed from chromosomal DNA, requires RNA polymerase II and this enzyme is located in the nucleus. In a paper
published in 1967, Joseph Kates and McAuslan showed that virus-specific mRNA is not synthesized de novo and must be present at the time of infection. All available evidence
pointed to the presence of RNA polymerase within the vaccinia virus virion. Thirty years ago this was difficult to conceive because virions were thought to be metabolically
inert and no virion-associated enzyme had previously been described in any animal virus. It was at this stage that Kates and McAuslan published their study on the ability of
vaccinia virus cores to synthesize virus-specific RNA. Kates and McAuslans discovery played a seminal role in studies which led to the detection of virion-associated enzyme
activities in other viruses. Attempts to infect cells with nucleic acid recovered from some RNA viruses proved unsuccessful and the possibility began to be considered that their
genomes may be negative-sense. If so, a transcriptase would be required to act on the genomic RNA to produce a positive sense, complementary copy. Because a mammalian
enzyme with such activity does not exist, it was considered that it must be brought into the host cell by the infecting virus, that is, the transcriptase would have to be a virion-
associated enzyme. Such an RNA-dependent RNA transcriptase was found, in 1968, to be present in reovirus virions (by Aaron Shatkin and Jean Sipe), and two years later in
vesicular stomatitis virus virions (by David Baltimore and Alice Huang), and, in 1971, in influenza virions (by David Bishop, Jack Obijeski and Robert Simpson). Of course, in
1970 an RNA-dependent DNA polymerase was found in retrovirus virions (by Howard Temin and David Baltimore) this enzyme, reverse transcriptase, became so famous
that in some ways the other virion-associated transcriptases became lost in time.
page 340

1967 Joseph Kates, Brian McAuslan discovery of viral DNA-dependent RNA polymerase (vaccinia virus)
Mareks disease is a highly contagious viral neoplastic disease of
chickens. The disease is characterized by the presence of T cell
lymphomas as well as the infiltration of nerves and organs by
lymphocytes. The discovery of Mareks disease virus involves more
individuals than most of the other discoveries described so far in
this book it resembles more the discoveries of recent times. The
discovery began with evidence provided by Peter Biggs and Jim
Payne at the Houghton Poultry Research Station, England, that
Mareks disease and avian lymphoid leukosis were separate entities.
Next was their demonstration that the agent was cell-associated.
These experiences were shared with Ben Burmester and his
colleagues at the USDA Regional Poultry Laboratory, East Lansing,
Michigan, with whom Biggs and Payne had a close relationship.
Next, Tony Churchill was recruited to join Biggs and his Houghton
Group to provide greater expertise in cell culture techniques.
Using the knowledge of the cell-associated nature of the infectious
agent, he inoculated chick kidney cell cultures with tumor tissue
from Mareks disease infected chickens he found a cytopathic
effect proof of the replication of a virus in the cells required
back-titration in chickens. From the character of the cytopathic
effect in cell culture (syncytia, intranuclear inclusions), the etiologic
agent was suspected of being a herpesvirus; Robert Dourmashkin,
of the Imperial Cancer Research Fund, Mill Hill, London, carried
out electron microscopic studies and, indeed, found a herpesvirus.
Biggs initiated correspondence with Robert Huebner at the National
Peter M. Biggs Ben Roy Burmester (1910-2009) Cancer Institute, who expressed disbelief that a herpesvirus could
be oncogenic - he felt strongly that all cancers were associated with
retrovirus oncogenes. Eventually, Huebner became convinced.
In 1967, Biggs visited East Lansing he told Burmester and his
colleagues about the latest findings and learned that they had also
had success in finding the virus, using duck embryo fibroblast cell
cultures. Keyvan Nazerian did the electron microscopy in East
Lansing and found the same herpesvirus. Proof that the herpesvirus
was the causative agent of Mareks disease was difficult because of its
cell-associated nature; conclusive evidence was the demonstration
by the Houghton group that an attenuated variant of the virus
protected chickens from Mareks disease. Mareks disease virus
together with the closely related herpesvirus of turkeys now form the
genus Mardivirus in the subfamily Alphaherpesvirinae of the family
Herpesviridae.
Jzsef Marek
(1868-1952) who Churchill AB, Biggs PM. Agent of Mareks disease in tissue culture.
The complex pathogenesis of Mareks disease in chickens, Nature 1967;215:528-530.
first described the characterized by persistent virus infection, unique virus shedding
disease named for in feather follicle dander, and lethal T cell lymphomas Biggs PM. History of Mareks disease. In: Hirai K, editor. Mareks
him in 1907 Disease. Berlin: Springer; 2001. p. 1-24.
page 341

1967 Peter Biggs, Anthony Churchill, Ben Burmester, others discovery of Mareks disease virus (herpesvirus)
The separation of proteins from complex mixtures, one of the
major challenges in biochemistry in the early 20th century, was first
advanced by Arne Tiselius, who made electrophoretic separations
in unstable liquid columns. In following years, proteins separated by
their electrical charge were stabilized in place using various supports
such as starch blocks, polyacrylamide gels, etc. These methods could
only resolve proteins having distinctive isoelectric points, but it soon
became apparent that most of the mixtures of interest comprised
proteins with closely overlapping isoelectric points. Jacob Maizel
and his colleagues, at Albert Einstein Medical Center in New York,
had been working with protein separations for some time when a
method of solubilizing chloroplasts using urea and SDS (sodium
dodecyl sulfate, an anionic detergent) was seen in the literature by
Bill Joklik, and was incorporated into the polyacrylamide gels in
Maizels ongoing attempts to separate the proteins of poliovirus.
The separation principle in SDS-gel electrophoresis is not protein
net charge, but is sieving in the polyacrylamide gel matrix due to
the unfolded extended conformation of proteins in the presence of
the detergent and urea. This soon led Maizel and his colleagues to
their breakthrough studies, moving from poliovirus, to adenovirus,
and then to reovirus virion proteins. As initially employed, the
method gave sharper bands than earlier methods, and although
there was a sense that the observed resolution was to some degree
correlated with molecular weight, this had not yet been established.
This correlation was finally resolved using known molecular weight
proteins as markers in the same or parallel gels indeed, there was
an inverse correlation between mobility and protein molecular
Jacob V. Maizel, Jr. Ulrich K. Laemmli weight. The next advance was the development of discontinuous
buffer systems and stacking gels by Ulrich Laemmli that further
enhanced the sharpness of protein bands. Polyacrylamide gel
electrophoresis (PAGE) became one of the most enabling methods in
the modern era of protein chemistry and molecular virology. The first
SDS-polyacrylamide gel in the literature was of poliovirus proteins
(the graph here is from the 1965 paper) the sliced gel showed
quite clearly that the virus mRNA was directing the synthesis of four
capsid proteins plus at least ten noncapsid proteins.
Summers DF, Maizel JV Jr, Darnell JE Jr. Evidence for virus-specific
noncapsid proteins in poliovirus infected HeLa cells. Proc Natl Acad
Sci USA. 1965;154:505-513.

The four poliovirus virion proteins, labeled with Maizel JV Jr. SDS polyacrylamide gel electrophoresis. Trends
14
C; cell extracts treated with SDS and urea and Biochem Sci. 2000;25:590-592.
electrophoresed in 10% polyacrylamide gel. Laemmli UK. Cleavage of structural proteins during the assembly of
y-axis: molecular weight, semi-log scale; x-axis: the head of bacteriophage T4. Nature 1970;227:680-685.
relative migration. page 342

1967 Jacob Maizel Jr., Ulrich Laemmli development of SDS polyacrylamide gel electrophoresis of proteins
One of the central problems in virology during the 1960s was
determining the mechanism by which viruses initiate their
replicative functions. Reoviruses were especially mysterious in this
regard because of their dsRNA genomes; dsRNAs were known to
be inactive as messengers (mRNAs). The first clue came from the
work of Kates and McAuslan, showing that vaccinia virus contains
an RNA polymerase this solved the conceptual problem. Shortly
thereafter Aaron Shatkin and Jean Sipe and Joseph Borsa and Angus
Graham showed that reoviruses contain a dsRNA-dependent RNA
transcriptase (polymerase). But, the story became much more
complicated as time went by. The polymerase (3 polymerase) of
reoviruses functions within the confined interior of the viral core
particle, which is only partially degraded upon entering the host cell
cytoplasm. Within the core particle, replication requires recruitment
of one capped, positive-sense strand for each of the 10 RNA genome
segments, along with an equal number of RNA-dependent RNA
polymerase molecules. Synthesis of minus-sense strands and then
full complements of positive strands accompanies this process (the
3 polymerase molecules switch mode to drive each synthetic step).
Nascent positive-strand RNA is capped and methylated by other
core-associated enzymes and then serves as mRNA for translation
of viral proteins it also serves as genomic positive strands for
packaging into virions. The structure of the reovirus core shows
that 3 polymerase molecules lie just inside the inner capsid shell
and close to five-fold vertex positions, where there are RNA exit
pores. Ten of the twelve five-fold exit positions are associated with
a polymerase molecule, probably with a single genome segment
dedicated to each. [Perhaps all 12 five-fold exit sites are used
by Colorado tick fever virus, which is a member of the family
Reoviridae and has 12 genome segments!] This organization suggests
that during transcription the template (minus-sense) strand of the
Aaron J. Shatkin (1934-2012) Jean D. Sipe genomic RNA is pulled through the polymerase, which remains
more or less in place, and that the transcript, as it emerges from
In the models, the polymerase the polymerase, is fed into the pore for delivery outside. It has been
3 (red) is anchored to considered that the polymerase would have to stay tethered at the
the inner surface of the pore for this scheme to work
icosahedral core shell, making
contact at the five-fold vertex Shatkin AJ, Sipe JD. RNA polymerase activity in purified reoviruses.
position. The polymerase is Proc Natl Acad Sci USA. 1968;61:1462-1469.
oriented with its transcript Borsa J, Graham AF. Reovirus: RNA polymerase activity in purified
exit channel facing a small virions. Biochem Biophys Res Commun. 1968;33:895-901.
channel through the inner
capsid shell, suggesting that Tao Y, Farsetta DL, Nibert ML, Harrison SC. RNA synthesis
nascent RNA is passed to the in a cagestructural studies of reovirus polymerase 3. Cell
exterior for translation. 2002;111:733-745.
page 343

1968 Aaron Shatkin, Jean Sipe discovery of dsRNA-dependent RNA polymerase (reovirus)
Werner Henle (1910-1987) Gertrude S. Henle (1912-2006) Diagnostic blood film: differential >50% lymphocytes, 10%
atypical lymphocytes (large, irregular nuclei)
Werner and Gertrude Henle were early pioneers in Epstein-Barr virus (EBV) research. They had worked with Anthony Epstein in 1964, helping to establish the relationship
between the virus and Burkitts lymphoma. By 1966, they had developed an immunofluorescent test to detect EBV viral antigens, which not only facilitated Burkitts lymphoma
research, but had also linked the virus to nasopharyngeal carcinoma. In 1968, the Henles identified EBV as the causative agent of infectious mononucleosis (IM). As Werner
Henle wrote in his Annotated Bibliography: The answer to our search [for an illness caused by EBV] came quite unexpectedly right in our laboratory when one of our
technicians reported ill in August 1967. She was seronegative for EBV and had been a frequent, more or less willing, donor of leukocytes for EBV transmission/transformation
studies carried out by Volker Diehl, a German postdoctoral fellow. Our technician returned to work five days later but developed under our eyes a rubella-like rash. Her
physician had considered either rubella or IM as probably diagnoses. We obtained blood from her for serological tests as well as yet another leukocyte culture, thinking more
of rubella than IM. The heterophil antibody test, however, turned out to be positive. The rash was thus not due to rubella but to ampicillin which she had received and which
for unknown reasons can be the cause of a rash in IM patients. We noted next with astonishment that our technician had developed antibodies to EBV since the last time
we tested her and observed in due course her leukocytes, which formerly never survived in culture, grew this time into a permanent lymphoblastoid cell line with a small
percentage of EBV-producing cells. These were the first exciting clues that EBV was the long sought cause of IM. It was fortunate that we never took our summer vacation in
August since in our absence this event would probably have passed unnoticed. The Henles then found that 100% of 42 serum samples from IM patients had EBV antibodies. In
a subsequent collaboration with James Niederman and Robert McCollum, prospective studies of college students clearly demonstrated that cases of IM were confined to those
individuals lacking EBV antibodies prior to their illnesses. Such antibodies appeared regularly in association with the disease and persisted for years or for life.
Henle G, Henle W, Diehl V. Relation of Burkitts tumor-associated herpes-type virus to infectious mononucleosis. Proc Natl Acad Sci USA. 1968;59:94-101.
Diehl V, Henle G, Henle W, Kohn G. Demonstration of a herpes group virus in cultures of peripheral leukocytes from patients with infectious mononucleosis. J Virol.
1968;2:663-669.
page 344

1968 Werner Henle, Gertrude Henle association of Epstein-Barr virus with infectious mononucleosis
Peter Wildy Joseph L. Melnick Nils Oker-Blom Victor Zhdanov
(1920-1987) (1914-2001) (1919-1995) (1914-1991)
The Four Founding Fathers

Gathering for the Opening Session


First International Congress for Virology
page 345 Helsinki, 1968 (536 registrants from 39 countries)

1968 Peter Wildy, Joseph Melnick, Nils Oker-Blom, Victor Zhdanov the First International Congress for Virology
International Congress for Place and Year of Chairman of Congress President of ICTV
Virology Congress & Editor(s) of Report
First Congress Helsinki, 1968 P Wildy, J Melnick, P Wildy
N Oker-Blom,
V Zhdanov
Second Congress Budapest,1971 J Melnick, P Wildy,
N Oker-Blom
Third Congress Madrid, 1975 P Wildy, J Melnick, F Fenner
N Oker-Blom
Fourth Congress Hague, 1978 P Wildy, R Matthews
J van der Want,
E Norrby, J Melnick
Fifth Congress Strasbourg, 1981 J van der Want R Mathews
Sixth Congress Sendai, 1984 F Murphy R Francki, C Fauquet,
D Knudson, F Brown
Seventh Congress Edmonton, 1987 E Norrby
Eighth Congress Berlin, 1990 M van Regenmortel
Ninth Congress Glasgow, 1993 B Mahy F Murphy, C Fauquet,
D Bishop, S Ghabrial,
Tenth Congress Jerusalem, 1996 R Pettersson
A Jarvis, G Martelli,
Eleventh Congress Sydney, 1999 J Almond M Mayo, M Summers
Twelfth Congress Paris, 2002 R Compans M van Regenmortel,
C Fauquet, D Bishop,
E Carstens, M Estes,
S Lemon, J Maniloff, M Mayo,
D McGeoch, C Pringle,
R Wickner
Thirteenth Congress San Francisco, 2005 H Klenk L Ball, C Fauquet, M Mayo,
J Maniloff, U Desselberger
Fourteenth Congress Istanbul, 2008 G Smith E Carstens
Fifteenth Congress Sapporo, 2011 K Nagata J Mackenzie
Sixteenth Congress Montreal, 2014 T Dermody E Carstens
Meeting of the Executive
Committee and Advisory Council
at the Second International
Congress for Virology, Budapest,
1971
Participants, clockwise from left: H. von
Magnus (Denmark); F. Fenner (Australia);
A.J. Rhodes (Canada); P. Wildy (U.K.);
E. Farkas (Hungary); N. Oker-Blom
(Finland); J.L. Melnick (USA); E. Norrby
(Sweden); G. Ivanovic (Hungary); L. Hirth
(France); J.H. Subak-Sharpe (U.K.);
V. Rennick (USA)

page 346

1968 Peter Wildy, Joseph Melnick, Nils Oker-Blom, Victor Zhdanov The First International Congress for Virology
The Beginnings of the International Congresses for Virology IAMS to proceed at once to form a section on Virology, to include all branches of the
by Joseph L. Melnick (abridged) science, and to arrange for future and regular International Congresses for Virology.
(2) Until such time as the IAMS establishes a Section in Virology with the responsibility of
How did the International Congresses for Virology begin? This question has been holding International Congresses for Virology, the Secretary-General of the International
asked repeatedly, so I was asked to write this article for the record. Virologists had been Congress for Virology is requested to continue in office and to proceed with arranging for
playing key roles in the affairs of the International Association of Microbiological Societies the Second International Congress for Virology in 1971...
many years prior to 1968, the year of the first Virology Congress in Helsinki. They included The scene now shifts to Mexico City in 1970. In view of the fact that virologists
Thomas M. Rivers (1939-47), Macfarlane Burnet (1953-58), Andre Lwoff (1962-70), and provided firm evidence of their existence by having held a successful independent
Victor M. Zhdanov (1970-74), each of whom served, in the years indicated, as President of Congress in Helsinki, and faced with the real possibility of virologists withdrawing from
the Executive Committee, and Sven Gard, who served as President of the 7th Congress for the IAMS to form their own organization, the delegates to the 10th International Congress
Microbiology in 1958. for Microbiology modified their statutes and agreed to the formation of a Section on
Among virologists who attended Congresses of the International Association of Virology. The virologists present at the Mexico City Congress met and organized their
Microbiological Societies (IAMS), in which virological reports were interspersed among Section. The elected officers were Joseph L. Melnick (Chairman), Peter Wildy (Vice-
presentations oriented toward a variety of other disciplines, it became clear that it would Chairman), Nils Oker-Blom (Secretary) and the elected Advisory Council consisted
be desirable to have a separate Section of the IAMS devoted to Virology. But in order to be of S. Gaidamovich (USSR), J. Hidaka (Japan), L. Hirth (France), E. Norrby (Sweden),
recognized by the IAMS, it was necessary to spring fully grown from the sea, as Aphrodite A. J. Rhodes (Canada), F. Fenner (Australia). The delegates voted to hold the Second
perhaps not as beautiful a body, but a functioning one nevertheless. Thus during the International Congress in 1971 in Budapest... aided by a Hungarian Host Committee:
IAMS Congress in Moscow in 1966, a small revolutionary group of workers felt that the G. Ivanovics, E. Farkas, and I. Dmk.
time was ripe for the International Virologists of the World to unite. The group included The Budapest Congress was a huge success, with 984 virologists from 45
Peter Wildy, Victor Zhdanov, Joseph Melnick, and Nils Oker-Blom. The affairs of the countries participating. Included in the Scientific Program of the Congress were five
IAMS at that time were run by a bureaucracy whose main interest had been focused on plenary sessions on topics of general interest, and thirty specialized workshops.
Bacteriology, and the Microbiology Congresses failed to provide sufficient opportunities At the opening ceremony of the VIIIth International Congress for Virology in
for virologists to meet and to discuss their rapidly developing discipline. Berlin in 1990, the Congress paid tribute to the four founding fathers. Unfortunately, Peter
...At the IAMS Congress in Moscow virologists were again disappointed: sessions Wildy and Victor Zhdanov were no longer with us. I was privileged to be present but Nils
devoted to Virology were few and far-apart, so that most fruitful virologic discussions Oker-Blom had previously committed himself to be lecturing in Nairobi.
were held impromptu, often at street corners while waiting for the Congress busses-which I would like to close these reminiscences with a quotation on the subject of
ran on infrequent and irregular schedules. Our group of revolutionaries proposed to the international virology from Peter Wildy, who was one of the leading figures in 20th
Executive Committee of the IAMS that the virologists be allowed to form a Section within century virology, and who played a key role in founding the International Congresses:
the IAMS. And if we were a Section, why not meet separately, preferably at different times Something must be said about International Virology. Up until 1966, virologists were
from the large congresses? The response to our petition was that virologists rightfully provided for by meetings held from time to time in different countries under the auspices
belonged among microbiologists, and could not be a separate body-and furthermore, that of the International Association of Microbiological Societies. These meetings steadily
no significant virologists group existed, so our request was without substance. grew in size and dullness and virologists became more and more disinterested. In 1966,
The Helsinki Congress in 1968 came into being with the aid of an International four mutineers (ringleader: J. Melnick) decided to run an International Virology meeting
Committee that was recruited from 39 countries. In attendance at that Congress were 536 independently. The meeting was successful and followed by other more comprehensive
virologists, each of whom, in accordance with the admission regulations established by programmes in Budapest, Madrid, The Hague and Strasbourg and since then in Sendai,
the International Committee, had to have contributed at least 3 significant publications Edmonton, and Berlin. Several points have become clear. First, it is important for the
to the worlds virologic literature. The fact that so many persons qualified for attendance health of virology and for the interests of virologists that meetings be held separately from
indicated how large and significant the literature of our science had already become. others dealing with microbiological disciplines, in order to attain the maximum chance
I had the pleasure of serving as Secretary-General of the Helsinki Congress. of personal encounter. Second, the breadth of virology is great enough to sustain varied
In my opening remarks I said: Now, this week, by our meetings and discussions of our combinations of interest. Indeed, considerable ingenuity is required to ensure mutually
laboratory work, we shall reaffirm our existence. Speaking not only for myself but also for fruitful exchanges. Third, the heterogeneity of virologists is such that it has often been
each of the participants, I wish to express the appreciation that is owed to Professors Peter questioned whether it is wise to continue with such large congresses. So far, I believe they
Wildy, Victor Zhdanov, and Nils Oker-Blom, with whom I have had the pleasure of serving have been a powerful force of good communication. I hope that they remain so.
on the Convening Committee, to the 39 members of the International Committee, and I believe that the Virology Congresses continue to be a powerful force of good
particularly to Professor Oker-Blom and his colleagues, to the University of Helsinki... communication and I believe that they will be so in the future.
At the end of the weeks program the following resolutions were adopted... (1) Be
it resolved that the members of the First International Congress for Virology petition the Melnick JL. The beginnings of the international congresses for virology.
page 347 Arch Virol. 116;293-300:1991.

1968 Peter Wildy, Joseph Melnick, Nils Oker-Blom, Victor Zhdanov The First International Congress for Virology
Kathleen Danna

Daniel Nathans (1928-1999) Hamilton Smith Werner Arber


In the early 1960s, Werner Arber and Daisy Dussoix, a PhD student, found that bacteria can distinguish between their own DNA and
the foreign DNA of an invading bacteriophage they found that the bacterial DNA is protected by methylation and the foreign DNA is
cleaved and further degraded by specific enzymes. The enzymes doing this cleavage were called restriction and modification enzymes,
and are now known as restriction endonucleases. Arber provided the theoretical framework for all future work and he also isolated the Thomas J. Kelly
first enzyme, EcoBI. Hamilton Smith, Kent Wilcox and Thomas Kelly verified Arbers hypothesis by purifying both bacterial restriction Arber W, Dussoix D. Host controlled
and modification enzymes and showing that they specifically cut DNA and methylated the DNA, respectively. Starting in 1971, Daniel modification of bacteriophage
Nathans and Kathleen Danna pioneered in the application of restriction enzymes to the study of the DNAs of several organisms, starting lambda. J Mol Biol. 1962;5:18-36.
with that of simian virus 40 (SV40). They used the restriction enzyme endonuclease R, discovered by Smith and Wilcox to produce Smith HO, Wilcox KW. A restriction
specific fragments of SV40 DNA and showed that these fragments could be nicely separated from one another by polyacrylamide gel enzyme from Hemophilus
electrophoresis. [We now know that the original preparation of endonuclease R contained two enzymes, the one characterized by influenzae. I. Purification and
Smith, HindII, the second HindIII the SV40 cleavage pattern was the product of both enzymes.] It was Nathans who made the key general properties. J Mol Biol.
intuitive leap that cleavage patterns could be used to produce a physical map of SV40 DNA, as well as indicating the origin of replication 1970;51:379-391.
and the location of specific genes. These pioneering studies set the stage for modern recombinant DNA technology and molecular
biology the photograph of the first published gel provided immediate visualization of just how powerful the combination of restriction Danna K, Nathans D. Specific
endonucleases and gel electrophoresis would be. Today, more than 3,600 restriction enzymes are recognized, representing more than Cleavage of simian virus 40 DNA
250 specific cleavage patterns more than 600 of these enzymes are available commercially. by restriction endonuclease of
Hemophilus Influenzae. Proc Natl
Acad Sci USA. 1971;68:2913-2917.
page 348

1968> Werner Arber, Hamilton Smith, Daniel Nathans, others discovery of restriction endonucleases
In 1969, Robert Huebner and George Todaro proposed the oncogene
hypothesis they proposed that normal cells must contain genes related
to those found in retroviruses, since viral proteins could often be found
in cells of apparently uninfected animals, especially chickens and mice.
Such genes were believed to be transmitted vertically, integrated into the
chromosomes of germ-line cells, and expressed in response to a variety of
stressors. Since some retroviruses were known to be highly oncogenic in
animals, it was also proposed that tumor-inducing genes, e.g., retroviruses
genes (viral oncogenes) might also be transmitted through the germ line as
a consequence of ancient infection. Activation of these endogenous viral
oncogenes by substances we recognize as carcinogens chemicals, radiation,
other viruses could serve to initiate a neoplastic process. Although their
hypothesis that most cancers were caused by expression of retroviral genes
was not correct, their work did lead to the identification of the first retroviral
oncogene, src, and to the realization that these viral genes were derived from
functional cellular genes or proto-oncogenes. They proposed that cellular
proto-oncogenes are precursors of transforming cancer genes. In 1981,
Huebner wrote (abridged): The viral oncogene theory was inescapable
given the years of cumulative evidence culminating in our 1969 report
in PNAS [It was our view] that cancer was the result of derepression
of inherent species-specific endogenous oncogenes often linked in mice
and chickens with endogenous retroviruses. Genetic abnormalities or
deficiencies, environmental carcinogens, and oncogenic viruses were thus
inducers of such oncogenes. Another key factor was our observation that the
endogenous leukemia and sarcoma viruses in chickens, mice, rats, and cats
actually carried movable oncogenes, since src genes could be rescued from
primary hosts and transmitted as transforming oncogenes to cultured cells
of a wide variety of animal hosts, including humans The viral oncogene
hypothesis expressed a new concept in which it was postulated that all
carcinogenesis was mediated through activation of an endogenous host cell
gene present in every vertebrate cell. Recently, it became apparent that the
Robert J. Huebner (1914-1998) oncogene determinants of cancer represented also the required determinants
Symbols for oncogenes are three lowercase for immunoprevention. Vaccines and IgG preparations incorporating
characters in italics. These may be preceded immunity to src and leu gene expressions have recently been used to prevent
by a c or v in roman type indicating virtually all types of cancers in mice and rats, including cancers produced
whether the origin was chromosomal or viral. by chemicals and DNA tumor viruses. These successful immunoprevention
E.g., scr, c-src, v-src. Symbols for the protein studies, we believe, provide excellent models for development of comparable
products of oncogenes are three characters in prevention of cancer in man. Of course, in the years since Huebner and
roman type with the initial letter capitalized. Todaro conceived this grand hypothesis much has changed much of the
E.g., Src. However, one often sees all three hypothesis has been proven wrong and the promise of immunoprevention
letters capitalized, e.g., SRC, or all lower of human cancers by vaccines against any retrovirus antigen is still a work in
case, e.g., src the convention has not been progress.
followed very well. Huebner RJ. Todaro GJ. Oncogenes of RNA tumor viruses as determinants of
George Todaro cancer. Proc Natl Acad Sci USA. 1969;64:1087-1094.
page 349

1969 Robert Huebner, George Todaro development of the viral oncogene hypothesis
Lassa virus, human liver, massive
Sonja Buckley (1919-2005) Jordi Casals-Ariet (1911-2004) hepatocellular necrosis
1 plastic-embedded section, polychrome stain
Events leading to the discovery of Lassa virus in 1969 were dramatic.
Two nurses in a mission hospital in Lassa, Nigeria, died but one
was first flown to a larger hospital in Jos. There, an attending nurse
also became very ill; she was evacuated to Columbia-Presbyterian
Hospital in New York. She survived. Clinical specimens were sent
to the Yale Arbovirus Research Unit in New Haven, where Lassa
virus was isolated by Jordi Casals and Sonja Buckley. Later, while
working with the virus, Casals became very ill and was successfully
treated with convalescent serum from the surviving nurse; however,
a laboratory technician at Yale died. Work at Yale was terminated
and shifted to CDC where Thomas Monath and colleagues further
characterized the virus, unraveled its natural history and prevalence
and discovered its reservoir host, the multimammate rat, Mastomys
(Praomys) natalensis. Today, Lassa hemorrhagic fever is recognized
as an important disease in West Africa.
Lassa hemorrhagic fever field research station, Nigeria, 1972 Buckley SM, Casals J. Lassa fever, a new virus disease of man from
Thomas Monath, Jordi Casals, Kent Campbell, Verne Newhouse, Mission Hospital Nurses West Africa. Am J Trop Med Hyg. 1970;19:680-691.
page 350

1969 Sonja Buckley, Jordi Casals-Ariet discovery of Lassa virus, Lassa hemorrhagic fever
Epizootic diarrhea of newborn calves has for centuries been a
plague of cattle husbandry. The disease attacks calves within
four or five days of birth, causing at times significant mortality
a 30% loss is not uncommon. Attempts at experimental
transmission of the disease had been made from time to time,
but these had given equivocal results, probably because the
etiologic agent(s) is ubiquitous, so that controls as well as
inoculated animals became ill or did not because of maternal
immunity. Charles Mebus, Marvin Twiehaus and their colleagues
first succeeded in transmitting infection in a reproducible way
by inoculating gnotobiotic calves by intraduodenal tube (and
later by feeding) with bacteria-free filtrates of diarrheal feces.
Calves developed diarrhea usually within 24-48 hours, and
65nm diameter virus particles were found in large numbers in
their feces. The virus was described as reovirus-like; the name
rotavirus was coined in 1973 by Thomas Flewett. The virus
was adapted to grow in cell culture, and extensive pathology
and pathogenesis studies were done using histopathology
and frozen-section immunofluorescence of intestinal tissues.
Thomas Flewett, one of the discoverers of human rotaviruses
stated: In the early days of virology, workers on human, animal,
insect and plant viruses were all acquainted with each other and
knew each others work. But with proliferation of laboratories,
diversification of interests, and multiplication of journals, this
happy state has not existed for some time. Oblivious of the work
of Mebus and his colleagues in Nebraska or not appreciating
its importance, groups in 1973 in Birmingham, Melbourne and
Toronto were independently searching for virus particles in feces
or intestinal biopsies of children. this story is continued under
the year 1973, when Ruth Bishop, Ian Holmes, Thomas Flewett,
Charles A. (Chuck) Mebus Peter Middleton and their colleagues discovered the first human
rotavirus.
Mebus CA, Underdahl NR, Rhodes M, Twiehaus M. Calf
diarrhea (scours): Reproduced with a virus from a field outbreak.
Univ Nebraska Res Bull. 1969;233:1-16.
Mebus CA, Underdahl NR, Rhodes MB, Twiehaus MJ. Further
studies on neonatal calf diarrhea virus. Proc Annu Meet US
Rotavirus Anim Health Assoc. 1969;73:97-99.
(top) purified virus, negative contrast
Mebus CA, Kono M, Underdahl NR, Twiehaus MJ. Cell culture
electron microscopy
propagation of neonatal calf diarrhea (scours) virus. Can Vet J.
(bottom) virions assembling at edge
1971;12: 69-72.
of an intracytoplasmic inclusion
body (virus factory), cell culture, thin Flewett TH, Woode GN. The rotaviruses: brief review. Arch
section electron microscopy Virol. 1978;57:1-23.
page 351

1969 Charles Mebus, Norman Underdahl, Marvin Twiehaus discovery of bovine rotavirus (the first rotavirus)
Knowing is not enough; we must apply.
Willing is not enough; we must do.
Goethe
Motto of the Institute of Medicine
U.S. National Academy of Sciences

Members of the Board on Medicine,


as it became the Executive Committee
of the Institute of Medicine in 1970:
Robert J. Glaser, M.D., President pro tem
John R. Hogness, M.D., first President
Walsh McDermott, M.D.
Joseph S. Murtaugh
Ivan L. Bennett, Jr., M.D.
Samuel M. Nabrit, Ph.D.
Charles G. Child III, M.D.
Irvine H. Page, M.D.
Julius H. Comroe, Jr., M.D.
Henry W. Riecken, Ph.D.
John T. Dunlop, Ph.D.
Dwight L. Wilbur, M.D.
Walter A. Rosenblith, Ing. Rad.
Rashi Fein, Ph.D.
Lucile P. Leone, M.A.
Bryan Williams, M.D.
Irving M. London, M.D.
Adam Yarmolinsky, LL.B.
Colin M. MacLeod, M.D.
Albert Einstein Alonzo S. Yerby, M.D.
on the grounds of the National Academy of Sciences Eugene A. Stead, Jr., M.D.
page 352

1970 Irvine Page, others founding of the Institute of Medicine of the U.S. National Academy of Sciences
From the time of Pasteur, rabies virus had been singular antigenically,
genetically and taxonomically singular. In fact, when the African viruses that
were to become the members of the genus Lyssavirus, family Rhabdoviridae,
that is the rabies-like viruses, were first isolated there was no notion of their
relationship to rabies virus. The first two of these viruses were discovered in
arbovirus field projects and so were expected to be members of one of the taxa
housing the arboviruses. L.R. Boulger and James Porterfield isolated a virus from
Eidolon helvum fruit bats at Lagos Island, Nigeria, in 1956; they called it Lagos
bat virus, a possible arbovirus, and distributed it widely to reference centers in
the hope of finding out more about its relationship to other arboviruses. Graham
Kemp isolated Mokola virus from organs of Crocidura spp. shrews in 1968,
although its reservoir host species is still unknown. Later two strains of Mokola
virus were isolated from cases of central nervous system disease in Nigerian
children. Lagos bat virus and Mokola virus were sent to the Yale Arbovirus
Research Unit in 1969,where Robert Shope, using all the classic arbovirus
identification methods confirmed that the two viruses behaved like other
ungrouped arboviruses and that the two viruses cross-reacted by complement-
fixation testing. Shope had been collaborating with Frederick Murphy at the
Centers for Disease Control and Prevention trying to classify the rhabdoviruses
of vertebrates (quite a few had been identified by electron microscopy by this
Robert Ellis Shope (1929-2004) time). The two viruses were found to be rhabdoviruses, but more so, they were
identical to rabies virus in their appearance in infected cells (intracytoplasmic
inclusion bodies Negri bodies and virions budding in association with these
inclusion bodies quite unlike other rhabdoviruses such as vesicular stomatitis
viruses). Murphy telephoned Shope with this exciting news and suggested that
the two viruses be tested against rabies virus this was done immediately and
a serologic relationship was found. A few years later, in South Africa, Courtney
Meredith read about the rabies-related viruses and thought of them when a virus
he isolated from the brain of a man bitten on the lip by a bat did not react in the
rabies immunofluorescence test, although the patient had died of what appeared
to be typical rabies. This virus was found to be another rabies-like virus; it
was named Duvenhage virus. Since then, several other viruses that, except for
political sensitivities, might be called rabies virus have been isolated in Europe,
including the United Kingdom, and Australia. Recently, four viruses genetically
quite distant from rabies virus have been found in bats in central and southeast
Asia. The genus Lyssavirus is rather well populated now, after having only one
member for 66 years.
Shope RE, Murphy FA, Harrison AK, Causey OR, Kemp GE, Simpson DI, Moore
DL. Two African viruses serologically and morphologically related to rabies
Lagos bat virus, hamster brain virus. J Virol. 1970;6:690-692.
thin section electron microscopy Tignor GH, Murphy FA, Clark HF, Shope RE, Madore P, Bauer SP, Buckley SM,
Meredith CD. Duvenhage virus: morphological, biochemical, histopathological
and antigenic relationships to the rabies serogroup. J Gen Virol. 1977;37:595-611.

page 353 Frederick A. Murphy Shope RE. Rabies-related viruses. Yale J Biol Med. 1982;55:271-275.

1970> Robert Shope, Frederick Murphy, others discovery of rabies-like viruses (the genus Lyssavirus)
Frank Fenner (1914-2010) David Ogilvie White (1931-2004)
Frank Fenner, later recalling book writing in 1969-1970: I had always felt somewhat guilty being called a Professor, having
never given a course of lectures (I was never invited to lecture to students in the faculties of the university), and now I was never Medical Virology, First Edition, 1970:
likely to. So I thought that I might occupy the spare time I had, always early in the morning before anyone else arrived and at Frank Fenner, David O. White.
irregular times on most days, to write a textbook on virology for medical students. I persuaded David White, who was by then Medical Virology, Second Edition, 1976:
Professor of Microbiology at the University of Melbourne, who I knew to be a first-class teacher of virology, to be a co-author. Frank Fenner, David O. White.
The first edition, entitled Medical Virology, 390 pages long, was published by Academic Press in 1970. It received excellent Medical Virology, Third Edition, 1986:
reviews and sold very well. M.A. Epstein, writing in Nature, said: Despite the risk of writing a rave notice, I am still filled with David O. White, Frank Fenner.
wonder on reflecting on the seeming ease with which extremely complicated topics have been reduced to an orderly survey of Medical Virology, Fourth Edition, 1994:
the basic facts involved, together with suitable explanations of their significance. This book can be used, therefore, both as an David O. White, Frank Fenner.
introductory textbook and as a very reliable and solid reference book. More than 25,000 copies of the first edition were sold.
Fenner F. Nature, nurture and chance. The lives of Frank and Charles Fenner.
Canberra: Australian National University ePress; 2006. page 354

1970 Frank Fenner, David White publication of book, Medical Virology (currently four editions)
Viral RNA-dependent RNA polymerases were
discovered in the early 1960s in studies of
mengovirus and poliovirus when it was observed
that these viruses were not sensitive to actinomycin
D, a drug that inhibits cellular DNA directed
RNA synthesis. This lack of sensitivity suggested
that there was a virus specific enzyme that could
copy viral RNA from an RNA template. It was
found that in these positive-sense RNA viruses
this enzyme is encoded in the virion RNA and is
transcribed and translated directly. The advantage
of this for the positive sense RNA viruses is that
there is no intermediate stage, so replication can
be quick and often very productive. However,
purified genomic RNA from viruses like vesicular
stomatitis viruses (VSV) is non-infectious, and
this observation led to the discovery that the
genomic RNA is complementary to its mRNA
these are the negative-sense RNA viruses. David
Baltimore, Alice Huang and Martha Stampfer
discovered the gene encoding the RNA-dependent
RNA polymerase and the polymerase itself in
1970. The transcription and replication scheme of
viruses like the VSVs first requires production of
complementary strand RNAs, which are then used
as mRNAs. Even so, there are quite a few differences
in the transcription, translation and replication
schemes employed by the many negative-sense RNA
viruses. In 1973, Alice Huang also suggested that
the virion polymerase of VSV serves its distinctly
dual functions as transcriptase and replicase in
complex fashion. Recently, it has been found that
David Baltimore Alice S. Huang there are two RNA-dependent RNA polymerases in
VSV, each a multiprotein complex, each containing
Baltimore D, Franklin RM. A new ribonucleic acid the viral L and P proteins (the polymerase), but
polymerase appearing after mengovirus infection of each containing different cellular proteins. Thus,
L-cells. J Biol Chem. 1963;238:3395-3400. the mechanism for carrying out the two different
Baltimore D, Huang AS, Stampfer M. Ribonucleic acid functions, transcription and replication, seems
synthesis of vesicular stomatitis virus, II. An RNA to have evolved in the simplest way using two
polymerase in the virion. Proc Natl Acad Sci USA. distinct enzymatic complexes rather than some
1970;66:572-576. mysterious single complex that would have to shift
from one function to the other.

page 355

1970 David Baltimore, Alice Huang discovery of RNA-dependent RNA polymerase in vesicular stomatitis virus
Extracellular mature C particles
thin section electron microscopy
Baltimore D. RNA-dependent DNA polymerase
in virions of RNA tumour viruses. Nature
1970;226:1209-1211.
Temin HM, Mizutani S. RNA-dependent DNA
polymerase in virions of Rous sarcoma virus.
Howard M. Temin (1934-1994) David Baltimore Nature 1970;226:1211-1213.
Through many experiments done over many years, the principles by which genetic information is transferred were demonstrated. DNA provided the information that was
transcribed into mRNA, which was then used for translation into proteins. DNA>mRNA>proteins the central dogma. However, in 1970, it was shown that the central
dogma did not cover everything. In 1961, Howard Temin began gathering evidence that was inconsistent with the central dogma. Temin, working on Rous sarcoma virus
(RSV), felt that the best explanation for the often dormant behavior of the virus was a proviral non-replicating state in infected cells. Since RNA is notoriously unstable,
Temin proposed that the RNA genome of RSV is converted into a DNA provirus. He amassed data suggesting that RSV-transformed cells have a DNA complementary to
RSV genomic RNA. Separately, David Baltimore had been studying the replication of vesicular stomatitis virus (VSV) and found that the its genome is complementary to
its mRNA thus was born the idea of negative sense viruses. One puzzle was how a negative sense virus can initiate infection its RNA must either be copied by a host
cell enzyme or the virus must enter the cell with its own RNA-dependent RNA polymerase. In 1970 Baltimore, with Alice Huang and Martha Stampfer, found the virion-
associated polymerase. This led directly to Baltimores discovery of reverse transcriptase; he hypothesized that other viruses must also solve the problem of initiating infection
by employing a virion-associated polymerase. After being lucky enough to obtain a large amount of Rauscher murine leukemia virus, Baltimores search for an RNA-dependent
DNA polymerase took only a few days. He found that the polymerase activity was ribonuclease (RNase)-sensitive and actinomycin D-resistant, suggesting that the virion-
mediated DNA synthesis employed an RNA not a DNA template. As with VSV, the virion-associated polymerase allowed the Rauscher virus genome to serve as the template
for nucleic acid synthesis without having first to make new protein(s). Temin and Satoshi Mizutani came to the same conclusion at the same time actually, the experiments
done by the two groups to identify the template for the viral DNA synthesis, the virion RNA, were remarkably similar. The polymerase was soon referred to by the editors of
Nature as reverse transcriptase and the viruses as retroviruses. Back-to-back papers were published with great fanfare, and soon a broader impact of the discovery became
evident: the ability to convert mRNA into DNA permitted creation of cDNA libraries, the enzyme became crucial in cloning technologies and the rise of the biotechnology
industry, and 15 years later the enzyme was at the heart of research into the cause of AIDS and the discovery of human immunodeficiency virus (HIV).
page 356

1970 Howard Temin, David Baltimore, colleagues discovery of the reverse transcriptase of retroviruses
1--D-ribofuranosyl-1,2,4-triazole-3-carboxamide [Ribavirin]

Ribavirin
(brand names: Copegus, Rebetol, Ribasphere, Vilona and Virazole)

Ribavirin was discovered in 1970 as part of a systematic search for antiviral and antitumor activity in
synthetic nucleosides at ICN Pharmaceuticals, Inc. (later Valeant Pharmaceuticals International) by
laboratory director Roland K. Robins, chemist Joseph T. Witkowski (1901-1986) and their colleagues. This
was inspired in part by discovery in the 1960s of the antiviral activity of naturally-occurring purine-like
nucleoside antibiotics. The earlier compounds were too toxic to be clinically useful, but they served as the
starting point for pharmaceutical chemists interested in antivirals and antimetabolic chemotherapeutic
agents. In 1972, it was reported that ribavirin was active against several RNA and DNA viruses in cell
culture and/or in animals, all without undue toxicity. Experimental data indicated that the drug might
Roland K. Robins (1926-1992) be useful in treating influenza, hepatitis B, polio, measles, canine distemper, smallpox, West Nile virus
and an unnamed technician infection, dengue and a number of viral hemorrhagic fevers, including Lassa fever, yellow fever, Crimean-
Sidwell RW, Huffman JH, Khare GP, Allen LB, Witkowski JT, Congo hemorrhagic fever, and hantavirus pulmonary syndrome fever. Ribavirin also exhibits activity
Robins RK. Broad-spectrum antiviral activity of virazole: 1--D- against some DNA viruses in cell culture. Although ribavirin protected mice against mortality from
ribofuranosyl-1,2,4-triazole-3-carboxamide [Ribavirin]. Science influenza A and B viruses, in human trials results were mixed and FDA did not approve its use. In 1980,
1972;177:705-706. ICN was allowed to market ribavirin, in inhalant form, for respiratory syncytial virus infection in children,
but it was used for this indication only infrequently. By the time oral ribavirin was finally approved by
the FDA in 1998 as part of a combination treatment (with interferon- derivatives) for hepatitis C, the
original patents had expired, and ribavirin had become essentially a generic drug. Ribavirin is a prodrug,
which when metabolized becomes an analogue of purine nucleotides used in the replication of RNA. How
it affects viral replication was unknown for many years and this affected its wider approval, but in recent
years it has been found that a major mode of action is as a hypermutagen, inducing lethal mutations in
the RNA-dependent RNA polymerases central to the replication of RNA viruses. There are still important
impediments to ribavirin use: clinical treatment requires high drug dosage, which leads to significant side
effects. The thrust of ongoing research is to combine ribavirin with other drugs, thereby allowing use of a
smaller dose.
page 357

1970 Roland Robins, Joseph Witkowski, colleagues synthesis of ribavirin, the first broad spectrum antiviral drug
After the discovery of poly(A) tails on viral
mRNAs in 1970 by Joseph Kates, the same tails
were found on most mRNAs of eukaryotes.
James Darnell and his colleagues, as well
as several other groups, studied these in
eukaryotic cells and found that they served
several functions: the tails promote processing
and export of mRNAs from the nucleus,
facilitate translation, and protect the mRNAs
from degradation. It was found that nearly
all eukaryotic mRNAs possess a poly(A) tail
(50-300 nucleotides in length) at their 3 end.
Most of the eukaryotic mRNAs are post-
transcriptionally polyadenylated in the nucleus.
However, most positive sense RNA viruses
replicate in the cytoplasm, and so have evolved
their own mechanisms to acquire their polyA
tails: they either encode a 3 poly(A) sequence
on the end of their genomic RNA (positive
strand) and/or a 5 poly(U) sequence on the
negative sense RNA used as their replicative
intermediate, or they employ reiterative
copying (stuttering) on short 3 (U) sequences
on their negative sense RNA strand. Poly(A)
tails and other terminal additions to RNAs are
so important that cells have even evolved repair
mechanisms to assure that damage is not fatal
the RNA viruses enjoy such repair afforded by
the host cell that they are usually committed to
destroy.
Joseph R. Kates James E. Darnell Jr.

Kates J. Transcription of vaccinia virus genome and the occurrence of polyriboadendylic acid
sequences in mRNA. Cold Spring Harbor Symposium on Quantitative Biology 1970;35:743-752.
Darnell JE Jr, Wall R, Tushinski RJ. An adenylic acid rich sequence in mRNA of HeLa cells and its
possible relationship to reiterated sites in DNA. Proc Natl Acad Sci USA. 1971;68:1321-1325.

Edmonds M, Vaughan MH, Nakazato H. Polyadenylic acid sequences in the heterogenous nuclear
RNA and rapidly labeled poly-rRNA of HeLa cells: possible evidence for a precursor relationship.
Proc Natl Acad Sci USA. 1971;68:1336-1340.
Viral mRNA circularization at the ribosome enhances
Lee SY, Mendecki J, Brawerman G. A polynucleotide segment rich in adenylic acid in rapidly translation. The poly(A)-binding protein (PABP)
labeled poly-rRNA composed of mouse sarcoma 180ascites cells. Proc Natl Acad Sci USA. interacts with the translation initiation factor eIF4G
1971;68:1331-1335. and the 5 cap-binding protein eIF4E, brings the 5
and 3 ends of the mRNA together. page 358

1970 Joseph Kates James Darnell Jr., others discovery of the 3 poly(A) tail on many viral and cellular mRNAs
Ornithodoros spp

Phlebotomus papatasi

Aedes triseriatus
Walter Plowright (1923-2010) Robert B. Tesh Douglas M. Watts
Of all the infectious agents, the Rickettsiae and the viruses have been most successful in employing vertical transmission in their arthropod hosts in their quest for long-
term survival. One reason is that Rickettsiae and certain viruses have developed a capacity for persistent cellular infection without disturbing host functions too much. In
many cases the viruses that are capable of transovarial (or transstadial) transmission in their arthropod vectors use this mechanism as a secondary or backup life cycle, for
use when their primary horizontal transmission cycle fails. For example, it has long been thought (but is largely unproven) that transovarial virus carriage might explain the
overwintering of certain arboviruses in areas where adults cannot survive. Some viruses do, however, employ vertical transmission in their primary life cycle. The history of
our understanding of the transovarial transmission phenomenon is long, complex and difficult to assign attributions. The table included here (abridged from the review by
Burgdorfer and Varma) illustrates this. As long ago as 1906 there was an unconfirmed report that yellow fever virus was occasionally transmitted to human volunteers by
transovarially infected Aedes aegypti mosquitoes. Transovarial infection of ticks by certain viruses was the first to be considered important, but here the older literature is not
often included in databases. The more modern literature based upon more modern laboratory methods gives precedent to the proof of the transovarial transmission of (1)
African swine fever virus by ticks; (2) La Crosse virus by mosquitoes, and (3) vesicular stomatitis viruses by phlebotomine flies (sandflies).
Daubney R, Hudson JR. Nairobi sheep disease. Parasitology 1931;23:507-524.
Chumakov MP. Studies on virus encephalitides. 6. Transmission of tick-borne encephalitis to the offspring in Ixodidae ticks and the question of
natural reservoirs of this infection. Med Parazitol Parazitar Bolezni. 1944;6:38-40 (In Russian). Date Virus / Arthropod Discoverers
Benda, R. The common tick, Ixodes ricinus as a reservoir and vector of tick borne encephalitis. I. Survival of the virus (strain B3) during the
development of the tick under laboratory conditions. J Hyg Epidemiol Microbial Immunol (Prague). 1958;2:314-330. 1931 Nairobi sheep disease virus / tick Rhipicephalus appendicutatus R. Daubney, J.R. Hudson
Blakovi D, ehek J. Ticks as virus vectors in Eastern Europe. In: Maramorosch K, editor. Biological transmission of disease agents. New York:
Academic Press; 1960. p. 192-196/ 1944> Tick-borne encephalitis viruses / tick Ixodes persulcatus, I. ricinus M.P. Chumakov, et al.; R. Benda, et al.; D. Blakovi, J. ehek
ehek J. Transovarial transmission of tick-borne encephalitis virus by ticks. Acta Virol. 1962;9:220-226.
Florio L, Miller MS, Mugrage ER. Colorado tick fever. Isolation of the virus from Dermacentor andersoni in nature and a laboratory study of the 1950 Colorado tick fever virus / tick Dermacentor andersoni L. Florio, M.S. Miller, E.R. Mugrage
transmission of the virus in the tick. J Immunol. 1950;64:257-263.
Avakian AA, Lebedev AD. Natural focalization of haemorrhagic fevers. J Microbial Moscow. 1955;4:20-26 (In Russian, English translation: 1955 Omsk hemorrhagic fever virus / tick Dermacentor reticulates A.A. Avakian, A.D. Lebedev
NAMRU 3-T 147).
Plowright W, Perry CT, Pierce MA. Transovarial infection with African swine fever virus in the argasid tick, Ornithodorus moubata porcinus 1960 Crimean-Congo hemorrhagic fever virus / Hyalomma marginatum D. Blakovi, J. ehek
Walton. Res Vet Sci 1970;11:582-584.
Tesh RB, Chaniotis BN, Johnson KM. Vesicular stomatitis virus (Indiana serotype): transovarial transmission by phlebotomine sandflies. Science 1970 African swine fever virus / Ornithodoros spp W. Plowright, C.T. Perry, M.A. Pierce
1972;175:1477-1479.
Watts, DM, Pantuwatana S, DeFoliart GR. Transovarial transmission of La Crosse virus (California Encephalitis Group) in the mosquito, Aedes 1972 Vesicular stomatitis virus (Indiana) / sandfly Phlebotomus papatasi R.B. Tesh, B.N. Chaniotis, K.M. Johnson
triseriatus. Science 1973;182:1140-1141.
Burgdorfer W, Varma GR. Transstadial and transovarial development of disease agents in arthropods. Ann Rev Entomol. 1967;12:347-376. 1973 La Crosse virus / mosquito Aedes triseriatus D.M. Watts, S. Pantuwatana, G.R. DeFoliart
page 359

1970> Walter Plowright, Robert Tesh, Douglas Watts, others transovarial transmission of viruses by arthropods
Salmi AA, Grnroos JA, Halonen PE. Mixed
Viral Diagnostic Methods, 1970-1980s
haemadsorption with herpes simplex virus. Ann Med Exp
Biol Fenn. 1968;46:103-108. Direct examination of clinical specimens
Histopathology, ultrastructural pathology
Halonen PE, Murphy FA, Fields BN, Reese DR.
Hemagglutinin of rabies and some other bullet-shaped Detection of viruses by electron microscopy
viruses. Proc Soc Exp Biol Med. 1968;127:1037-1042. Detection of viral antigens
Herrmann KL, Halonen PE, Stewart JA, Casey HL, Enzyme immunoassay (EIA, ELISA, TR-FIA)
Ryan JM, Hall AD, Caswell KE. Evaluation of serological Radioimmunoassay
techniques for titration of rubella antibody. Am J Public Immunofluorescence
Health Nations Health. 1969;59:297-304.
Immunochromatography
Sarkkinen HK, Halonen PE, Arstila PP. Comparison of Immunohistochemistry
four-layer radioimmunoassay and electron microscopy for
Detection of viral nucleic acids
detection of human rotavirus. J Med Virol. 1979;4:255-260.
Polymerase chain reaction (PCR)
Sarkkinen HK, Halonen PE, Arstila PP, Salmi AA.
Hybridization (in situ, Southern blot, dot-blot)
Detection of respiratory syncytial, parainfluenza type
2, and adenovirus antigens by radioimmunoassay and Oligonucleotide fingerprinting,
enzyme immunoassay on nasopharyngeal specimens from Restriction endonuclease mapping
children with acute respiratory disease. J Clin Microbiol. Virus isolation
1981;13:258-265. Virus isolation in cultured cells
Meurman OH, Hemmil IA, Lvgren TN, Halonen PE. Virus isolation in animals or chick embryos
Time-resolved fluoroimmunoassay: a new test for rubella Serology
antibodies. J Clin Microbiol. 1982;16:920-925.
Enzyme immunoassay (EIA, ELISA, IgM-EIA)
Calisher CH, Meurman O, Brummer-Korvenkontio
Serum neutralization assay
M, Halonen PE, Muth DJ, 1985. Sensitive enzyme
immunoassay for detecting immunoglobulin M antibodies Immunoblotting (western blotting)
to Sindbis virus and further evidence that Pogosta disease is Indirect immunofluorescence assay
caused by a western equine encephalitis complex virus. Hemagglutination-inhibition assay
Pekka Eljas Halonen (1927-2001) J Clin Microbiol. 1985;22:566-571. Complement-fixation assay
Rapid viral diagnostic methods, applicable to many virus infections, made great strides in the 1970s and 1980s. This followed an era when most viral diagnostic tests were
done in public (state or city) laboratories, an era when results were only available weeks after submission of specimens. Often, the tests used centered on virus isolation and
identification, and on paired-sera serology. Needless to say, such test results had little or no clinical relevance, but were said to have public health purposes (surveillance,
outbreak investigation, etc.). Starting in Scandinavia, especially in Turku, Finland, this changed when better diagnostic technologies became available and were married
to systems for rapid testing and reporting; usually results reached clinicians caring for hospitalized patients, especially children, on the same day as specimen submission.
Diagnostic tests became part of the standard-of-care, and were relied upon in the real-time decisions of clinicians treating seriously ill patients. This era started with
immunofluorescence tests for viral antigens in respiratory diseases and was followed by radioimmunoassays and enzyme immunoassays. When tests were found to have
similar sensitivity and specificity, simplicity and rapidity became the bases for choosing methods and reagents. Pekka Halonen was the leader of this movement, which grew in
1977 into the The European Group for Rapid Laboratory Viral Diagnosis. Halonens influence was straightforward: Its the diagnosis, not the method To be useful the tests
must be rapid, specific, high-throughput Whatever method is best in a given circumstance with a given virus must be used The methods will keep changing Indeed, the
introduction of monoclonal antibody reagents and antigens produced by recombinant DNA technologies in the 1980s did keep changing the methods. In the United States,
this wave of progress in clinical virology never quite took hold and by the turn of the century clinical diagnostic labs were doing less and less virus diagnosis except by the use
of kit tests much of this failing was due to the cost-recovery system. The continuing revolution in methods, now usually based in virus genomic sequencing and very high
throughput automated equipment, is still not linked to clinical care in the United States or in many other developed countries. page 360

1970> Pekka Halonen, others linkage of rapid virus diagnostics to clinical care, especially pediatric clinical care
Some Nonhuman Primate Viruses:
Adenoviruses (numerous types)
B virus
Benign epidermal monkeypox virus
Callitrichid hepatitis (LCM) virus
Cytomegaloviruses
Ebola virus
Encephalomyocarditis virus
Epstein-Barr virus
Hepatitis A virus
Hepatitis B virus
Hepatitis C virus
Herpes simplex virus 1
Herpes simplex virus 2
Herpesvirus ateles
Herpesvirus saimiri
Herpesvirus tamarinus
Marburg virus
Measles virus
Molluscum contagiosum virus
Monkeypox virus
Papillomaviruses
Rabies virus
Simian agent (SA8 virus)
Simian foamy viruses
Simian hemorrhagic fever virus
Simian hepatitis A virus
Simian immunodeficiency viruses
Simian varicella virus
Seymour S. (Sy) Kalter (1918-2007) Richard L. Heberling SV40 virus
There are about 200 species of nonhuman primates comprising four basic Seymour Kalter and Richard Heberling Tanapox virus
groups, and viruses have been isolated from each. Approximately thirty species founded the NIH / WHO Collaborating Yabapox virus
have been used in medical research, with ten being used most commonly: Center for Reference and Research Yellow fever virus
1. Prosimians (lemurs, aye-ayes, bushbabies, tarsiers) are primitive animals in Simian Viruses, at the Southwest
and are not widely used in research. Foundation for Research and Education,
2. Old World monkeys are the macaques (most commonly used are the rhesus San Antonio, Texas, in 1970.
macaque, Macaca mulatta, and the cynomologous or long-tailed macaque,
Macaca fascicularis), baboon (Papio spp), patas monkey (Erythrocebus
patas) and vervet or African green monkey (Chlorocebus aethiops). Kalter SS, Heberling RL. Comparative virology of primates. Bacteriol Rev. 1971;35:310-364.
3. New World monkeys are the marmosets (Callithrix spp), squirrel monkey Kalter SS, Heberling RL. Primate viral diseases in perspective. J Med Primatol. 1990;19:519-
(Saimiri sciureus), spider monkey (Ateles spp), and owl monkey (Aotus spp). 535.
4. Apes. The chimpanzee (Pan troglodytes) is the only ape that has been used
in research and this is coming to a close. Kalter SS, Heberling RL, Cooke AW, Barry JD, Yian PY, Northam WJ. Viral infections of
nonhuman primates. Lab Animal Sci. 1997;47:461-467.
page 361

1970> Seymour Kalter, Richard Heberling development of nonhuman primate virology


The question whether SV40 virus can cause human tumors
has been one of the most highly controversial topics
in cancer research during the last 40 years. The debate
began with the discovery of SV40 virus as a contaminant
of poliovirus vaccines used in >100 million children and
adults between 1955 and 1963. Concerns were reinforced
by the finding that SV40 virus transforms human cells
and induces tumors in animal models. SV40 DNA has
been found in a variety of human cancers (mesotheliomas,
brain tumors, osteosarcomas, non-Hodgkin lymphomas);
however, the question remained whether these were linked
causally to the presence of SV40 virus. In 2002, the Institute
of Medicine published a report entitled Immunization
Safety Review: SV40 Contamination of Polio Vaccine and
Cancer, which concluded that: The biological evidence is
strong that SV40 is a transforming virus (able to transform
normal cells into malignant cells); the biological evidence
is of moderate strength that SV40 exposure could lead to
cancer in humans under natural conditions; the biological
evidence is of moderate strength that SV40 exposure from
polio vaccine is related to SV40 infection in humans; and
concerns about exposure to SV40 through inadvertent
contamination of polio vaccines are significant because of
the seriousness of cancers as the possible adverse health
outcomes and because of the continuing need to ensure and
protect public trust in the nations immunization program.
Others, especially anti-vaccine activists went much
further, condemning all or most current antiviral vaccines.
However, several more recent studies indicate that older
conclusions were flawed most studies describing the
recovery of SV40 DNA sequences from tumors have
Janet S. Butel Keerti Shah not been reproducible. Contamination with laboratory
Butel JS, Lednicky JA. Cell and molecular SV40 plasmids was identified as a possible source of false
biology of simian virus 40: implications for positive results, and low-level immunoreactivity of human
human infections and disease. J Natl Cancer sera to SV40 was likely the result of cross-reactivity with
Inst. 91:119-134, 1999. antibodies to the SV40-related human polyomaviruses BK
Poulin DL, DeCaprio JA. Is there a role for SV40 and JC viruses. SV40 immunoreactivity in patients with
in human cancer? J Clin Oncology 24:4356-4365, suspect tumors was no greater than that in controls. In
2006. epidemiologic studies, the increased incidence of some of
the suspect tumors in the 1970s and 1980s was not related
Shah KV. SV40 and human cancer: A review of to the risk of exposure to SV40-contaminated vaccines. In
recent data. Int J Cancer 2006;120:215-223. summary, although many people were exposed to SV40
by polio vaccination the most recent evidence does not
support the conclusion that SV40 has contributed to the
Simian vacuolating virus (SV40), cell culture development of human cancers.
page 362

1970s> Janet Butel, Keerti Shah, others controversy whether SV40 virus (in polio vaccines) causes brain tumors
Gertrude Belle Elion (1918-1999) George H. Hitchings (1905-1998) Richard J. Whitley
In 1944, Gertrude Elion was hired as an assistant to George Hitchings in Burroughs-Wellcomes research labs. Her first assignments
were in purine chemistry. In the late 1950s, advances in biosynthesis pointed Hitchings and Elion to applications for their research
they invented rational drug design, replacing the trial-and-error approach of the day. They developed drugs based on purine analogs /
antagonists. In 1959, they were awarded a patent for 2-amino-6-mercaptopurine (6-MP; Purinethol) and methotrexate, the first major drugs
to treat leukemia. In 1962, they were awarded a patent for azathioprine (Imuran), the first drug used to suppress the immune rejection of
transplanted organs, especially kidneys. Next, Elion developed allopurinol (Zyloprim), used to treat gout. Finally, in the 1960s Elion and
Hitchings developed a screening program for antiviral drugs, which resulted in the discovery of acyclovir (Zovirax) in 1974. A preclinical
investigation brought the drug to clinical trials in 1977 and the first form of the drug, for topical use against herpes keratitis, was made
available in 1982. Their early antiviral drug work was done before the structure of DNA and the anabolic pathways involved in the utilization
of purines for nucleic acid synthesis had been imagined. The work of Elion and Hitchings opened a wide field of laboratory and clinical
research as several prominent clinical virologists explored the application of rationally selected candidate drugs: Richard Whitley, Lawrence
Corey and others advanced the use of acyclovir and derivative drugs in the treatment of the various diseases caused by the herpesviruses.
Hitchings GH, Elion GB, Falco EA, Russell PB, VanderWerff H. Studies on analogs of purines and pyrimidines. Ann NY Acad Sci.
1950;52:1318-1335.
Elion GB, Furman PA, Fyfe JA, de Miranda P, Beauchamp L, Schaeffer HJ. Selectivity of action of an antiherpetic agent,
9-(2-hydroxyethoxymethyl) guanine. Proc Natl Acad Sci USA. 1977;74:57165720.
Whitley RJ, Soong S-J, Dolin R, Galasso GJ, Chien LT, Alford CA. Adenine arabinoside therapy of herpes simplex encephalitis: National
Institute of Allergy and Infectious Diseases Collaborative Antiviral Study. N Engl J Med. 1977;297:289-294.
page 363

1970s> Gertrude Elion, George Hitchings, others rational drug design, acyclovir for herpesvirus infections
Radioimmunoassays (RIAs) were first described in 1960 by Rosalyn
Yalow and Solomon Berson and immediately caught the imagination
of many investigators; over the next decade many specific assays were
developed, including several for viral diagnostics. In many laboratories
around the world special facilities were built for safely working with
the amounts of radioisotopes required for the labeling of antigens
or antibodies, but safety concerns persisted. In the early 1970s, the
idea of using enzyme labels was met with skepticism: how could so
large a molecule as an enzyme be attached to an antigen or antibody
without sterically hindering the immunochemical reaction between
antigen and antibody? Peter Perlmann (1919-2005) and Eva Engvall
at Stockholm University in Sweden and Anton Schuurs and Bauke
van Weemen at the Research Laboratories of NV Organon, Oss,
The Netherlands, independently invented enzyme immunoassays
(EIAs / ELISAs), the former first measuring IgG in rabbit serum
with alkaline phosphatase as the reporter label, the latter first
quantifying human chorionic gonadotropin concentrations in urine
with horseradish peroxidase as the reporter label. In the early 1970s,
blood-bank screening for viral diseases such as hepatitis B (HBsAg)
was done either by (semi)automated RIA or nonradioactive but rather
cumbersome hemagglutination tests. In 1976, Organon Teknika
developed and marketed a highly successful EIA system for hepatitis
B surface antigen (HBsAg), in 96-well microtiter plates. This became
the first commercially produced EIA. Other diagnostic tests followed,
most prominently EIAs for human immunodeficiency virus antibodies
in the 1980s.
Engvall E, Perlmann P. Enzyme-linked immunosorbent assay
(ELISA). Quantitative assay of immunoglobulin G. Immunochemistry
1971;8:871-874.
van Weemen BK, Schuurs AHWM. Immunoassay using antigen-
enzyme conjugates. FEBS Letts. 1971;15:232-236.

Eva Engvall, Anton Schuurs, Peter Perlmann, Bauke van Weemen There are many different types of EIAs / ELISAs for the detection of
viral antigens or antibodies in serum or other clinical specimens. One
of the most common is the sandwich ELISA, in which the moiety (say
an antibody) to be detected is sandwiched between an antigen fixed
on the surface of the wells of a 96 well plate and a chromogenically-
conjugated anti-Ig, which is added last. Common chromogens
The first EIA kit, are alkaline phosphatase and horseradish peroxidase. Common
HEPANOSTIKA test chromogen substrates, each yielding a different color, are ABTS: 405-
kit for hepatitis B 410nm (purple); TMB: non-stopped 620-650nm (red), stopped 450nm
(HBsAg) (green, as shown here); OPD: non-stopped 450nm (green), stopped
Organon Teknika, 490nm (blue); pNPP: 405-410nm (purple), and BluePhosTM: 595-
The Netherlands 650nm (yellow).
page 364

1971 Eva Engvall, Bauke van Weemen, colleagues development of enzyme-linked immunoassays (EIAs, ELISAs)
I: dsDNA viruses (Adenoviruses, Herpesviruses, Poxviruses)
II: (+) sense ssDNA viruses (Parvoviruses)
III: dsRNA viruses (Reoviruses)
IV: (+) sense ssRNA viruses (Picornaviruses, Togaviruses)
V: (-) sense ssRNA viruses (Orthomyxoviruses, Rhabdoviruses)
VI: ssRNA-RT viruses (Retroviruses)
VII: dsDNA-RT viruses (Hepadnaviruses)
David Baltimore
David Baltimore: The world of animal viruses appears to offer an unfathomable diversity, but as the molecular biology of the replication of many viruses has been studied, a
pattern of behavior has emerged. The viruses can be divided into classes, each of which has its own method of transmitting its genetic information from one generation to the
next and its own style of expressing its genetic information. Although in some cases the data are still fragmentary, it is possible to outline these systems and to place them in a
formal scheme. Viruses have evolved to the point where they are concerned mainly with two processes: duplication of their genetic material and controlled expression of the
information inherent in their genetic material.... There are two end products of the viral genetic system: new genetic material and messenger ribonucleic acid (mRNA). The one
process which all viruses must perform is mRNA synthesis. The central role of mRNA derives from the fact that viruses use cellular ribosomes and soluble factors to translate
their mRNA. Therefore, the job of the virus is almost finished once it has made mRNA, except for certain minor alterations in the functioning of the translation machinery.
One important aspect of the transcriptional systems of animal viruses has emerged during the last few years: the role of virion-associated nucleic acid polymerases. The initial
discovery of the virion-associated DNA-dependent RNA polymerase of vaccinia virus, followed a year later by the discovery of the double-stranded RNA-dependent RNA
polymerase of reovirus, opened up a new concept of how viruses carry out their initial transcriptional processes. The finding that vesicular stomatitis virus and Newcastle
disease virus both have virion-associated RNA-dependent transcriptases has led us to see such enzymes as the rule rather than the exception. Finally, the existence of an
RNA-dependent DNA polymerase in the virions of the RNA tumor viruses has led us to the concept that, whenever the first function performed by virion nucleic acid after its
introduction into the cell is the transfer of its information to another nucleic acid, the enzyme responsible for this transfer is likely to be found in the virion. Viruses which
have a double-stranded DNA genome can express and duplicate their genetic material by processes which are at least formally identical to those used by cells. Viruses with
other types of genomes require special systems for replication and transcription. These processes make up the viral genetic system, and it is possible to group viruses according
to general properties of their genetic systems.
Baltimore D. Expression of animal virus genomes. Bacteriol Rev. 1971;35:235-241.
page 365

1971 David Baltimore system of virus categorization based on the replicative path to mRNA
In 1971 Theodore Diener and his colleagues (William B. Raymer, Muriel OBrien, others) showed that the
causative agent of potato spindle tuber disease was the potato spindle tuber viroid (PSTVd), which is a
130,000 dalton (341-364 nt, varying with strain this is at least three orders of magnitude smaller than
even the most diminutive virus) single-stranded circular RNA molecule that has no coat protein, and which
does not code for any protein. Despite their small size, and therefore extremely limited genetic information,
viroids are replicated autonomously (requiring no helper virus) in the nucleus or chloroplasts of plant cells
by host RNA-dependent RNA polymerase. In some ways, viroids resemble transposons, but taxonomically
they have been accorded their own taxon. The painstaking work leading to these conclusions took six
years, particularly since the discovery had to stand up against anti-dogma skeptics. One key was the
development of a bioassay for the PSTVd viroid quantitating the infectious moiety was difficult because
of the very slow development of the disease in potatoes, often taking several years to become manifest.
The new bioassay was based on infection of tomatoes, where plants become dramatically stunted in a few
weeks. Overall, the discovery brought about a revolution in plant virology as more and more diseases
were found to be caused by viroids presently, there are at least 53 viroid diseases, some commercially
important, including cadang-cadang, a devastating disease of commercial coconut palm plantations in
several parts of the world.
It has been proposed that human hepatitis D virus (hepatitis delta virus, HDV) may have originated as a
viroid. Evidence in support of this stems from the fact that both HDV and viroids exist as single-stranded,
linear and closed circular RNAs that have rod-like structures. Likewise, both HDV and viroids contain
RNA sequences that can form catalytically active structures, that is ribozymes. During replication, these
catalytic RNAs are required in order to produce unit length copies of the genome. Neither HDV nor viroids
encode their own polymerase; instead, their replication requires a host polymerase that can utilize RNA
as a template. RNA polymerase II has been implicated as the polymerase responsible for the replication
of HDV. Normally RNA polymerase II utilizes DNA as a template and produces mRNA. Consequently, if
HDV indeed utilizes RNA polymerase II for its replication, it would be the only known pathogen capable of
using a DNA-dependent polymerase as an RNA-dependent polymerase.
Diener TO, Raymer WB. Potato spindle tuber virus: a plant virus with properties of a free nucleic acid.
Science 1967;158:378-381.
Theodore Otto Diener Diener TO. Potato spindle tuber virus. IV. A replicating, low molecular weight RNA. Virology
1971;45:411-428.
Diener TO. Discovering viroidsa personal perspective. Nat Rev Microbiol. 2003;1:75-80.
Taylor JM. Hepatitis delta virus. Virology 2006;344: 71-76.

Secondary structure of potato spindle tuber viroid, 341-364 nucleotides, varying with strain.
Nucleotides in central area that are common to most viroids are highlighted in yellow/red

Potatoes, infected and uninfected with


potato spindle tuber viroid (PSTVd)
page 366

1971 Theodore Diener discovery of viroids (infectious naked RNA molecules)


Sylvia D. Gardner Anne Field (1936-2007)
BK virus was discovered at the Virus Research Laboratory, Public Health Laboratory Service, Collindale, London,
during a study by Sylvia Gardner of cytomegalovirus infection in kidney transplant recipients. Urine collected from
a patient after transplantation was found to contain numerous cells bearing viral inclusions. Electron microscopy
by Anne Field demonstrated virus particles that resembled papillomaviruses. Inoculation of the urine into rhesus
monkey kidney cells and human embryonic kidney cells produced a cytopathic effect and confirmed that the
virus was not a papillomavirus. The new virus and named BK virus after the initials of the patient from whom it
was isolated. Epidemiological studies at Collindale in the 1970s and 1980s showed that up to 90% of some human
populations became infected by BK virus by adulthood. Following transplantation, 10% to 60% of allograft recipients
excreted virus in their urine. However, viral infection was usually asymptomatic or associated with only transient graft
dysfunction. The first case of clinically significant BK virus interstitial nephritis was diagnosed by a needle biopsy of
an allograft in 1993, after which BK virus-associated interstitial nephritis was reported from most kidney transplant
centers around the world. In recent years, an etiological role of BK virus in human kidney cancers has been advanced,
Renal allograft biopsy, BK virus nephropathy:
but the evidence is disputed and the view most experts is that there is not an etiologic association.
(top) intranuclear inclusion in tubular epithelial
Gardner SD, Field AM, Coleman DV, Hulme B. New human papovavirus (BK) isolated from urine after renal cell; (bottom) electron microscopy of inclusion
transplantation. Lancet 1971;1:1253-1257. showing that it is comprised of virions arranged in
page 367 paracrystalline array

1971 Sylvia Gardner, Anne Field, colleagues discovery of BK virus, etiologic agent of kidney and bladder disease
Thottapalayam virus has been isolated from Suncus murinus (Asian house
shrew, musk shrew) in India, Afghanistan, Pakistan, India, Sri Lanka, Nepal,
Bhutan, Burma, China, Taiwan, and Japan. This is the only hantavirus not
isolated from a rodent; whether the virus is naturally harbored by this
insectivore host or represents spillover from a rodent reservoir host is
unknown, but some evidence suggest the virus co-evolved with this host.
Carey DE, Reuben R, Panicker KN, Shope RE, Myers RM. Thottapalayam
virus: a presumptive arbovirus isolated from a shrew in India. Indian J Med
Res 1971;59:1758-1760.
Song J-W, Baek L-J, Schmaljohn CS, Yanagihara R. Thottapalayam virus, a
prototype shrew-borne hantavirus. Emerg Infect Dis. 2007;13:980-985.

Donald E. Carey Jin-Won Song

Geographic distribution of Suncus murinus


(blue = native; red = introduced)

Robert E. Shope (1929-2004) Richard Yanagihara Suncus murinus (Asian house shrew, musk shrew)
page 368

1971 Donald Carey, Jin-Won Song, Robert Shope, Ric Yanagihara, others Thottapalayam virus (the first hantavirus)
Bert Geoffrey Achong (1928-1996) Michael Anthony Epstein Simian foamy virus 1
cell culture, thin section electron microscopy

Simian foamy viruses (SFVs) have been reported in 1-4% of persons


occupationally exposed to nonhuman primates in zoos, primate centers,
and laboratories, in North America and in Europe. More recently, naturally
acquired SFV infections were described in hunters in Cameroon and in
a person who had had contact with Macaca fascicularis macaques in
Indonesia. About 70-90% of non-human primates born in captivity are
infected with SFVs, but no disease has been associated with these infections.
Achong BG, Mansell PW, Epstein MA, Clifford P. An unusual virus in
cultures from a human nasopharyngeal carcinoma. J Natl Cancer Inst.
1971;46:299-307.
Heneine W, Switzer WM, Sandstrom P, Brown J, Vedapuri S, Schable CA,
Khan AS, Lerche NW, Schweizer M, Neumann-Haefelin D, Chapman LE,
Folks TM. Identification of a human population infected with simian foamy
viruses. Nat Med. 1998;4:403-407.
Herchenrder O, Renne R, Loncar D, Cobb EK, Murthy KK, Schneider J,
Mergia A, Luciw PA. Isolation, cloning, and sequencing of simian foamy
viruses from chimpanzees (SFVcpz): high homology to human foamy virus
(HFV). Virology. 1994;201:187-199.
Thomas Folks Paul Luciw
page 369

1971 Bert Achong, Michael Epstein, Paul Luciw, Thomas Folks, others simian foamy virus infection in humans
George Martin Baer (1936-2009) Franz Steck (1932-1982) Alexander Wandeler
The concept of vaccinating wildlife rabies reservoir host species against rabies was developed by
George Baer and his colleagues at the U.S. Centers for Disease Control in 1971. They did extensive
containment-housed animal experiments to show safety and efficacy; however, because of regulatory
entanglements they were not allowed to do field tests. Two veterinarians from the Swiss Rabies
Center, Franz Steck and Alexander Wandeler, used the method to immunize foxes against rabies; they
experimented with encapsulating vaccine and placing capsules in chicken heads. When unsuspecting
foxes ate the chicken heads, they broke open the capsules and were immunized. Starting in 1978, the
Swiss started dropping vaccine-containing chicken heads in the Rhone Valley by helicopter, but after
Steck was killed in a helicopter crash other methods of distribution were devised. The method was
a great success and within a few years Switzerland was free of rabies (the fox was the only reservoir
host present in the country). The method, improved over the years in several ways, was taken up by
other European countries and finally in Canada and the U.S., where the variety of rabies reservoir (above) Rabies, Animals,
host species again called for modifications of bait delivery, but where again there has been great Europe, 1983 after 30 years
success in reducing wildlife rabies and consequent transmission to domestic animals and humans. of conventional rabies control

Baer GM, Abelseth MK, Debbie JG. Oral vaccination of foxes against rabies. Am J Epidemiol. (left) Rabies, Animals,
1971;93:487-490. Europe, 2008 after 26
Steck F, Wandeler A, Bichsel P, Capt S, Hafliger V, Schneider L. Oral immunization of foxes against years of bait-delivered oral
rabies: laboratory and field studies. Comp Immunol Microbiol Infect Dis. 1982;5:165-171. vaccination of foxes
page 370

1971 George Baer, Franz Steck, Alexander Wandeler, colleagues oral vaccination of wildlife against rabies
The discovery of Norwalk virus is the first instance of bypassing classical tissue-culture and
experimental animal virology in necessary favor of direct virology or particle virology, as
described Albert Kapikian. In 1969, his lab at the NIH turned to the study of the etiology of acute
nonbacterial (viral) gastroenteritis, where it was already known from human volunteer studies
involving the feeding of bacteria-free stool ultrafiltrates, that virus(es) were involved as important
etiologic agents. Newer techniques developed for the detection of fastidious viruses, such as organ
culture, were tried by research groups in the U.S., the United Kingdom and Japan, still without
success. In 1970, at NIH fecal ultrafiltrates from four gastroenteritis outbreaks were studied in
human volunteers one of the outbreaks occurred in an elementary school in Norwalk, Ohio in
1968. During a 2-day period, 50% of the students and teachers developed gastroenteritis, along
with 32% of contacts. Standard lab studies failed to reveal an etiologic agent. An ultrafiltrate from
a secondary case in the Norwalk outbreak was administered orally to 3 volunteers, 2 of whom
developed gastroenteritis. Minimal virus characterization studies were conducted using this
ultrafiltrate. Then, everything changed: Kapikian had studied in the lab of Anthony Waterson and
June Almeida at the Royal Postgraduate Medical School, University of London, and had brought the
immunoelectron microscopy (IEM) technique back to the NIH, where still the Norwalk agent could
not be cultivated. After 20 months of developmental work, Kapikian examined Norwalk agent stool
ultrafiltrates, using a volunteers convalescent serum as the source of antibody. He stated, glistening
aggregates of nonenveloped, antibody-coated 27-nm, virus-like particles, which resembled
rhinoviruses, were visualized. [The virus is now known to be 35-39nm in diameter.] It was clear that
further studies were needed to determine the significance of this finding this was done by showing
that volunteers who became ill developed a serologic response to the 27-nm particles visualized
by IEM. An antibody-rating system was developed, in which dilutions of pre- and post-challenge
serum were reacted with the Norwalk agent ultrafiltrate and examined by negative contrast electron
microscopy, that is by IEM. When not coated by antibody the icosahedral virions appear to have a
Star of David pattern on their surface; when heavily coated by antibody virion structure is almost
totally obscured. The final proof involved testing paired acute and convalescent sera from patients
who developed naturally occurring illness during the outbreak in Norwalk, Ohio it was reasoned
Albert Z. Kapikian that it would be highly unlikely that the newly discovered 27-nm particle was an adventitious agent
if the naturally occurring cases developed an antibody response to it. Success here, with negative
controls used in blinded observations of IEM reactions, was a major link in establishing the etiologic
association of the virus and the disease. Soon, the same method showed that there were several
distinct Norwalk-like viruses. During the early 1990s, cloning and sequencing of Norwalk-like
viruses led to the development of sensitive molecular assays [e.g., reverse transcription-polymerase
chain reaction (RT-PCR), nucleotide hybridization probes, and enzyme-linked immunosorbent
assays (ELISA) based on recombinant-DNA expressed viral antigens]. Using these assays, it was
shown that Norwalk-like viruses are the cause of a high percentage of foodborne and waterborne
gastroenteritis outbreaks. The viruses are now classified as belonging to the genus Norovirus, family
Caliciviridae.
Kapikian AZ, Wyatt RG, Dolin R, Thornhill TS, Kalica AR, Chanock RM. Visualization by immune
electron microscopy of a 27nm particle associated with acute infectious nonbacterial gastroenteritis.
J Virol. 1972;10:1075-1081.
Kapikian AZ. The discovery of the 27-nm Norwalk virus: an historic perspective. J Infect Dis.
Norwalk virus, without and with antibody (IEM) 2000;181S2:S295-302.
page 371

1972 Albert Kapikian, colleagues discovery of Norwalk virus (the first human calicivirus, the first norovirus)
By the mid-1960s, several members of Paul Bergs team at Stanford University, including
Dale Kaiser, had trained at the Pasteur Institute and followed the research of Franois Jacob
and Jaques Monod closely. In a 1965 presentation, Kaiser suggested that the mechanisms at
work in bacteriophage might have analogues in mammalian tumor viruses such as SV40
virus. Berg was fascinated by the implications: could viruses be used to study gene regulation
in mammalian cells, as they had been in bacterial cells? He felt that the understanding of
gene expression in bacteria was becoming clear, but there was still little known about gene
expression in higher life forms. Eager to explore such fresh territory, Berg spent several years
preparing for the work that would lead to the development of recombinant DNA technologies.
One immediate problem was that the DNA of obvious vectors SV40 and polyoma viruses
was so small that it was unlikely that it could pick up genes from other sources. To overcome
this limitation, Berg and his team set out to combine SV40 DNA with a plasmid found in E.
coli. The plasmid consisted of phage DNA together with three genes from E. coli. By splicing
the plasmid into the SV40 DNA, they created a recombinant DNA molecule that had both
the ability to enter a mammalian cell and the ability to carry new genes from other sources
when it did so. The technique that Berg and his team devised for splicing two DNA molecules
together utilized an array of enzymes: first, they cut open the circular SV40 and plasmid
DNAs. Working from the knowledge that phage DNA strands have cohesive or sticky ends
that allow complimentary base pair bonding, they created their own sticky ends using another
enzyme to add strings of thymine (T) and adenine (A) nucleotides to the ends of each DNA.
Then finally, the A and T strands of the two DNAs were annealed using DNA polymerase,
ligase, and other enzymes. This complex procedure, resulting in the first recombinant DNA
molecule, was described by Berg, David Jackson, and Robert Symons in 1972. Because such
a revolutionary technique was considered by some to create unpredictable occupational or
environmental hazards, Berg did not insert the recombinant DNA molecule into a bacterium
rather, within a few years he took the leading role in organizing the scientific community to
address the issue of recombinant DNA safety (he served as chair of the National Academy of
Sciences Committee on Recombinant DNA Molecules and in 1975 organized the Asilomar
Conference on Recombinant DNA). Within a year of Berg and his teams publication, Stanley
Paul Berg Cohen, Herbert Boyer, and others had discovered that a restriction enzyme, EcoR1, would
create the necessary sticky ends on almost any DNA molecule, which greatly simplified the
recombination process and made it possible to splice together any DNA from any source this
was the beginning of genetic engineering and the biotechnology industry.
Jackson DA, Symons RH, Berg P. Biochemical method for inserting new genetic information
into DNA of simian virus 40: circular DNA molecules containing lambda phage genes and the
galactose operon of Escherichia coli. Proc Natl Acad Sci USA. 1972;69: 2904-2909.
Berg P, Mertz JE. Personal reflections on the origins and emergence of recombinant DNA
technology. Genetics 2010;184:9-17.

Method used by Jackson, Symons and Berg


in 1972 for constructing SV40-dvgal 120
recombinant DNA in vitro
page 372

1972 Paul Berg, Dale Jackson, Robert Symons development of recombinant-DNA technology
Stanley Norman Cohen Herbert Boyer Stanley Cohens molecular cloning diagram

In 1972, Stanley Cohen from Stanford University and Herbert Boyer from the University of California San Francisco met in Honolulu at a U.S.-Japan conference on bacterial
plasmids. Boyer gave a paper on the nature of the cleavage of DNA carried out by the restriction enzyme EcoRI, and Cohen gave a paper on his procedure for transfecting E.
coli with plasmid DNA. They made a now-famous late evening excursion to a delicatessen in Waikiki for pastrami sandwiches and immediately began planning collaborative
experiments. The work involved Cohens plasmids and Boyers EcoRI enzyme, with analysis of recombinant products in each laboratory. Within four months they and their
team members had a breakthrough the first success involved one of Cohens plasmids, pSC101, a tetracycline-resistance gene of E. coli, which they transferred from one
strain of E. coli to another. They soon moved on to more complicated cloning experiments. They joined tetracycline-resistant plasmids with kanamycin-resistant plasmids
and inserted them into E. coli. Next, they showed that genes could be transferred between species: a Staphylococcus aureus plasmid was spliced into E. coli plasmids and then
inserted into E. coli. They then cloned genes from a South African toad (Xenopus laevis) into E. coli. They wrote three landmark papers in 1973 and 1974, which revolutionized
biological research and launched the multi-billion dollar biotechnology industry.
Cohen SN, Chang AC, Boyer HW, Helling RB. Construction of biologically functional bacterial plasmids in vitro. Proc Natl Acad Sci USA. 1973;70:3240-3244.
Chang AC, Cohen SN. Genome construction between bacterial species in vitro: replication and expression of Staphylococcus plasmid genes in Escherichia coli.
Proc Natl Acad Sci USA. 1974;71:1030-1034.
Morrow JF, Cohen SN, Chang AC, Boyer HW, Goodman HM, Helling RB. Replication and transcription of eukaryotic DNA in Escherichia coli.
Proc Natl Acad Sci USA. 1974;71:1743-1747.
page 373

1973 Stanley Cohen, Herbert Boyer application of recombinant DNA technology, start of genetic engineering
After Stanley Cohen, Herbert Boyer, and their colleagues
succeeded with the first cloning experiments in 1973, concerns
over biosafety led to a request that the National Academy of
Sciences address the situation. In response, Paul Berg convened
a meeting of seven scientists, which produced an open letter
to the scientific community (published in Science) requesting a
moratorium on recombinant DNA research until the hazards of
such research could be assessed by an international conference of
scientists, and procedures developed for containment of possible
hazards. This provoked a wide range of commentary, as well as
protests, but the voluntary moratorium was universally observed.
The first conference was held at the Asilomar Conference Center
near Monterey in 1973. Although no problems arising from
working with recombinant infectious agents were uncovered,
several recommendations were made; the proceedings were
published by the Cold Spring Harbor Laboratory Press (Alfred
Hellman, Michael N. Oxman, Robert Pollack, editors). The
second conference was organized by Berg, Maxine Singer, and
Richard Roblin, and held at the Asilomar Conference Center,
this one in 1975. There were about 140 participants; it was
recommended that the moratorium on experiments be lifted
Maxine Singer, Norton Zinder (1928-2012), Sydney Brenner, Paul Berg and replaced with guidelines governing such research. In 1976,
the NIH issued its first set of Guidelines for Research Involving
at Asilomar, 1973 Recombinant DNA. As the guidelines took hold, research
Time Magazine cover art, 1977 under seemingly appropriate containment recommenced with
a bang. The first principle for dealing with potential risks was
Doomsday: Tinkering With Life
that containment facilities should be made an essential part
It is one of the lowliest of natures creatures, a rod
of experimental design. A second principle was that the level
-shaped beastie less than a ten-thousandth of an
of containment should match the estimated risk as closely as
inch long [Escherichia coli]... Yet this tiny parcel
possible varying risk levels were identified (table). Biological
of protoplasm has now become the center of a
barriers to limit the spread of recombinant DNA were also
stormy controversy that has divided the scientific
included in the guidelines; these included fastidious bacterial
community, stirred fearsoften farfetchedabout
hosts that were unable to survive in natural environments and
tampering with nature, and raised the prospect of
equally fastidious vectors (plasmids, bacteriophages, viruses).
unprecedented federal and local controls on basic
scientific research. Hellman A, Oxman MN, Pollack R, editors. Biohazards in
biological research. Proceedings of a conference held at the
CLASSIFICATION OF INFECTIOUS MICROORGANISMS BY RISK GROUP Asilomar conference center, Pacific Grove, California, January 22-
Following upon the Asilomar Conference of 1973 24, 1973. New York: Cold Spring Harbor Laboratory Press; 1973.
Risk Group 1 Agents that are not associated with disease in healthy adult humans. Berg PD, Baltimore D, Boyer HW, Cohen SN, Davis RW, et al.,
Agents that are associated with human disease which is rarely serious and for which preventive or Letter: potential biohazards of recombinant DNA molecules.
Risk Group 2 Science 1974;185: 303.
therapeutic interventions are often available.

Risk Group 3
Agents that are associated with serious or lethal human disease for which preventive or therapeutic Berg PD, Baltimore D, Brenner S, Roblin RO, Singer MF.
interventions may be available (high individual risk but low community risk). Summary statement of the Asilomar conference on recombinant
Risk Group 4
Agents that are likely to cause serious or lethal human disease for which preventive or therapeutic DNA molecules. Proc Natl Acad Sci USA. 1975;72:1981-1984.
interventions are not usually available (high individual risk and high community risk).
page 374

1973, 1975 Paul Berg, others Asilomar conferences and proceedings: Biohazards in Biological Research
Robert H. Purcell, Albert Z. Kapikian, Stephen M. Feinstone Hepatitis A virus
Negative contrast electron microscopy
Albert Kapikians success in discovering and characterizing Norwalk virus as a major cause of acute gastroenteritis provided the impetus and methodology for seeking other
previously undetected human viral pathogens over time, he developed immune-electron microscopy (IEM) to a fine art. Using the method in carefully controlled studies
(samples from human volunteer studies and outbreaks, IEM results recorded from blinded/coded reactions, etc.), Kapikian, Robert Purcell and Stephen Feinstone, at the NIH,
succeeded in visualizing and identifying the virus responsible for hepatitis A. Prior to this, even the diagnosis of acute viral hepatitis was problematic; diagnosis was largely
based upon clinical and clinical chemistry evidence, but the overlap between what turned out to be hepatitis B and then hepatitis C virus infections was confounding. In
addition, in the 1960s, a number of unclassified, so-called candidate hepatitis viruses were isolated from patients with acute hepatitis, all of which proved inconsequential.
Further, a number of agents belonging to well-known virus families were also isolated from patients with hepatitis, again just confounding. Prior to 1973, it was known that
hepatitis A was transmissible by oral ingestion of ultrafiltered stool extracts from infected patients. Thus, the IEM method was employed on stool ultrafiltrates, not serum or
other clinical specimens. In the first experiments, ultrafiltered stool specimens from hepatitis A infected volunteers were incubated with dilutions of convalescent serum from
a hepatitis A patient. The 27nm virus was found in two of four specimens from subjects suffering acute illness but in none of four controls. Large numbers of experiments
followed, with stool specimens and convalescent serum from other sources (including Saul Krugmans Willowbrook State Hospital and outbreaks at Holy Cross College and
in American Samoa). The work was quickly confirmed by other investigators, including several working on food-borne acute hepatitis outbreaks. The same stool specimens
reactive by IEM infected chimpanzees and marmosets, with the same 27nm virus particles appearing in their stools.
Feinstone SM, Kapikian AZ, Purcell RH. Hepatitis A: detection by immune electron microscopy of a virus-like antigen associated with acute illness.
Science 1973;182:1026-1028.
page 375

1973 Stephen Feinstone, Albert Kapikian, Robert Purcell discovery of hepatitis A virus
Human rotavirus, varying degrees of stain penetration
into the three-layered virion
In 1973, Ruth Bishop, at the Royal Childrens Hospital in
Melbourne, was studying childhood diarrhea and concluded
that a nonbacterial infectious agent was probably to blame. She
teamed up with Geoff Davidson, Ian Holmes and Brian Ruck; by
negative contrast electron microscopy they found a previously
unknown virus in the first bowel biopsy sample examined and
in many following samples. When their finding was published,
confirmation came from around the world and soon the virus
Ruth F. Bishop Ian H. Holmes (1931-2010) was recognized as the most common cause of severe diarrhea in
infants and young children worldwide.
Bishop RF, Davidson GP, Holmes IH, Ruck BJ. Virus particles in epithelial cells of
duodenal mucosa from children with acute non-bacterial gastroenteritis. Lancet
1973;1:1281-1283.

Global distribution of rotavirus mortality. Each dot = 1000 deaths


~527,000 rotavirus-related deaths per year in children <5 years of age
Thomas Flewett (1922-2006) Albert Z. Kapikian page 376

1973 Ruth Bishop, Ian Holmes, Thomas Flewett, Albert Kapikian, others discovery of human rotaviruses
Most of the critical developments in DNA, RNA and protein electrophoresis
occurred in a brief spurt around 1971-1973. The first major development was
SDS- polyacrylamide gel electrophoresis, which was most useful in separating
smaller molecules such as proteins. For separating gene-sized fragments of DNA,
agarose gel electrophoresis was developed by Joe Sambrook, Bill Sugden and Phillip
Sharp at Cold Spring Harbor Laboratory. This method separated DNA molecules
ranging from several hundred nucleotides in length to over 10,000 nucleotides.
The phosphate groups in the DNA backbone are negatively-charged, giving DNA
molecules an overall negative charge; smaller molecules migrate through the pores
of the agarose gel faster than larger molecules. Initially, agarose gels were run in
tubes, tapered at the end to stop the slippery agarose from sliding out, and then
later on vertical slabs, using special frames to prevent slippage and intricate combs
to allow the even loading of samples. These gels were, in turn, replaced by the
horizontal slab gels, which are most common today. Early on, radioactive DNA
fragments were analyzed by autoradiography and scintillation counting of gel slices;
then, Piet Borst and Cees Aaij at the University of Amsterdam introduced the
use of the fluorescent dye, ethidium bromide (added to the conductive medium),
which binds to DNA and fluoresces when illuminated with ultraviolet light. The
size of DNA fragments were and still are determined by comparison with markers
(DNA fragments of known sizes). Viruses were among the first DNAs studied by
this method, for example adenovirus DNA, as studied by Michiaki Takahashi,
Takeo Ogino and Koichi Baba. When viral DNAs were cut with various restriction
Joseph Sambrook Phillip A. Sharp enzymes, complete restriction maps of viral genomes followed quickly.
Sharp PA, Sugden B,
Sambrook J. Detection of
two restriction endonuclease
activities in Haemophilus
parainfluenzae using analytical (Left) One of many agarose gel
agarose-ethidium bromide electrophoresis formats
electrophoresis. Biochemistry (vertical slab format)
1973;12:3055-3063. (Below) Ethidium bromide
Aaij C, Borst P. The gel stained agarose gel showing
electrophoresis of DNA. separated DNA fragments
Biochim Biophys Acta
1972;269:192-200.
Borst P. Ethidium DNA
agarose gel electrophoresis:
How it started. IUBMB Life
2005;57:745-747.
Takahashi M, Ogino T, Baba K.
Separation of adenovirus DNAs
of different molecular length
by agarose-gel electrophoresis.
page 377 Piet Borst Biken J. 1968;11:69.

1973 Joseph Sambrook, others development of agarose gel electrophoresis of DNA / ethidium bromide staining
Adherent dendritic cell and macrophage
Zanvil A. Cohn (1926-1993) and Ralph A. Steinman (1943-2011) (scanning electron microscopy, colorized)
Dendritic cells were discovered in 1973 by Ralph Steinman and Zanvil
Cohn at Rockefeller University. At the time, they were trying to understand
the induction of immune responses in mice. Steinman noticed unusual cells
in the spleen that acted like macrophages but looked nothing like them.
Naming them dendritic cells, for their branching tree-like shape, they were
initially unaware of their significance. However, over the next several years
Steinman discovered that these cells were the central players in triggering
the immune response dendritic cells, not macrophages, are the main
T cell-stimulating cells. In the 1990s, he found that immature dendritic
cells position themselves at sites that are most vulnerable to invasion by
pathogens skin, respiratory tract, intestines and genital organs. They also
localize in the T-cell areas of the spleen and lymph nodes. When stimulated
by exposure to a pathogen, these cells undergo an incredible maturation
process, increasing antigen-capture and antigen-presenting capacities
and increasing cytokine signalling to T lymphocytes. Today, dendritic cell
research is central to subjects as diverse as autoimmune disease, cancer and
viral vaccine development.
Steinman RM, Cohn ZA. Identification of a novel cell type in peripheral
lymphoid organs of mice. J Exp Med. 1973;137:1142-1162. page 378

1973> Ralph Steinman, Zanvil Cohn discovery of dendritic cells and their role in antigen presentation
Ari Helenius Multiple portals of virus entry into mammalian cells. (1) Clathrin-mediated entry
(vesicular stomatitis virus), (2) fusion-entry (HIV), (3) macropinocytosis-mediated entry
(vaccinia virus, HIV), (4) phagocytosis-like-mediated entry (herpes simplex virus), (5)
phagocytosis-mediated entry (bacteria), (6) caveolin-mediated entry (simian virus 40)
There is a very large literature on virus entry and its routes and mechanisms; it is impossible to attribute primacy because over the years so many different approaches
were used to study phenomena from electron microscopy to molecular genetics. Both non-enveloped and enveloped viruses share the same main steps and routes of
virus entry, which begin with attachment to cell-surface receptors and end with the delivery of the viral genome into the cell cytoplasm. The mechanisms underlying this
are now known to be quite complex and diverse. After binding to cellular receptors, viruses use two main routes to enter the cell: endocytic and non-endocytic routes. The
endocytic route is usually accomplished by transport in clathrin-coated vesicles or pits, but non-clathrin-coated pits, macropinocytotic vesicles and caveolae are also used.
The non-endocytic route involves directly crossing the plasma membrane at neutral pH. Some viruses use both pathways (e.g., HIV). The complex processes are regulated
and mediated by membrane proteins once viral and cellular membranes come into close proximity. For both enveloped and non-enveloped viruses, entry into cells involves
important conformational changes of viral entry proteins and/or host-cell receptors, which are induced by low endosomal pH. Viruses may leave endosomes by penetration
(for non-enveloped viruses) or fusion (for enveloped viruses). After entry into the host cell, many viruses are transported through the cytoplasm as nucleoprotein complexes
using nuclear localization signals on the viral nucleoprotein complex to target entry into the nucleus. Elucidation of the molecular mechanisms and the dynamics of the
conformational changes driving virus entry are key to our understanding of virus tropism and pathogenesis, and are key to development of entry inhibitors and novel vaccine
immunogens much contemporary research is being done in this area.
Miyamoto K, Morgan C. Structure and development of viruses as observed in the electron microscope. XI. Entry and uncoating of herpes simplex virus. J Virol. 1971;8:910-918.
Helenius A, Kartenbeck J, Simons K, Fries E. On the entry of Semliki forest virus into BHK-21 cells. J Cell Biol. 1980;84:404-420.
Dimitrov DS. Virus entry: molecular mechanisms and biomedical applications. Nat Rev Microbiol. 2004;2:109-122.
page 379

1973> Councilman Morgan, Ari Helenius, others discovery of the variety and complexity of virus entry mechanisms
In the late 1960s, Daniel Nathans colleagues, Bernard Roizman
and Robert Wagner, left The Johns Hopkins University School
of Medicine, and he decided to redirect his research to the
animal virology niche they left unoccupied after some
thought he selected the simplest DNA tumor virus, SV40. Even
though the SV40 genome was known to be only about 5,000
base pairs of double-stranded circular DNA, its ability to grow
lytically in monkey cells and to cause permanent tumorigenic
transformation of rodent cells suggested that there was much
more to learn. Nathans spent a sabbatical in 1969 with Leo Sachs
and Ernest Winocour at the Weitzman Institute in Israel learning
how to grow and assay SV40 virus. While there, he received
a letter from his colleague, Hamilton Smith, describing an
enzymatic activity by the bacterium Hemophilus influenzae that
degraded DNA from foreign cells but did not degrade its own
DNA. Smith mentioned evidence that this enzyme cleaved DNA
at specific nucleotide sequences. Nathans immediately realized
the implications of this discovery, that restriction enzymes might
be used to dissect the genome of SV40 virus and show something
about how the virus works. Nathans returned to Hopkins with
radiolabeled SV40 DNA in his luggage, and he set about testing
his ideas. Using Smiths H. influenzae restriction enzyme to cut
SV40 virus DNA (actually, the cleavage activity was due to a
mixture of two restriction enzymes), Nathans and his graduate
student Kathleen Danna cleaved SV40 DNA into 11 specific
pieces. These results were published in their 1971 classic paper in
the Proceedings of the National Academy of Sciences. Immediately,
it was understood that a genome map should be developed
Daniel Nathans (1928-1999) Kathleen J. Danna structural genes, early and late genes, genes affecting host cell
tumorigenic transformation, the origin of replication, etc., needed
to be mapped. Together with Danna and George Sack, a medical
fellow, Nathans determined the specific order of each fragment
Autoradiogram of 14C-labeled SV40 DNA in relation to the others and constructed the first map of a viral
from Nathans and Dannas classic paper genome. This map was constructed by isolating overlapping
depicting 11 distinct HINDII/III restriction enzyme partial digestion products and determining their constituents.
fragments of virus DNA as separated by Their identification of the origin of SV40 DNA replication was the
SDS-polyacrylamide gel electrophoresis first genetic signal to be positioned on a eukaryotic viral genome.
Danna K, Nathans D. Specific cleavage of simian virus 40 DNA
by restriction endonuclease of Hemophilus influenzae. Proc Natl
Acad Sci USA. 1971;68:2913-2917.
Danna K, Sack GH Jr, Nathans D. Specific studies of simian virus
40 DNA. VII. A cleavage map of the SV40 genome. J Mol Biol.
1973;78: 363-368.
Modern SV40 genome map
page 380

1973 Daniel Nathans, others completion of restriction enzyme map of a viral genome (SV40 virus)
The classic experiment of Zinkernagel
and Doherty demonstrating that
antigen recognition by cytotoxic T
lymphocytes (CTLs) exhibits major
histocompatibility complex (MHC)
restriction. H-2k haplotype mice were
infected with LCM virus to induce
Peter C. Doherty Rolf M. Zinkernagel CTLs specific for the virus; several days
From Robert Blanden (John Curtin School of Medical Research website, 1996): In 1973 it was known that the major histo- later their spleen cells (which included
compatibility gene complex (MHC) encoded the antigens responsible for stimulating a response by T lymphocytes that led to the CTLs) were isolated and incubated
rejection of tissue grafts exchanged between incompatible individuals. However, since grafting was an artificial event performed by with LCM-infected target cells of the
transplantation surgeons and experimental immunologists, the real biological function of the MHC was a mystery. Rolf Zinkernagel same or different haplotype that were
and Peter Doherty solved the mystery. The experiments that they performed made it clear that when T lymphocytes recognized intracellularly labeled with 51Cr and
and killed a virus-infected cell, they were recognizing not only a component of the virus but simultaneously also recognizing major either infected or not with LCM virus.
histocompatibility molecules on the surface of the infected cell. Thus, the major histocompatibility antigen molecules were a means As measured by release of 51Cr into the
of focusing T cell recognition on cell surfaces so that they could perform the essential functions leading to elimination of infected culture supernatant, the CTLs only
cells and recovery from viral infection. Following the precedent set by Zinkernagel and Doherty, other workers showed that the killed syngeneic virus-infected target
basic principles discovered by them also applied to the mechanisms by which a subset of T lymphocytes helped B lymphocytes to cells here H-2k haplotype LCM-
produce antibodies.... Over two decades later, much of the molecular detail of T cell recognition and the cell biology connected with infected target cells. Later studies with
immune responses mediated by T lymphocytes has been worked out, but these aspects of immunology still constitute an enormous congenic and recombinant congenic
proportion of current immunological research. Doherty and Zinkernagels discovery has been relevant to clinical medicine as well. mouse strains showed that the CTLs
It relates both to efforts to strengthen the immune response against invading viruses and microorganisms and the means to combat and the virus-infected target cells
certain forms of cancer (via vaccines, stimulants of innate immunity), and to efforts to diminish the effects of autoimmune reactions. must share class I MHC molecules on
their surfaces, encoded by the K or
Zinkernagel RM, Doherty PC. Restriction of in vitro T cell-mediated cytotoxicity in lymphocytic choriomeningitis within a syngenic
D regions of the MHC. Thus, antigen
and semiallogeneic system. Nature 1974;248:701-702.
recognition by CD8+ CTLs is class I
Doherty PC, Zinkernagel RM. A biological role for the major histocompatibility antigens. Lancet 1975;1:1406-1409. MHC restricted.
page 381

1974 Peter Doherty, Rolf Zinkernagel discovery of how the cellular immune system recognizes virus-infected cells
Csar Milstein (1927-2002) Georges Khler (1946-1995)
The history of the development of monoclonal antibodies suffers from two issues that went unresolved for many years: first, there are multiple, conflicting claims of primacy,
and second, there is great criticism that the key discovery, with its great commercial value, was not protected by patents. In the 1970s, it was known that the B-cells that
proliferate in multiple myelomas produce antibodies with unique (and unknown) specificities, but it was not possible to produce identical antibodies specific to a given antigen.
Despite the controversy over primacy, the work of Csar Milstein and Georges Khler from the Medical Research Council Laboratory of Molecular Biology at Cambridge
was found to be so crucial, so innovative that they were awarded the 1984 Nobel Prize in Physiology or Medicine. Their key idea was to fuse cultured immortal myeloma
cells with healthy antibody-producing B-cells the resulting fused cells were called hybridomas. Milstein was experienced in culturing mouse myeloma cells that produced
immunoglobulins of unknown specificities; determining these unknown specificities was a major research focus in his and many other labs. Khler, a postdoc in Milsteins
lab, carried out the fusions of myeloma cells with antibody-producing B-cells (spleen cells from immunized mice). The resulting hybridomas combined two attributes: the
immortal properties of the myeloma cells, and the specificity of the antibody produced by the B-cells. The first hybridoma (designated P3-X63 Ag8) and the next two produced
monoclonal antibodies against different antigens of sheep red blood cells, a good choice due to the simplicity and sensitivity of available assays. For the first time, large
quantities of a pure, specified antibody could be produced. The discovery revolutionized immunology, and the infectious disease sciences including virology, and generated
many clinical and diagnostic applications. The last sentence of their first paper in Nature states, Such cultures could be valuable for medical and industrial use. To some
extent, for some years after the discovery the failure by the British Medical Research Council to protect this value with patents overshadowed the significance of the original
research, but today the scholarly merit of the discovery is fully recognized.
Khler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975;256:495-497.
Tansey EM, Catterall PP. Monoclonal antibodies: a witness seminar in contemporary medical history. Med Hist. 1994; 38: 322-327.
Milstein C. The hybridoma revolution: an offshoot of basic research. BioEssays 1999;21:966-973. page 382

1974 Georges Khler, Csar Milstein development of monoclonal antibodies


Pioneering studies by Vigdis Torsvik, Wolfram Zillig and their colleagues identified the
first viruses of archaeal organisms [although the Archaea resemble Bacteria in their
cellular structure and genome organization, their DNA replication, transcription and
translation machineries and several other qualities are unique they represent a separate
kingdom, with several phyla]. Although initial studies suggested that the viruses were
similar to tailed bacteriophage like T2, further research revealed morphologies and
genomes indicative of the need to create at least seven new families. Some of the viruses
are rod-shaped, some with claw-like end structures, others are icosahedral, some with
large turrets or spikes at vertices, while yet others are fusiform or lemon-shaped or
bottle-shaped. All but one of the viruses contain double-stranded DNA genomes, but
their genomes contain few recognizable genes suggesting that they might have unique
evolutionary origins.
Torsvik T, Dundas ID. Bacteriophage of Halobacterium salinarium. Nature 1974;248:680-
681.
Zillig W, Reiter W-D, Palm P, Gropp F, Neumann H, Rattenberger M. Viruses of Archaea.
In: Calender R, editor. The Bacteriophages. New York: Plenum Press;1988. p. 517-558.
Prangishvili D, Forterre P, Garrett RA. Viruses of the Archaea: a unifying view. Nature
Rev Microbiol. 2006;4:837-848.
Lawrence CM, Menon S, Eilers, BJ, Bothner B, Khayat R, Douglas T, Young MJ.
Wolfram Zillig (1925-2005) Vigdis Torsvik Structural and functional studies of archaeal viruses. J Biol Chem. 2009;284: 12599-
12603.

David Prangishvili Patrick Forterre


Some of the viruses of Archaea, each representing a unique taxon
page 383

1974 Wolfram Zillig, Vigdis Torsvik, Ian Dundas discovery of viruses of Archaea, many with odd morphologies
In a famous paper from 1842, Rigoni Stern
analyzed death certificates of women in Verona
and noted a high frequency of cervical cancer
in married women, widows and prostitutes,
but rare occurrence in virgins and nuns. He
concluded that the development of this type of
cancer is related to sexual contact. Attempts
to link cervical cancer to an infectious agent
failed until 1974, when after 10 years of effort
Harald zur Hausen and his colleagues provided
the first proof of the etiologic role of human
papillomaviruses (HPVs). These studies were
grounded in anecdotal reports of rare malignant
conversion of genital warts into squamous cell
carcinomas. In 1982, zur Hausen, Lutz Gissmann
and Ethel-Michelle de Villiers found HPV 6 DNA
in biopsies from three invasive tumors using
technology that led further to the quantitative
association of other specific virus genotypes
with cervical carcinoma and other carcinomas
(anogenital carcinomas, head and neck cancers).
Then, together with his collaborators, zur Hausen
identified HPV16 and HPV18 in cervical cancers.
Today, it is known that 99.7 % of cervical cancers
are a result of human papillomavirus 16 and
18 infections this is the case, even though
there are approximately 138 distinctive human
papillomaviruses (as discriminated by PCR and
full genome sequencing). Over 5% of all human
cancers, globally, can be attributed to HPVs. zur
Hausens research led directly to the development
Harald zur Hausen of HPV vaccine, which was introduced in 2006.
zur Hausen H, Meinhof W, Scheiber W,
Bornkamm GW. Attempts to detect virus-
specific DNA in human tumors. I. Nucleic acid
hybridizations with complementary RNA of
human wart virus. Int J Cancer. 1974;13:650-656.
zur Hausen H. Condylomata acuminata and
human genital cancer. Cancer Res. 1976;36:794.
Drst M, Gissmann L, Ikenberg H, zur Hausen H.
A papillomavirus DNA from a cervical carcinoma
and its prevalence in cancer biopsy samples from
different geographic regions. Proc Natl Acad Sci
Global cervical carcinoma mortality USA. 1983;80:3812-3815. page 384

1974 Harald zur Hausen, colleagues discovery of association of human papillomaviruses and cervical carcinoma
Hans Gelderblom Porcine circovirus, negative contrast electron microscopy
Porcine circovirus was first recognized as a contaminant of the widely used pig kidney cell line PK-15 in 1974 in Germany and
described as picornavirus-like. The virus, and now all its fellow members of the family Circoviridae, is very small (the smallest viruses
replicating autonomously in eukaryotic cells 16-18nm in diameter), nonenveloped, with single stranded DNA (~1,800 nt) forming
a circular genome. Initially, the virus from the cell line did not induce disease in pigs but in the 1990s strains of the virus were
associated with a newly-emerged disease syndrome in pigs described as postweaning multisystemic wasting syndrome (PMWS).
Further, many other circoviruses were discovered, all pathogens: chicken anemia virus, beak and feather disease virus of parrots,
columbid circovirus of pigeons, goose circovirus, canary circovirus. Genetic analyses strongly suggest that porcine circovirus, which
has become a global problem, is of relatively recent origin, having recently switched hosts from birds to swine. In 2010, porcine
circovirus was found to be a contaminant of human rotavirus vaccines, but since the virus is not known to cause disease in humans
or other animals the vaccines have been left on the market.
Tischer I, Rasch R, Tochtermann G. Characterization of papovavirus- and picornavirus-like particles in permanent pig kidney cell
lines. Zbl Bakt Hyg I Abt Orig A. 1974;226:153-167.
Tischer I, Glederblom H, Vettermann W, Koch MA. A very small porcine virus with circular single stranded DNA. Nature
1982;295:64-66.
Tischer I, Mields W, Wolff D, Vagt M, Griem W. Studies on epidemiology and pathogenicity of porcine circovirus. Arch Virol.
1986;91:271-276.

page 385

1974 Ilse Tischer, Hans Gelderblom discovery of porcine circovirus (the first circovirus)
Yvonne Edna Cossart Anne Field (1936-2007) The typical slapped cheek rash of fifth
disease (also called erythema infectiosum)
Yvonne Cossart graduated in medicine and then science from the University of Sydney in 1957-1959 and then completed her training as a pathologist at the Royal Postgraduate
Medical School in London. She was, by good luck during a class excursion, introduced to the virologists at the Central Public Health Laboratories at Colindale where there
was a vacancy. She took the job and stayed for 14 years, mostly working on viral hepatitis. In the mid-1970s, she noted an anomalous reaction of a normal blood donors
serum in an immunoprecipitation assay for hepatitis B (the anomaly occupied position 19 in plate B). When Cossart excised the precipitate and examined it by negative
contrast electron microscopy she saw ~25nm icosahedral virus particles this was the first human parvovirus, named parvovirus B19. Using the same immuneprecipitation
technique, antibodies to parvovirus B19 were found in a high proportion of normal adults. The most common illness caused by parvovirus B19 was identified a few years later,
during outbreaks in the United Kingdom of fifth disease (also called erythema infectiosum), a highly contagious childhood exanthem long suspected of having a viral cause.
Fifth disease takes its name from an old list of common childhood exanthems, named in the order of when they were first reported: measles, scarlet fever, rubella, Dukes
(or fourth) disease, fifth disease, and roseola infantum or exanthem subitum or sixth disease. [Most experts question the validity of fourth disease.] Parvovirus B19 has also
been implicated in several other diseases in 1981, John Pattison and his colleagues linked the virus to a severe complication of sickle cell disease in children, called transient
aplastic crisis (an aplastic anemia).
Cossart YE, Field AM, Cant B, Widdows D. Parvovirus-like particles in human sera. Lancet 1975;1:72-73.
Pattison JR, Jones SE, Hodgson J, Davis LR, White JM, Stroud CE, Murtaza L. Parvovirus infections and hypoplastic crisis in sickle-cell anaemia. Lancet 1981;1:664-665.
Anderson MJ, Lewis E, Kidd IM, Hall SM, Cohen BJ. An outbreak of erythema infectiosum associated with human parvovirus infection. J Hyg (London) 1984;93:85-93. page 386

1975 Yvonne Cossart, Anne Field, colleagues discovery of parvovirus B-19 and its association with fifth disease
C. Richard (Dick) Madeley Human astrovirus 1, negative contrast electron microscopy
Astroviruses were discovered in 1975 by C. Richard Madeley and Bonnie P. Cosgrove, using electron microscopy to examine diarrheal stool
samples from infants. The appearance of vrisions was unique and immediately distinguished them from other viruses seen in diarrheal
stools their appearance also suggested their name about 10% of the 28-35nm virions show a characteristic five- or six-pointed star
motif on their surface. Since the viruses (there are at least eight serogroups of human astroviruses) were not initially cultivable in cell
cultures, diagnosis relied heavily on electron microscopy enzyme-linked immunosorbent assay (ELISA)-based kits have now simplified
diagnostics and epidemiologic research. Several studies have found them to be the second most important cause of symptomatic
viral enteric infection in the young, worldwide (after rotavirus). Human astroviruses have also been associated with disease in the
immunosuppressed; the viruses are a common cause of acute diarrhea in patients infected with human immunodeficiency virus (HIV), and
prolonged astrovirus shedding has been observed in bone marrow transplant recipients. Astroviruses have also been found in the feces of a
variety of animals: cattle, sheep, pigs, cats, dogs, turkeys and chickens, ducks, mice, cheetahs, sea lions, dolphins and bats. Strikingly, more
than 100 genetically distinct astroviruses have been detected in various bat species.
Madeley CR, Cosgrove BP. Letter: Viruses in infantile gastroenteritis. Lancet 19;2:124-125, 1975.
page 387

1975 C.Richard Madeley, colleagues discovery of human astroviruses (the first astroviruses)
Aaron J. Shatkin (1934-2012) Bernard Moss

Capping of mRNA is an early posttranscriptional event, unique to eukaryotes, that strongly influences subsequent processing, nuclear export, stability, recognition by
ribosomes, and translation of mRNA. Accordingly, viruses of eukaryotes, whether they reside in the nucleus or cytoplasm, must solve the capping problem for efficient
replication. The 5 cap (also termed an RNA 7-methylguanosine cap), which is a modified guanine nucleotide, is added to the 5 end of eukaryotic messenger RNAs shortly
after the start of transcription. The 5 end of the mRNA being synthesized is bound by a cap-synthesizing complex associated with RNA polymerase. This enzymatic complex
catalyzes the multi-step chemical reactions that are required for mRNA capping. The discovery of 5 capping of viral mRNAs followed a long, complex path, with the earliest
work done in Japan. It was greatly hindered by the primitive state of nucleic acid sequencing at the time. In 1968, K-I. Miura spent time at the NIH with Aaron Shatkin working
on reovirus genomic RNA. He continued this work when he returned home to the National Institute of Genetics in Mishima, and brought Yasuhiro Furuichi and others into
the lab to pursue not only 5 capping but also 3 polyadenylation. However, it was several years before the phenomenon of capping became understood. This story was reviewed
by Furuichi and Aaron Shatkin in 2000. Understanding of the enzymatic mechanistics followed much later, dependent upon the great advances in sequencing in recent years.
Miura K-I, Watanabe K, Sugiura M, Shatkin AJ. The 5-terminal nucleotide sequence of the double-stranded RNA of human reovirus. Proc Natl Acad Sci USA. 1974;71:3979-
3983.
Furuichi Y, Morgan M, Muthukrishnan S, Shatkin AJ. Reovirus mRNA contains a methylated, blocked 5-terminal structure: m7G(5)ppp(5) GmpCp. Proc Natl Acad Sci USA.
1975;72:362-366.
Wei CM, Moss B. Methylated nucleotides block 5-terminus of vaccinia virus mRNA. Proc Natl Acad Sci USA. 1975;72:318-322.
Furuichi Y, Shatkin AJ. Viral and cellular mRNA capping: Past and prospects. Adv Virus Res. 2000;55:135-184.
page 388

1975 Aaron Shatkin, Bernard Moss, colleagues discovery of 5 cap on messenger RNAs (vaccinia virus, reovirus)
Edwin Southern developed the southern blot technique in 1973 while at Edinburgh University,
but his paper was not published until 1975 because the Journal of Molecular Biology didnt
think it was interesting or significant. He spent two more years performing experiments to
show that his method was important. The method is used for the detection of specific DNA
sequences in DNA samples, say the genomic DNA of viruses or reverse-transcribed DNA
of RNA viruses. Other blotting methods (i.e., Western blot, Northern blot) employ similar
principles, but are used to detect RNAs and proteins they have been named playing off
Southerns name. Southern blots were also used for genetic fingerprinting (e.g.,paternity
testing, forensic testing) prior to the development of microsatellite markers (RFLPs) for
these purposes. The concept of the Southern blot was also used in the creation of microarray
technology. When double-stranded DNA molecules are separated into single strands, by heat
denaturation, and then allowed to reanneal, the efficiency of hybridization between any two
strands is dependent upon their sequence complementarity. This is the central principle upon
which the Southern blot and many other DNA detection techniques are based. In order to
detect whether a specific sequence, such as particular viral gene, is present in a DNA sample, a
probe is made, consisting of the strand complementary to the sequence of interest. The probe
contains a label to indicate the presence of the target sequence. More specifically, Southerns
technique is carried out by first cutting the DNA specimen into manageable fragments
by digestion with restriction endonucleases. Then, the restriction fragments generated
are separated by agarose gel electrophoresis. Next, they are denatured with a strong alkali
solution. The DNA fragments are now single-stranded but remain in the gel matrix in their
original position as determined by their size. Prior to Southerns technique, DNA fragments
were not immobilized in the gel and were frequently lost during the detection process. The
beauty of Southerns technique is that the single-stranded fragments are transferred onto
a more robust membrane where they can be permanently immobilized. It is this transfer
Edwin Mellor Southern that is the blotting step. The agarose gel is placed in contact with a membrane (originally
nitrocellulose, now nylon) and a blotting paper is overlaid; the ends of the blotting paper are
placed in a strong salt solution and a mass of dry highly absorbent paper is placed on top of
the membrane and weighted down. The salt solution is drawn up by capillary diffusion into
the blotting paper and then passed through the gel and membrane into the absorbent paper.
As the solution passes between the gel and the membrane, it carries the single-stranded DNA
and vertically transfers it to the membrane where it is immobilized. Large DNA fragments are
transferred inefficiently and may move horizontally leading to blurred bands in the final result;
in order to reduce this, DNA fragments are nicked by exposing the membrane to UV light.
Negatively charged DNA sticks to the positively charged membrane but cannot pass through
it. The fragments may be permanently immobilized in the membrane by baking or cross-
linking with UV light. Probes (usually about 750 bp in length) are chosen with high DNA
binding efficiency they may be a cDNA clone, a genomic DNA fragment, an oligonucleotide
of a known sequence that is complementary to the target DNA, etc., and may be labeled,
usually with 32P or with digoxigenin, biotin, or fluorescent-tagged nucleotides. Once the probe
has hybridized with the target sequences on the membrane, detection of radio-labeled probes
involves exposure of the membrane to X-ray film; detection of other labeled probes usually
involves various light-generating methods.
Southern EM. Detection of specific sequences among DNA fragments separated by gel
page 389 electrophoresis. J Mol Biol. 1975;98:503-517.

1975 Edwin Southern development of southern blotting


Ebola virus
negative contrast
Karl M. Johnson Patricia A. Webb (1925-2005) electron microscopy,
from the first cassette,
among the first few
micrographs taken,
the day the virus was
discovered at CDC,
1976

Johnson KM, Lange JV, Webb PA, Murphy FA. Isolation and partial
characterization of a new virus [Ebola virus] causing acute hemorrhagic
fever in Zaire. Lancet 1977;309:569-571.
Murphy FA, van der Groen G, Whitfield SG, Lange JV. Ebola and
Marburg virus morphology and taxonomy. In: Pattyn SR, editor. Ebola
virus haemorrhagic fever. Amsterdam: Elsevier; 1978.
Pattyn SR, editor. Ebola virus haemorrhagic fever. Proceedings
of an international colloquium on ebola virus infection and other
haemorrhagic fevers held in Antwerp, Belgium, 6-8 December, 1977.
Frederick A. Murphy Guido van der Groen Amsterdam: Elsevier; 1978. page 390

1976 Karl Johnson, Patricia Webb, Frederick Murphy, Guido Van der Groen, others discovery of Ebola virus
The Discovery of Ebola Virus, 1976 gloves) and working in a hood (CDCs old Biological Safety Cabinet, BSC, quite primitive
by todays standards), I prepared specimens for examination by negative contrast electron
The story of the discovery of Ebola virus starts with a history of the development of
microscopy. I put the electron microscopy grids in a sealed box, told the staff not to go
biocontainment facilities at the U.S. Centers for Disease Control. After working on
near the hood room where I had worked and trundled off to the electron microscope lab.
Marburg virus in 1967, in the midst of the lethal outbreaks in Europe it became clear that
What I saw raised the hairs on the back of my neck! I saw telltale long filaments and was
CDC needed permanent biocontainment facilities, so plans were drawn up for a glove-box
sure it was Marburg virus. I was the only one still at CDC who had worked on Marburg
containment lab. It was constructed in a tin building, the Maximum Security Laboratory,
virus when it appeared in 1967. I had also done a project on Marburg virus in 1977 with
otherwise known as Building 8, located adjacent to the main viral diseases laboratory
David Simpson and his colleagues from Porton Down, the British high containment
building and was opened in 1968. Completion of the Building 8 facility coincided with
lab, so I thought I really knew enough about this unique virus. I shut down the electron
the emergence of Lassa fever in West Africa. Work on Lassa virus at Yale University
microscope and went back to the room in which I had prepared the specimen. I cloroxed
(Yale Arbovirus Research Unit) was terminated after one person died and another, the
the hell out of the hood where I had done the preparation, carried my discard pan with
famous virologist, Jordi Casals, nearly died. Immediately, I was put in charge and worked
gown and gloves, etc., to the autoclave and ran it myself. Then I went back to the electron
with Tom Monath and Bill Gary conducting the experiments that began to unravel the
microscope and called Karl and Patricia to come and take a look. I shot a cassette of
natural history of Lassa virus. Tom Monath later moved to West Africa and discovered the
pictures and with wet negatives (not good for the enlarger) I made prints, which were
reservoir host of the virus, the rodent, Mastomys natalensis, and showed how prevalent
available within minutes.
the human infection was. The Building 8 Laboratory became a part of my Viral Pathology
Branch, in the Division of Viral Diseases. The work expanded from an emphasis on Along with Karl I carried these dripping prints to the office of the director of the CDC
diagnostics to work on the pathogenesis of the viral hemorrhagic fevers. Washington (I cannot remember why we did not stop at other offices in the chain of command).
Winn and David Walker later developed animal models of several of the highly pathogenic It was a dramatic moment: the director, David Sencer, was sitting at the far end of the
human viruses, including Lassa virus. By 1976, Karl Johnson and Patricia Webb were very long table in the conference room along with a guest. He said hed be with us in a
scheduled to take over the Maximum Security Laboratory and a new facility. The latter moment. As we gathered at the other end of the table, others started coming in and it
was another tin building, a gift from the NIH it had never been used and delay followed got a bit noisy as plans were discussed about what to do next. When the noise got out
delay in completing it, all the while Karl becoming more and more frustrated [it was not of hand, David Sencer and his guest joined us. The dripping prints seemed to say it all.
completed until 1977 this was to be the first high containment lab to make use of space Marburg virus was the only virus that looked like the virus particles in the pictures.
suits (positive pressure personnel protection suits), invented by Karl Johnson]. As we sat at the table discussing the ramifications of our findings, Patricia came in and
whispered in Karls ear. He turned and repeated her findings: Patricia had done a two-
In the meantime, in October 1976, Ebola virus appeared while we were still using the old
way cross immunofluorescent antibody test, using the newly isolated virus, a reference
Maximum Security Laboratory (MSL / Building 8), but at least we had Karl and Patricia
Marburg virus, serum from one of the surviving Zaire patients and a reference Marburg
Webb with all their experience and wisdom, and we had the great technical skills of Bill
antiserum. The two did not cross: the virus causing the epidemic in Zaire was something
Gary, Jim Lange and Herta Wulff. There were two nearly simultaneous epidemics of
new. We went across the hall and David Sencer called James Gear, director of the national
hemorrhagic fever in Zaire (now Democratic Republic of Congo) and the Sudan. The
laboratory in Johannesburg, South Africa, the most senior public health infectious disease
outbreak in the Sudan was as dramatic as that in Zaire, but it is the latter that stands out
figure in Africa. Plans for a team to go to Zaire were formulated. Incredible excitement,
in my memory as if it were yesterday. We heard that the Zaire epidemic was centered at
based in sound reasoning and experienced planning, dominates my memory. David
a missionary hospital in the village of Yambuku and seemingly all the staff of the hospital
Sencers guest, who joined in the discussion as if he were an old hand turned out to be the
had died. The whole region was being quarantined by the Zaire army and there were
then junior Senator from Georgia, Sam Nunn.
rumors of devastation over a large area. CDC sent a team into the area and they found
that things were bad enough, but not quite as bad as the rumors. Between the Zaire and Within days, Karl was appointed leader of a WHO international field team and left with
Sudan epidemics there were 550 cases of severe hemorrhagic fever and 430 deaths. The a large international entourage for Kinshasa and Yambuku. Patricia and I and our staffs
CDC team was delayed by a regional quarantine in getting out with their specimens. On continued to work for the next several months on what was soon to be named Ebola virus.
October 11, specimens were received in Atlanta and Patricia Webb used all of her acumen The rest of the story is well described in Richard Prestons book, The Hot Zone.
to save the day. When she opened the package she found that the tubes containing blood One of the dripping electron micrographs from that first cassette was the image that
and tissues from patients were broken. Instead of taking the whole box to the autoclave, became rather famous. It is the shepards crook image of an Ebola virion that is now
she carefully (wearing a single pair of surgical gloves, surgical gown and mask) squeezed all over the Internet, the image that high school students often still ask me for. Let me
out a drop of fluid from the surrounding cotton packing material. She inoculated this into add one prideful caption in this regard: years later my dear friend Karl Johnson, chatting
Vero cell cultures, along with a double dose of antibiotics. with a group of old friends, said, Fred is not really a scientist, he is just a photographer, a
Two days later the cells showed traces of cytopathology, so she gave me a few drops of the photographer who uses very big, very expensive cameras. So true!
supernatant fluid from the cultures. Wearing the same sort of garb (surgical gown, mask,
Frederick A. Murphy, from his unpublished personal family memoir, 2004.
page 391

1976 Karl Johnson, Patricia Webb, Frederick Murphy, Guido Van der Groen, others discovery of Ebola virus
Between 1970 and 1984, Harold Varmus and J. Michael Bishop set out to
resolve one of the great contemporary challenges in biomedical research: to
understand how retroviruses cause cancer. During the 1960s, Howard Temin had
hypothesized that retroviruses made double-stranded DNA copies of themselves
(termed proviruses), which could be inserted into the DNA of infected cells;
he suggested that proviruses implanted oncogenes in the DNA of host cells,
resulting in cancer. This was the era of Robert Huebners virogene-oncogene
hypothesis, that is, that all forms of cancer arise from the activation of oncogenes
embedded in normal cells through infection of germline cells by retroviruses.
Varmus and Bishop set out to add experimental detail to these hypotheses: to
detect proviruses in infected cells and to track these proviruses in time and
place. They chose Rous sarcoma virus (RSV), a retrovirus that triggers cancer
in chickens and carries the first known viral oncogene, src. They set out first to
demonstrate the presence in the genomic DNA of chicken cells sequences that
derived from the RSV src oncogene a needle in a haystack in the days before
restriction mapping and gene cloning. Varmus devised a radioactive molecular
probe that could identify src genes amid the mass of host cell DNA. Working
with Dominique Stehelin, they made the surprising discovery that the RSV src
oncogene was nearly identical to a sequence in the normal cellular DNA of several
different species of birds. Then, working with Deborah Spector, they found a
src proto-oncogene in fish as well as in several mammals, including mice, cows,
and humans. They hypothesized that the many different forms of cancer all arise
from a common genetic mechanism involving specific genes present in normal
cells of all species. When these normal cellular genes undergo mutations, the
J. Michael Bishop and Harold E. Varmus result can be cancer. Their work refocused thinking on the ultimate origins of
cancer, directing attention away from the viral genome to the host genome. The
fact that src sequences may be found in the normal genome of a wide range of
species and had been there through evolutionary species divergence indicated
that src originated in normal cells; it was captured from the genome of a host
cell by an invading retrovirus far in the evolutionary past in an event known as
viral transduction. The process by which retroviruses capture host cell proto-
oncogenes damages these genes in ways that can turn them into full-fledged
oncogenes, capable of inducing malignant growth directly when the virus infects
a new host. The fact that the cellular version of src (c-src) survived through many
stages of evolution also indicated that it must perform a vital function, most likely
in directing the growth and development of cells, although its precise role was not
at first understood. From all this, Varmus and Bishop also reasoned that src was
the archetype of an array of genes that give rise to cancer, and that the oncogenes
Varmus HJ, Bishop JM, Vogt P. Appearance of virus-specific DNA in mammalian cells of other retroviruses likely also stemmed from cellular proto-oncogenes. More
following transformation by Rous sarcoma virus. J Mol Biol. 1973;74:613-626. than a hundred proto-oncogenes have since been found. Finally, Varmus and
Stehelin D, Varmus HJ, Bishop JM, Vogt P. DNA related to the transforming gene(s) of Bishop proposed that cancer can ensue when so-called tumor suppressor genes,
avian sarcoma viruses is present in normal avian DNA. Nature 1976;260:170-173. genes that control the expression of proto-oncogenes, themselves undergo
mutation or are deleted, leaving the latter free to divide unhindered. It is thus the
Spector D, Varmus HJ, Bishop JM. Nucleotide sequences related to the transforming gene
behavior of both proto-oncogenes and tumor suppressor genes that drives the
of avian sarcoma virus are present in DNA of uninfected vertebrates. Proc Natl Acad Sci
malignant growth of cancer cells.
USA. 1978;75:4102-4106. page 392

1976 J. Michael Bishop, Harold Varmus discovery of the cellular origin of retroviral oncogenes
Rudolf Rott (1926-2003) Christoph Scholtissek Influenza virus and host cell
After influenza viruses were found to have a segmented genome, reassortants obtained in the laboratory became invaluable tools to address many important problems in
influenza virology. The polygenic nature of influenza virus pathogenicity was unraveled over several years, initially by the work of Kenneth Fraser, Edwin Kilbourne and others,
and then in great depth by Rudolf Rott and Christoph Scholtissek, working in the veterinary faculty (Fachbereich Veterinrmedizin, der Justus-Liebig-Universitt), Giessen,
Germany. The latter made reassortants between highly virulent and avirulent strains of avian influenza virus, exchanging separately each of the eight genome segments of each
virus strain. The virulence of the progeny viruses they obtained varied in complex patterns. They showed that some genes worked better together than others, that there was an
optimal combination of genes, an optimal gene constellation, which favored viral survival in nature and determined virulence. They recognized the preeminent role of the viral
hemagglutinin (and its enzymatic cleavability and consequent activation) and the related role of the viral neuraminidase, but they also found that the three parts of the virion
polymerase complex played important roles in determining the virulence of reassortants. Importantly, Rott and Scholtissek initiated the distinction of the two key phenotypic
characteristics of influenza (and all other)viruses virulence (the relative pathogenicity of each virus strain) and transmissibility (the relative capacity of each virus strain to
cause epidemics). To this, we may add other phenotypic characteristics of various strains that are important, such as ease of adaptation and virus yield for vaccine production,
ease of evasion of host immune mechanisms, etc.
Scholtissek C, Harms CE, Rohde W, Orlich M, Rott R. Correlation between RNA fragments of fowl plague virus and their corresponding gene functions.
Virology 1976;74:332-344.
Rott R, Orlich M, Scholtissek C. Attenuation of pathogenicity for fowl plague virus by recombination with other influenza A viruses nonpathogenic for fowl: Nonexclusive
dependence of pathogenicity on hemagglutinin and neuraminidase of the virus J Virol. 1976;19:54-60.
page 393

1976 Rudolf Rott, Christoph Scholtissek concept that an optimum constellation of genes determines virulence
Criteria for Proof of Disease
Causation 1976
A unified concept appropriate for viruses
as causative agents of disease based on the
HenleKoch postulates, by Alfred S. Evans,
1. Prevalence of the disease is significantly higher
in subjects exposed to the putative virus than in
those not so exposed.
2. Incidence of the disease is significantly higher
in subjects exposed to the putative virus than in
those not so exposed (prospective studies).
Springer-Verlag 3. Evidence of exposure to the putative virus is
1993 present more commonly in subjects with the
disease than in those without the disease.
4. Temporally, the onset of disease follows
exposure to the putative virus, always
following an incubation period.
5. A regular pattern of clinical signs follows
exposure to the putative virus, presenting a
graded response, often from mild to severe.
6. A measurable host immune response, such as
an antibody response and/or a cell-mediated
response, follows exposure to the putative
virus. In those individuals lacking prior
experience, the response appears regularly, and
Plenum in those individuals with prior experience, the
Fourth Edition response is anamnestic.
Alfred S. Evans (1918-1996) 1997 7. Experimental reproduction of the disease
The classic concept of necessary criteria to prove the causation of an infectious disease was elaborated by Jakob Henle (1809- follows deliberate exposure of animals to the
1885), and his student Robert Koch (1843-1910), and his student Friedrich Loeffler (1852-1915), in 1884-1890 the Henle- putative virus, but nonexposed control animals
Loeffler-Koch postulates. The postulates worked well for some bacterial diseases (e.g., anthrax, tuberculosis), but even Koch remain disease free. Deliberate exposure may
realized that they did not for other bacterial diseases (e.g., typhoid, cholera). In the latter case, he thought that fulfilling just be in the laboratory or in the field, as with
some of the criteria was satisfactory. Thomas Rivers (1888-1962) reviewed the postulates in regard to viral diseases in 1937 sentinel animals.
and found them wanting. He published a refined set of postulates, which were then edited by several other virologists as 8. Elimination of the putative virus and/or its
the technologies of virology advanced, but seemingly at each step along the way new viral diseases came along that defied vector decreases the incidence of the disease.
the current postulates (e.g., mononucleosis and Epstein-Barr virus, AIDS and HIV; cervical carcinoma and certain human
papillomaviruses). The next major step was taken by Alfred Evans, who in 1976 revisited the postulates and proposed a set 9. Prevention or modification of infection, via
that included more solid epidemiologic criteria (list). This set of postulates became widely used for many years, up until the immunization or drugs, decreases the incidence
need to add criteria based in the new, central role of molecular biology in all etiologic research. of the disease.

Evans AS. Causation and disease: the Henle-Koch postulates revisited. Yale J Biol Med. 1976;49:175-195. 10. The whole thing should make biologic and
epidemiologic sense.
Evans AS. Causation and disease: a chronological journey. Am J Epidemiol. 1978;108:249-258. page 394

1976 Alfred Evans criteria for proof of viral disease causation: the Henle-Koch postulates revisited again
The 1976 Swine Flu Episode (the Swine Flu Fiasco)
In February 1976, Private David Lewis, aged 18, collapsed and died at Ft. Dix, New Jersey. Five additional patients
developed respiratory illness in the next week (eventually there were 155 similar cases). Samples were sent to
the Influenza Branch at CDC; it was quickly determined that the illness in the soldiers had been caused by an
H1N1 virus, that is a swine influenza virus, then thought to be similar to the virus that caused the 1918-19
pandemic in which 500,000 Americans died. The implications of the discovery were complicated by a number of
unfounded scientific inferences about the virus, but in any case it was suggested that there was the possibility of
an extremely severe flu epidemic and many deaths the following winter and that it was crucial to do something
quickly, namely mount an emergency vaccination program. These inferences first developed in the Influenza
Branch, then in the CDC directors office (David Sencer, director), then in the Office of the incoming Secretary
of the Department of Health, Education and Welfare (Joseph Califano, secretary designate) and finally in the
White House (Gerald Ford, president). At the same time, many informal meetings held in the hallways and
offices of the Division of Viral and Rickettsial Diseases of CDC, and elsewhere, urged caution. The failure of
the virus to take off, to infect more soldiers at Ft. Dix, was seen as similar to other circumstances when swine
influenza had been seen in the past: in such instances there were a few human infections, often with serious
clinical presentation, but nothing more. Such virologic and epidemiologic chatter was more removed from the
political scenes being played out at high levels than most virologists realized. In any case, President Ford made a
decision to initiate a massive immunization program. A group of well-known scientists supported the program
to vaccinate all 196 million Americans. Albert Sabin and Jonas Salk, who were not normally the best of friends,
appeared with President Ford on television. Congress approved funds totaling $135 million and 150 million
doses of vaccine were prepared, 40-45 million doses of which were used. Within weeks, cases of Guillain-Barre
syndrome were seen and vaccination was stopped a total of 4,181 claims were filed, amounting to $3.2 billion
[but by 1993 only $93 million had been paid out]. Articles in the Atlanta Constitution of those days describe the
rest of the story: David Sencer [the director of CDC] was at the peak of his powers, 52 years old, with a proven
track record When Jimmy Carter was inaugurated, he made Joseph Califano Secretary of Health, Education,
and Welfare. Califanos first act was to call Sencer to Washington and fire him The public reason was the
David J. Sencer (1924-2011) failed swine flu program and associated costs... Sencer, along with almost all proponents of the [vaccination]
program, argued that dollars were less important than lives. That is the crux of the problem. That is why Sencer
was fired That is how the CDC became a political beast rather than a scientific institution After hearing
about Sencers removal, Dr. Alexander Langmuir of Harvard University and former Chief Epidemiologist at CDC
said, This is the politicization of the CDC, and every important man is going to leave as fast as he can find a
new job. From that day on, the CDC changed from an effective, professional organization dedicated to the
control of communicable diseases into an outlet for political agendas, social engineering, and the manipulation
of public opinion The Atlanta Constitution eventually softened its tone and came to support CDC through
thick and thin, in keeping with the sense of civic pride in CDC that still pervades Atlanta. At a national level,
the episode, often called the Swine Flu Fiasco, affected all public vaccination programs and perhaps all public
health infectious disease programs, driving future leaders to a more conservative, less risk-taking, view of their
responsibilities.
Top FH Jr, Russell PK. Swine Influenza A at Fort Dix, New Jersey (January- February 1976). IV. Summary and
speculation. J Infect Dis. 1977;136 Suppl:S376-380.
Sencer DJ, Millar JD. Reflections on the 1976 swine flu vaccination program. Emerg Inf Dis. 2006;12:29-33.
Neustadt RE, Fineberg HV. The swine flu affair - decision-making on a slippery disease. Special report; 1978.
President Gerald Fords swine flu shot, 1976 Available online at http://www.nap.edu/catalog.php?record_id=12660.
page 395

1976 U.S. Government threat of swine flu epidemic, national emergency vaccination program
The 3,569 nucleotide genome of the
bacteriophage MS2, sequenced by Walter Fiers
and his colleagues in 1976
1 gggtgggacc cctttcgggg tcctgctcaa cttcctgtcg agctaatgcc atttttaatg
61 tctttagcga gacgctacca tggctatcgc tgtaggtagc cggaattcca ttcctaggag
121 gtttgacctg tgcgagcttt tagtaccctt gatagggaga acgagacctt cgtcccctcc
181 gttcgcgttt acgcggacgg tgagactgaa gataactcat tctctttaaa atatcgttcg
241 aactggactc ccggtcgttt taactcgact ggggccaaaa cgaaacagtg gcactacccc
301 tctccgtatt cacggggggc gttaagtgtc acatcgatag atcaaggtgc ctacaagcga
361 agtgggtcat cgtggggtcg cccgtacgag gagaaagccg gtttcggctt ctccctcgac
421 gcacgctcct gctacagcct cttccctgta agccagaact tgacttacat cgaagtgccg
481 cagaacgttg cgaaccgggc gtcgaccgaa gtcctgcaaa aggtcaccca gggtaatttt
541 aaccttggtg ttgctttagc agaggccagg tcgacagcct cacaactcgc gacgcaaacc
601 attgcgctcg tgaaggcgta cactgccgct cgtcgcggta attggcgcca ggcgctccgc
661 taccttgccc taaacgaaga tcgaaagttt cgatcaaaac acgtggccgg caggtggttg
721 gagttgcagt tcggttggtt accactaatg agtgatatcc agggtgcata tgagatgctt
781 acgaaggttc accttcaaga gtttcttcct atgagagccg tacgtcaggt cggtactaac
841 atcaagttaa atggccgtct gtcgtatcca gctgcaaact tccagacaac gtgcaacata
901 tcgcgacgta tcgtgatatg gttttacata aacgatgcac gtttggcatg gttgtcgtct
961 ctaggtatct tgaacccact aggtatagtg tgggaaaagg tgcctttctc attcgttgtc
1021 gactggctcc tacctgtagg taacatgctc gagggcctta cggcccccgt gggatgctcc
1081 tacatgtcag gaacagttac tgacgtaata acgggtgagt ccatcataag cgttgacgct
1141 ccctacgggt ggactgtgga gagacagggc actgctaagg cccaaatctc agccatgcat
1201 cgaggggtac aatccgtatg gccaacaact ggcgcgtacg taaagtctcc tttctcgatg
1261 gtccatacct tagatgcgtt agcattaatc aggcaacggc tctctagata gagccctcaa
1321 ccggagtttg aagcatggct tctaacttta ctcagttcgt tctcgtcgac aatggcggaa
1381 ctggcgacgt gactgtcgcc ccaagcaact tcgctaacgg ggtcgctgaa tggatcagct
1441 ctaactcgcg ttcacaggct tacaaagtaa cctgtagcgt tcgtcagagc tctgcgcaga
1501 atcgcaaata caccatcaaa gtcgaggtgc ctaaagtggc aacccagact gttggtggtg
1561 tagagcttcc tgtagccgca tggcgttcgt acttaaatat ggaactaacc attccaattt
1621 tcgctacgaa ttccgactgc gagcttattg ttaaggcaat gcaaggtctc ctaaaagatg
1681 gaaacccgat tccctcagca atcgcagcaa actccggcat ctactaatag acgccggcca
1741 ttcaaacatg aggattaccc atgtcgaaga caacaaagaa gttcaactct ttatgtattg
1801 atcttcctcg cgatctttct ctcgaaattt accaatcaat tgcttctgtc gctactggaa
1861 gcggtgatcc gcacagtgac gactttacag caattgctta cttaagggac gaattgctca
1921 caaagcatcc gaccttaggt tctggtaatg acgaggcgac ccgtcgtacc ttagctatcg
1981 ctaagctacg ggaggcgaat gatcggtgcg gtcagataaa tagagaaggt ttcttacatg
2041 acaaatcctt gtcatgggat ccggatgttt tacaaaccag catccgtagc cttattggca
2101 acctcctctc tggctaccga tcgtcgttgt ttgggcaatg cacgttctcc aacggtgcct
2161 ctatggggca caagttgcag gatgcagcgc cttacaagaa gttcgctgaa caagcaaccg
2221 ttaccccccg cgctctgaga gcggctctat tggtccgaga ccaatgtgcg ccgtggatca
2281 gacacgcggt ccgctataac gagtcatatg aatttaggct cgttgtaggg aacggagtgt
2341 ttacagttcc gaagaataat aaaatagatc gggctgcctg taaggagcct gatatgaata
2401 tgtacctcca gaaaggggtc ggtgccttta tcagacgccg gctcaaatcc gttggtatag
2461 atctgaatga tcaatcgatc aaccagcttc tggctcagca gggcagcgta gatggttcgc
2521 ttgcgacgat agacttatcg tctgcatccg attccatctc cgatcgcctg gtgtggagtt
2581 ttctcccacc tgagctatat tcatatctcg atcgtatccg ctcacactac ggaatcgtag

Walter Fiers Frederick Sanger 2641


2701
atggcgagac
tagagtccat
gatacgatgg
gatattctgg
gaactatttt
gcaatagtca
ccacaatggg
aagcgaccca
aaatgggttc
aatccatttt
acgtttgagc
ggtaacgccg
2761 gaaccatagg catctacggg gacgatatta tatgtcccag tgagattgca ccccgtgtgc
2821 tggaggcact tgcctactac ggtttcaaac cgaatctccg taaaacgttc gtgtccgggc
In 1976, Walter Fiers and his large team at the Laboratory of Molecular Biology of the University of Ghent were 2881 tctttcgcga gagctgcggc gcgcactttt accgtggtgt cgatgtcaaa ccgttttaca
2941 tcaagaaacc tgttgacaat ctcttcgccc ttatgctgat attgaatcgg ctacggggtt
the first to determine the nucleotide sequence of a virus, the single-stranded RNA bacteriophage MS2. In 1971, 3001 ggggagttgt cggaggtatg tcagatccac gcctttacaa ggtgtgggta cgactctcct
3061 cccaggtgcc ttcgatgttt ttcggtggga cggacctcgc tgccgactac tacgtagtca
Ray Wu had published the sequence of 12 nucleotides of the cohesive ends of the DNA of bacteriophage; this is 3121 gcccgcccac ggcagtctcg gtatatacca agactccgta tgggcggcta ctcgcggata
considered to be the first DNA sequence ever determined. The first DNA viral genome to be sequenced was that of 3181
3241
cccgtacctc
agcatgacag
gggtttccgt
tggccgctac
cttgctcgta
atagcgtggt
tcgctcgaga
tccatactgg
acgcaagttc
aggtgaagtc
ttcagcgaaa
accgacagta
the double-stranded DNA bacteriophage -X174 its 5,368 base pair genome was sequenced by Frederick Sanger 3301
3361
tgaagtccgc
tccctcagga
cggcgtgcgt
gtgtgggcca
attatgcgca
gcgagctctc
cttcggagtg
ctcggtagct
gctaacgccg
gaccgaggga
gttcccacat
cccccgtaaa
and his team in 1977, using the enzymatic method Sanger had developed in 1975. Several surprising features were 3421 cggggtgggt gtgctcgaaa gagcacgggt ccgcgaaagc ggtggctcca ccgaaaggtg
3481 ggcgggcttc ggcccaggga cctccccttg aagagagggc ccgggattct cccgatttgg
identified, including genes that partially overlap one another and the first clues that different organisms might have
3541 taactagctg cttggctagt taccaccca

slightly different codon usage patterns. The Sanger enzymatic sequencing method went on to become the basis for
all later sequence methods, including todays automated, high-throughput methods. In 1978, Walter Fiers and his team also reported the
Fiers W, Contreras R, Duerinck F, Haegeman G, Iserentant D, Merregaert J, Min Jou W, Molemans F, Raeymaekers complete sequence of the genome of SV40 virus5,224 bp:
A, Van den Berghe A, Volckaert G, Ysebaert M. Complete nucleotide-sequence of bacteriophage MS2-RNA - W. Fiers, R. Contreras, G. Haegeman, R. Rogiers, A. Van
primary and secondary structure of replicase gene Nature, 1976;260:500-507. de Voorde, H. Van Heuverswyn, J. Van Herreweghe, G.
Sanger F, Air GM, Barrell BG, Brown NL, Coulson AR, Fiddes CA, Hutchison CA, Slocombe PM, Smith M. Volckaert & M. Ysebaert. Complete nucleotide sequence
Nucleotide sequence of bacteriophage phi X174 DNA. Nature 1977;265: 687-695. of SV40 DNA. Nature 273:113-120, 1978.
page 396

1976-1977 Walter Fiers, Frederick Sanger complete sequencing of viral genomes (bacteriophages MS2 and 174)
Phillip A. Sharp Richard J. Roberts Richard Roberts and Phillip
Sharp made their discovery
By the 1970s the physical structure of the gene was firmly established from work on bacteria. The sequence of the DNA, the RNA, and the
of split genes in adenovirus
protein were thought to be colinearly organized and expressed what was true for bacteria must be true for higher forms of life. However,
when they examined a hybrid
more and more evidence began to appear that implied that this might not be universal. This led Phillip Sharp at MIT and Richard Roberts
between a viral mRNA and its
at Cold Spring Harbor Laboratory and their colleagues (Louise Chow, Richard Gelinas, Thomas Broker, Susan Berget, Claire Moore, Arnold
template DNA in the electron
Berk, Tim Harrison) to compare the complementarity of sequences expressed as cytoplasmic mRNAs during the late stage of adenovirus
microscope. They observed that
infection with the viral DNA from which it was transcribed. Hybridization of the mRNA encoding the adenovirus virion hexon protein
the mRNA was not encoded
to a restriction endonuclease fragment of the adenovirus genome generated a structure which by electron microscopy was seen to have
co-linearly in the DNA Instead,
three different length loops of viral DNA. That is, the single mRNA molecule corresponded to at least three well-separated segments in the
loops of unhybridized DNA
DNA molecule. Roberts and Sharp came to the conclusion that the genetic information in the gene was discontinuously organized in the
(A, B, C) were seen. The
genome. The discovery immediately led to intensive research by which the phenomenon was found to be present also in other viruses and in
interpretation was that mature
vertebrate cells. The discovery of split genes and RNA splicing also led to the concept of introns and exons, terms coined by Walter Gilbert.
messenger RNA was derived
Berget SM, Moore C, Sharp PA. Spliced segments at the 5 terminus of adenovirus 2 late mRNA. Proc Natl Acad Sci USA. 1977;74:3171- from discontinuous segments
3175. (exons) in the viral DNA. The
Chow LT, Gelinas RE, Broker TR, Roberts RJ. An amazing sequence arrangement at the 5 ends of adenovirus 2 messenger RNA. Cell intervening sequences (introns
1977;12:1-8. A, B, C) are excised during
page 397 mRNA maturation.

1977 Phillip Sharp, Richard Roberts, colleagues discovery of RNA splicing and split genes (adenovirus)
WHO
Smallpox
Eradication
Programme
D. A. Henderson Frank Fenner (1914-2010) Isao Arita
In 1958 Victor Zhdanov, Deputy Minister of Health of the USSR, called upon the World Health Assembly to undertake a global
program to eradicate smallpox; the proposal was accepted in 1959. At this point, 2 million people were dying from smallpox every year.
At first, progress was disappointing, so in 1967 the WHO formed a smallpox eradication unit, under the leadership D.A. Henderson,
with Isao Arita serving as the senior WHO staff member. The unit established a network of consultants who assisted in setting up
programs in all countries where smallpox was still present. The program centered on surveillance of cases (the most important failing
in most countries), the isolation of cases and the vaccination of everyone who lived close by cases (ring vaccination). Early on,
vaccine was provided by the United States and the Soviet Union, but by 1973 more than 80% of all vaccine was produced in developing
countries. The last major European outbreak occurred in 1972 in Yugoslavia. By the end of 1975, the disease persisted only in the Horn
of Africa Ethiopia and Somalia, where the program faced incredible challenges: poor roads and transport, civil war, famine, and many
refugees in transit. The last naturally occurring case of variola minor was diagnosed in Ali Maow Maalin, in Merca, Somalia, in 1977.
The last naturally occurring case of variola major had been detected in Rahima Banu, in Bangladesh, in 1975. In 1977, Frank Fenner
was named the chairman of the Global Commission for the Certification of Smallpox Eradication eradication was certified in each
country based upon intense on-the-ground searching for cases. It was Fenners great honor to announce to the World Health Assembly
in 1980 that global eradication had been achieved. His announcement was accepted unanimously by the World Health Assembly the
same day. He later said that making this announcement stood out most among his many achievements.
Fenner FD. Henderson DA, Arita I, Jeek , Ladnyi ID. Smallpox and its eradication. Geneva: World Health Organization; 1988.

Ali Maow Maalin, Merka, Somalia,


the last known natural case of smallpox page 398

1977 D.A. Henderson, Isao Arita, Frank Fenner, many others global eradication of smallpox
The first DNA sequences were obtained in the early 1970s
using laborious methods based on two-dimensional
chromatography. Then in 1975-77, Frederick Sanger at the
University of Cambridge invented a partial cleavage of end-
labeled DNA method and independently Walter Gilbert
and Allan Maxam at Harvard invented a chain termination
(dideoxy) method, both of which initiated the sequencing
revolution, even though it was the Sanger method that later
became the method of choice. Sangers method employs
dideoxynucleotide triphosphates (ddNTPs) as DNA replication
terminators this requires use of modified nucleotides
(dideoxyNTPs) that when incorporated terminate DNA strand
elongation. These ddNTPs are radioactively or fluorescently
labeled. The DNA sample is divided into four separate
sequencing reactions, each conting only one of the four chain-
terminating dideoxynucleotides (ddATP, ddGTP, ddCTP, or
ddTTP). This results in DNA fragments of varying length,
which are separated by size by polyacrylamide electrophoresis
(with a resolution of just one nucleotide); the DNA bands
are then visualized by autoradiography or UV light, and the
DNA sequence can be directly read off the X-ray film or gel
image. It is fluorescent dye-labeled ddNTPs that are read in
automated optical systems, the first invented by Leroy Hood,
that was the key in the first high-throughput machines.
Today several different methods are employed producing
thousands or millions of sequences at once (massively parallel
signature sequencing, polony sequencing, pyrosequencing,
reversible dye-terminator sequencing, sequencing by ligation,
Walter Gilbert Frederick Sanger ion semiconductor sequencing, DNA nanoball sequencing,
sequencing by hybridization, etc.).
Sangers End-labeled DNA Maxam AM, Gilbert WA. A new method for sequencing DNA.
Sequencing Method Proc Natl Acad Sci USA. 1977;74:560-564.
Single-stranded DNA is mixed with Sanger F, Coulson AR. A rapid method for determining
a primer and split into four aliquots,
sequences in DNA by primed synthesis with DNA polymerase.
each containing DNA polymerase,
four dideoxyribonucleotide J Mol Biol. 1975;94: 441-448.
triphosphates and a modified Sanger F, Nicklen S, Coulson AR. DNA sequencing with chain-
nucleotide replication terminator. terminating inhibitors. Proc Natl Acad Sci USA. 1977;74:
Each reaction proceeds until a 5463-5467.
replication-terminating nucleotide
is added. The mixtures are loaded
into separate lanes of a gel and
electrophoresis is used to separate
the DNA fragments. The sequence
of the original DNA strand is
page 399 inferred from the results.

1977 Walter Gilbert, Frederick Sanger development of the technology for rapid sequencing of DNA
In 1977, Mario Rizzetto and his colleagues in Torino,
Italy, while examining liver biopsies from individuals
infected with hepatitis B virus (HBV), discovered by
immunofluorescence a previously unrecognized nuclear
antigen that was subsequently shown to be a specific
marker of a novel human pathogen, hepatitis delta virus
(HDV). As discovered by John Gerin and his colleagues,
HDV is a unique defective RNA virus with similarities to
viroids, the subviral pathogens of higher plants. It is the
smallest animal virus (~1,679 nucleotides) and the only
RNA animal virus to possess a circular RNA genome.
For its replication the virus requires the helper function
of hepatitis B virus (HBV), which provides the essential
coat protein for virion assembly. Both superinfection
and coinfection of HBV carriers with HDV results in a
greater likelihood of experiencing liver failure in acute
infections and a rapid progression to liver cirrhosis,
with an increased chance of developing liver cancer
in chronic infections. In combination with hepatitis B
virus, hepatitis D has the highest mortality rate of any
the hepatitis infection ~20%. There are currently an
estimated 15 million HDV carriers worldwide, but there
has been a dramatic decline in the incidence has been
observed in the last decade, likely as a result of HBV
vaccination programs and improved socioeconomic
conditions.
Rizzetto M, Canese MG, Arico S, Crivelli O, Bonino F,
Mario Rizzetto John L. Gerin Trepo CG, Verme G. Immunofluorescence detection of a
new antigen/antibody system (d /anti-d) associated with
hepatitis B virus in liver and serum of HBsAg carriers.
Global Prevalence of HDV in HBsAg Positive People Gut 1977;18:997-1003.
Hess G, Shih JW, Kaplan PM, Gerin JL. The
demonstration of subtype (D or Y)-specific determinants
on the surface of the presumed hepatitis B virus. J
Immunol. 1977;119:1542-1544.
Rizzetto M, Shih JW, Gocke DJ, Purcell RH, Verme
G, Gerin JL. Incidence and significance of antibodies
to delta antigen in hepatitis B virus infection. Lancet
1979;2:986-990.
Rizzetto M, Purcell RH, Gerin JL. Epidemiology of HBV-
Hepatitis delta virus associated delta agent: geographical distribution of anti-
HBV proteins, blue delta and prevalence in polytransfused HBsAg carriers.
HDV proteins, green/red Lancet 1980;1:1215-1218.
HDV RNA, yellow page 400

1977 Mario Rizzetto, John Gerin, colleagues discovery of hepatitis delta virus (the only deltavirus)
Dengue infection can be asymptomatic or present in two clinical syndromes, dengue fever and dengue
hemorrhagic fever/dengue shock syndrome (DHF/DSS). Plasma leakage, hemorrhage and thrombocytopenia
characterize the latter. DHF/DSS is the result of the interaction of several virus and host factors. Some dengue
virus genotypes have more potential to produce DHF/DSS than others, but in addition, several host factors
interplay with virus factors: these include age (children at higher risk than adults), race, underlying chronic
diseases, nutritional status, sex and particular allelic variants of genes that encode cellular receptors. However,
secondary infection is considered the main risk factor for DHF/DSS this concept was first published by Scott
Halstead in 1977. He proposed that preexisting low-level dengue virus antibodies can modulate subsequent
infection by enhancing the infection of mononuclear phagocytes he called this antibody-dependent
enhancement (ADE), which he went on to show can suppress the cellular immune response and thereby increase
the extent of the infection and can generate inflammatory cytokines that contribute to the development of severe
disease. Single-serotype natural infections result in lifelong immunity to the infecting serotype but only short-
term cross-protection against heterotypic serotypes. There is also evidence to suggest that over time there is a
continuous selection of neutralizing-antibodies with increasing homologous reactivity and concurrent decrease
Scott B. Halstead in heterotypic reactivity. Studies in Thailand of first-time infected infants born to dengue-immune mothers and
children who had experienced a mild or primary asymptomatic dengue infection and then became secondarily
infected by a different dengue serotype had a 15-80 times higher probability of developing DHF/DSS than
other infants and children in one study, 99% of DHF/DSS cases had heterotypic antibodies to the dengue
serotype causing the severe disease. The cellular and molecular mechanisms acting in ADE are complex and to
some extent still mysterious; many studies are centered on how antibody-virus complexes are internalized into
mononuclear cells via their Fc receptor, resulting in infection of a higher number of target cells, which in turn
may lead to even more virus production.
Halstead SB, ORourke EJ. Dengue viruses and mononuclear phagocytes. I. Infection enhancement by non-
neutralizing antibody. J Exp Med. 1977;146:210-217.
Halstead SB, ORourke EJ. Antibody-enhanced dengue virus infection in primate leukocytes. Nature
1977;265:739-741.
Halstead SB. In vivo enhancement of dengue virus infection in rhesus monkeys by passively transferred antibody.
page 401 J Infect Dis. 1979;140:527-533.

1977 Scott Halstead discovery of immune enhancement in dengue hemorrhagic fever and dengue shock syndrome
Darwin provided the first unifying theory of the nature life on Earth and drew a famous diagram expressing the
universal tree of life. The basis for constructing this tree of life was fundamentally and dramatically changed in
1977 by the work of Carl Woese and his colleagues. Morphologic determinants of relationships among species
that were in place from the days of Linnaeus were replaced by similarities of mRNA gene sequences of related
organisms. Today, the simple tree concept has been adjusted by our knowledge of the occurrence of horizontal
gene transfer, but in most published works the three-branched unrooted tree has not been adjusted by the addition
of the viruses. Several reasons have been advanced to argue against adding viruses to the universal tree of life
[from Vincent Racaniello, http://www.virology.ws/2009/03/19/viruses-and-the-tree-of-life/ summary of the
argument of David Moreira and Purificacin Lpez-Garca]: (1) Viruses are not alive; (2) Viruses are polyphyletic
they did not evolve from a common ancestor; (3) There are no ancestral viral lineages, common genes, common
proteins; (4) Viruses do not have a structure derived from a common ancestor (e.g., cell membrane); (5) Viral
metabolic genes originated from cells; (6) Viral translation genes originated from cells, again by horizontal gene
transfer; (7) Viruses are gene robbers, not gene inventors (although they are major gene suppliers); etc.
Evidence of an ancient origin of viruses is controversial because of horizontal virus gene movement; people who
wish to see the viruses added to the universal tree of life cite recent studies that indicate that the viruses infecting
all three domains, Bacteria, Archaea and Eucarya, have a very ancient origin. One notion is that there are at least
two large DNA sequence-spaces on Earth, one represented by capsid-encoding viruses and another by ribosome-
encoding cells. Despite their probable distinct evolutionary origin, both sequence-spaces were and are connected
by intensive two-way gene transfer. It remains to be seen just how the viruses would be added to the universal tree
of life, except as a fourth domain (a domain without a root).
Woese CR, Fox GE. Phylogenetic structure of the prokaryotic domain: the primary kingdoms. Proc Natl Acad Sci
USA. 1977;74:5088-5090.
Fox GE, Stackebrandt E, Hespell RB, Gibson J, Maniloff J, Dyer TA, Wolfe RS, Balch WE, Tanner RS, Magrum
LJ, Zablen LB, Blakemore R, Gupta R, Bonen L, Lewis BJ, Stahl DA, Luehrsen KR, Chen KN, Woese CR. The
phylogeny of prokaryotes. Science 1980;209:457-463.
Carl Richard Woese (1928-2012) Moreira D, Lpez-Garca P. Ten reasons to exclude viruses from the tree of life. Nat Rev Microbiol. 2009;7:306-311.

The Virosphere by Claude Fauquet: taxonomy, but few implied


Darwins notebook phylogenetic relationships among the families of viruses page 402

1977 Carl Woese, colleagues recasting of The Tree of Life, phylogeny based on mRNA gene sequences
Ho Wang Lee Pyung Woo Lee Karl M. Johnson
Hemorrhagic fever with renal syndrome (HFRS) is the contemporary name for a group of clinically similar illnesses
that occur throughout the Eurasian landmass, with many older and local names: Korean hemorrhagic fever, epidemic
hemorrhagic fever, nephropathia epidemica, et al. Although these diseases were recognized in Asia for centuries, HFRS first
came to western attention when approximately 3,200 cases occurred from 1951 to 1954 among United Nations forces in
Korea. It was not until 1978 that the rodent reservoir for the etiologic agent of Korean hemorrhagic fever was found by Ho
Wang Lee, Pyung Woo Lee and Karl Johnson, who demonstrated that patient sera reacted with antigen in frozen sections
of lung of wild-caught Apodemus agrarius field mice and that the virus in these rodents could be passed from rodent to
rodent. The virus, Hantaan (HTN) virus, was grown in cell culture in 1981, thereby providing opportunity to study this
pathogen systematically. Many related viruses are associated with the syndrome, representing a significant gradient in
pathogenicity; generally, the less virulent variants are seen in northern Europe and the most virulent in eastern Asia but
there are exceptions mortality rates vary from 0.1% to 5-10%. Approximately 150,000 to 200,000 cases of severe HFRS,
Apodemus agrarius, the field mouse that involving hospitalization, are reported each year throughout the world, with more than half in China. Russia and Korea also
is the reservoir host of Hantaan virus, the report hundreds to thousands of cases each year.
etiologic agent of Korean hemorrhagic fever
(hemorrhagic fever with renal syndrome) Lee HW, Lee PW, Johnson KM. Isolation of the etiologic agent of Korean hemorrhagic fever. J Infect Dis. 1978;137:298-308.
Johnson KM. Nephropathia epidemica and Korean hemorrhagic fever: the veil lifted? J Infect Dis. 1980;141:135-136.
Johnson KM. The discovery of hantaan virus: comparative biology and serendipity in a world at war. J Infect Dis.
2004;190:1708-1710.
Lee HW. Hemorrhagic fever with renal syndrome in Korea. Rev Infect Dis. 1989;Suppl 4:S864-76.

page 403

1978 Ho Wang Lee, Pyung Woo Lee, Karl Johnson Hantaan virus, hemorrhagic fever with renal syndrome
Leland E. (Skip) Carmichael Max J. G. Appel Colin R. Parrish
Canine parvovirus type 2 (CPV) was first recognized by Leland Carmichael and Max Appel and their colleagues in 1978; the
virus appeared almost simultaneously in the United States, Australia and Europe and spread worldwide within two years.
The highly contagious virus causes cardiac and intestinal disease with high mortality especially in puppies. At first the canine
virus was thought to be a direct host-range extension mutant of feline panleukopenia virus (FPLV), but later research proved
that the canine virus evolved from an unknown carnivore parvovirus and was the result of accumulated mutations over the
preceding 10 years, mutations which affected its capacity to infect dogs, and to spread and cause disease. In contrast to other
carnivore parvoviruses, CPV continued to evolve rapidly after its emergence. Both a high mutation rate and positive selection
of mutations in the capsid gene appear to be the driving force for such rapid evolution. The original antigenic/genetic type
disappeared more than two decades ago, being replaced by a succession of new antigenic/genetic variants that have also spread
worldwide. The phylogenetic pathway of this evolution has been studied in detail by Colin Parrish and his colleagues. Each new
variant has required reformulation of the very effective vaccines.
Appel MJG, Cooper BJ, Greisen H, Carmichael LE. Status report: canine viral enteritis. J Am Vet Med Assoc. 1978;173:1516-
1518.
Parrish CR, CarmichaelLE. Antigenic structure and variation of canine parvovirus type-2, feline panleukopenia virus, and mink
enteritis virus. Virology 1983;129:401-414.
Carmichael LE. An annotated historical account of canine parvovirus. J Vet Med Series B. 2005;52:303-311. page 404

1978 Leland Carmichael, Max Appel discovery and characterization of canine parvovirus (CPV-2)
The history of RNA virus reverse genetics, now
usually defined as the ability to recover or rescue
infectious virus from complementary DNA
(cDNA), can be traced back to the report from
Charles Weissmanns lab on the recovery of the
RNA bacteriophage, Q, from cloned cDNA:
Taniguchi T, Palmieri M, Weissmann C. Q
DNA-containing hybrid plasmids giving rise to
QB phage formation in the bacterial host. Nature
1978;274:223-228.
Synthesis of a hybrid plasmid containing a
complete DNA copy of the Q genome. Q
RNA extracted from plaque-purified phage
was used as template for the synthesis of Q
minus strands by Q replicase. Q RNA and
purified Q minus strands were elongated with
AMP residues by polyA polymerase and used
as templates for reverse transcriptase, with
oligo dT as primer. The minus and plus cDNA
copies were hybridized and the incomplete
duplex was repaired with DNA polymerase.
Plasmid PCRI was cleaved with EcoRI and the
ends were trimmed back with 5 exonuclease.
The double-stranded Q DNA and the
PCRI DNA were elongated with dAMP and
dTMP residues, respectively, using terminal
nucleotidyl transferase. The two components
Charles Weissmann were hybridized and transfected into E. coli.

Before the advent of recombinant DNA and DNA sequencing technologies, genetic analysis of viruses involved random isolation and characterization of mutants, and rough
mapping of genes affected. With the availability of full genomic sequences for virtually all of the prototypical members of each virus family, genetic analysis can be conducted
with a reasonably complete foreknowledge of the genetic structure of the virus, the individual gene(s) of interest and their general function(s). Mutations can be deliberately
engineered in genes to further study their function(s). Termed reverse genetics, this process has come to dominate the genetic analysis of viruses. Reverse genetic analysis
involves two distinct manipulations: (1) the construction of a given mutation (not reviewed further here), and (2) the method for incorporation of the mutation into the virus of
interest. The principles governing these manipulations are dependent on the structure of the viral genome of interest and the strategy of its replication, but there are common
themes. For DNA or RNA viruses with relatively small genomes and for which the nucleic acid is infectious, incorporation of a given mutation into virus is a straightforward
matter of constructing the desired mutation in a full-length genomic clone in a prokaryotic vector, then transfecting cells with the mutant nucleic acid. For example,
engineered mutations have been constructed in both picornaviruses and polyomaviruses using full-length genomes. For DNA viruses that either are too large or complex to
be manipulated as a full-length clone or that contain genomes that are not infectious, incorporation of mutations into the viral genome can be accomplished via homologous
recombination and transfection of cells with the recombinant genome. For example, this has been used successfully to construct engineered mutations in vaccinia virus and
herpes simplex virus. For RNA viruses where the genomic nucleic acid is not infectious, protocols for incorporation of mutations into virus become more complex and tailored
to particular viruses. For example, construction of influenza virus recombinants requires co-transfection of multiple plasmids expressing the wild-type and mutant genomic
RNA segments plus several influenza replication proteins, all transcribed from an RNA polymerase promoter. From: Conditt, R. Principles of virology. In: Knipe D, Howley
PM, editors. Fields virology. Philadelphia: Wolters Kluwer, Lippincott Williams & Wilkins; 2007.
page 405

1978 Charles Weissmann, colleagues first reverse genetics experiment (bacteriophage Q)


Waterson AP,
Wilkinson L.
An introduction
to the history
of virology.
Cambridge:
Cambridge
Anthony Peter Waterson University
Press;1978.
(1923-1983) Lise Slver Wilkinson (1924-2012)
The 1978 book, An Introduction to the History of Virology, by Anthony Waterson and Lise Wilkinson, is unique. It came at a time just after the emergence of virology as a
distinct scientific discipline, and yet at a time when there were still tugs from the other microbiological disciplines that virology rejoin the fold. The book played a significant
role in bolstering the conviction of virologists to press on, to continue to build an independent community and to continue to build links with other new biological sciences:
cell biology, molecular biology, structural biology, et al. The authors were in just the right place to capture this perspective: Anthony Waterson was a professor in the
Department of Virology at the Royal Postgraduate Medical School, University of London, and had worked with many leading virologists characterizing many human viral
pathogens. Lise Wilkinson was a distinguished historian, at the time a fellow in the same department, and also associated with the Wellcome Institute for the History of
Medicine. Over the years, she wrote many articles on aspects of the history of virology. The following is abridged from the introduction to the book:
[Virologys] relatively recent origin means that the sources are copious and, on the whole, accessible, even if they are somewhat diffusely spread. Very much has happened
in a short time, so that the rate and acceleration of growth of knowledge have been so fast that the direction and flow of thought have been easy to follow. The origins are
various, but easy to discern: botany, plant pathology, human and veterinary medicine, and especially that activity known as hygiene on the continent of Europe and as
bacteriology in Great Britain and the United States, whether by doctors or veterinarians, have all contributed.... It was in fact a consideration of virology as an offshoot of
bacteriology, and an experience of the difficulties of working in a young subject under the terms of reference of an old, which supplied much of the stimulus to initiate the
current study.... The very element of near impossibility with which the early virologists were faced makes the subject a fertile one for the historian. The principal difficulties
were those intrinsic to the study of very small biological particles, and the pioneers had little in their armamentarium. However, techniques did emerge. Centrifuges grew in
speed and power, and microscopes were developed until their magnification went beyond what the specimen required.... The underlying theme of this book is the evolution
of the present concept of a virus. This present-day concept is essentially a chemical one, but that is not to imply that it is divorced from the mainstream of biological thinking;
[it is a rather good] vantage point from which to look back, so that we can better look forward. It is therefore essentially apocalyptic, in the proper sense of the word, i.e.
revelatory. It is possible to study the virological scientists of the past, and to know in the light of present knowledge exactly what they were studying, even though this was
not revealed to them in their day. It is, in other words, the story of the progressive unveiling of the nature of the virus particle. This history is therefore essentially, and indeed
deliberately, conceptual. It is a study of ideas and concepts, and the inter-reactions between these, on the one hand, and experiment and technique, on the other. That it is
inextricably interwoven with the administrative, the technical and the personal goes without saying, and to compose this history involved in itself the administrative, the
technical and the personal. A catalogue may be compiled by a computer, but stories can be told only by a person.... Individual virologists figure in these pages, but it is their
thinking as much as their doing with which we have been concerned, because the lesser (doing) is included in the greater (thinking). This is, above all, a story. It is to be
hoped that, as a story, it will be read with enjoyment no less than, as a history, it will be studied with interest. page 406

1978 Anthony Waterson, Lise Wilkinson publication of the book, An Introduction to the History of Virology
The first determination of the atomic structure of a virus was of tomato bushy stunt
virus determined to 2.9 resolution it was published by Stephen Harrison and his
colleagues in 1978. Solving the structure of the icosahedral plant viruses was a major
achievement, but it wasnt clear that it had a profound impact on virologists at the
time. However, it did mean that the structure of other viruses, animal viruses, might
also be determined. Picornaviruses seemed the obvious next goal. One reason was that
poliovirus had been crystallized by Carlton Schwerdt and Frederick Schaffer in 1955.
Harrison tells the following story: Apparently Schaffer had crystallized the Mahoney
strain of polio virus and his wife was going to bring it to England in her pocketbook.
The custom agent asked her what it was and when she said it was poliovirus, he said:
Lady, you cant bring poliovirus into England. She responded: But it is crystalline.
The custom agent must have realized the importance of this as he let her pass.
Then, Jim Hogle came to Harrisons lab to learn crystallography, planning to solve
the structure of polio virus. Hogle had been a graduate student at the University of
Wisconsin where he came under the influence of Roland Rueckert. Rueckert was
responsible for connecting Hogle with Harrison for the eventual goal of solving the
structure of polio virus and also for influencing Michael Rossmann to tackle the
rhinoviruses. In 1985, Hogle and his collaborators solved the structure of poliovirus
to a resolution of 2.9. One factor in achieving this goal had been that, because of the
vaccines, it was possible to work with the virulent Mahoney strain which gave more
stable crystals than the attenuated (Sabin) strains.
Steven C. Harrison James M. Hogle Harrison SC, Olson AJ, Schutt CE, Winkler FK, Bricogne C. Tomato bushy stunt virus
at 2.9 resolution. Nature 1978;276:368-373.
Hogle JM, Chow M, Filman DJ. Three-dimensional structure of poliovirus at 2.9
resolution. Science 1985;229:1358-1365.
Rossmann MG, Arnold E, Erickson JW, Frankenberger EA, Griffith JP, Hecht HJ,
Johnson JE, Kamer G, Luo M, Mosser AG, Rueckert RR, Sherry B, Vriend B. Structure
of a human common cold virus and functional relationship to other picornaviruses.
Nature 1985;317:145-153.

tomato bushy
stunt virus poliovirus

page 407
Marie Chow Michael G. Rossman

1978 Steven Harrison, James Hogle, others structure of viruses (tomato bushy stunt virus, poliovirus, rhinovirus)
The relationship between hepatitis B virus
(HBV) and primary hepatocellular carcinoma
(HCC) was first noted in the 1970s, when it
was found that HBV antigenemia corresponded
geographically with HCC. The prevalence of
antigenemia was found to be higher in cases of
HCC than in controls in both high- and low-
incidence areas for HCC. The epidemiologic
data supporting this causal relationship were
brought together in 1978 by Wolf Szmuness. This
was greatly advanced in 1981 by the prospective
study of Palmer Beasley and his colleagues in
which the risk of HCC over a 5-year period in
persons with persistent HBV antigenemia was
found to be 217 times greater than in people
without antigenemia. This relationship was
further strengthened by laboratory studies
demonstrating HBV DNA sequences integrated
into the chromosomal DNA of tumor cells, and
then by similar findings in studies of animals
infected with their own HBV-like viruses
(woodchuck hepatitis virus, Pekin duck hepatitis
R. Palmer Beasley Lu-yu Hwang virus). All this, in turn, greatly motivated the
drive for global HBV vaccination, since there
are still ~700,000 new cases of HCC annually,
worldwide [the fifth most common cancer in
men (523,000 cases, 7.9% of the total number
of cancers reported) and the seventh in women
(226,000 cases, 6.5% of total)]. This opportunity
has been turned into a global success story.
Blumberg BS, Larouz B, London WT, Werner
B, Hesser JE, Millman I, Saimot G, Payet M. The
relation of infection with the hepatitis B agent
to primary hepatic carcinoma. Am J Pathol.
Countries with most liver cancer 1975;81:669-682.
Szmuness W. Hepatocellular carcinoma and
the hepatitis B virus: evidence for a causal
association. Prog Med Virol. 1978; 24:4069.
Beasley RP, Hwang LY, Lin CC, Chien CS.
Hepatocellular Hepatocellular carcinoma and hepatitis B virus:
carcinoma A prospective study of 22,707 men in Taiwan.
classic arrangement of tumor Lancet 1981;318:1129-1133.
cells in trabecular and acinar
patterns. H&E Wolf Szmuness (1919-1982)
page 408

1978 Wolf Szmuness, Palmer Beasley, others relation of hepatitis B virus infection with hepatocellular carcinoma
Enquist LW, Madden MJ, Schiop-Stanley P,
Vande Woude GF. Cloning of herpes simplex
type 1 DNA fragments in a bacteriophage
Lynn W. Enquist lambda vector. Science 1979;203:541-544.
Abstracted from the 1979 paper by Enquist, Madden, Schiop-Stanley and Vande Woude:
Our approach to analysis of the large, complex DNA genome of herpes simplex virus type 1 (HSV1) was to insert DNA fragments into a coliphage vector and to grow
these hybrids in Escherichia coli. Only in this way were we able to pick out single large DNA fragments from the complicated mixture and amplify them in quantities suitable
for detailed study. To insert HSV1 DNA into the vector, we were required by the NIH Guidelines on Recombinant DNA to use the most stringent biological and physical
containment available: P4 combined biological and physical containment. The basic procedure for our experiments was as follows: the vector DNA was prepared and cleaved
with the restriction endonuclease EcoR1. Fragments of the cleaved vector DNA were separated and mixed with EcoR1-cleaved HSV1 DNA and joined by means of T4 DNA
ligase. The ligated DNA was then packaged into phage particles. The recombinant phage particles were used to infect disabled E. coli K12 and lysates containing recombinant
phage DNA were concentrated and the DNA extracted a certified treatment of this product effectively inactivated the recombinant phage and bacteria. After treatment
with ribonuclease and extraction with phenol the DNA was frozen for subsequent analysis. The vector is particularly suited for insertion of EcoR1 fragments; however,
fragments less than about 1 kbp or more than 15 kbp theoretically cannot be inserted into the vector because of size limitations of the phage head and limits of the packaging
mechanism. When HSV1 DNA was cleaved by EcoR1, about 15 fragments were obtained unfortunately, most of the fragments approached or exceeded the theoretical 15-
kbp size capacity of the vector. We sought to improve our chances of inserting large fragments that were close to the theoretical size limit by a procedure in which recombinant
DNA is added to a mixture of partially assembled virions, and the added DNA is encapsidated to form viable phage particles. In this procedure the larger DNA molecules are
preferentially packaged over smaller ones. Represented among the recombinant DNA molecules studied were fragments from about 50% of the HSV1 genome. The ability to
make large quantities of specific HSV DNA fragments allowed us to make structural and transcriptional analyses that could never be done before. The identification of viral
proteins by the hybrid arrest method was also simplified using the highly purified HSV DNA fragments.
page 409

1979 Lynn Enquist, colleagues cloning viral DNA fragments into phage vector (herpes simplex virus)
The protein p53 was discovered in 1979 by Lionel
Crawford, David Lane, Lloyd Old, and their co-workers.
Over the next ten years, the gene for p53 was found to act
as a tumor suppressor gene. Its role in the cell is to prevent
cell transformation leading to cancer. About 60 % of
human cancers contain mutations in the p53 gene. It now
appears that the function of the p53 protein is to respond
to damage to DNA, by either moving the cell into a lethal
apoptotic program or permitting cellular repair processes
to act prior to duplication of the DNA and cell division. In
this way, the p53 protein, sensing DNA damage, minimizes
mistakes in the genetic information and eliminates
the rise of genetically altered cells that lead to cancer.
Lane and Levine and their colleagues used antibodies
specifically reactive to SV40 large T antigen to show that
immunoprecipitation from transformed cells resulted in
the recovery of not only SV40 large T antigen itself but also
a cellular protein having an approximate molecular weight
of 53 kDa. Based on its size, this protein became known
as p53. Although this finding was significant in providing
the first evidence that the products of DNA tumor virus
oncogenes function through physical interactions with
cellular proteins, a decade of additional research was
required before the enormous effect of the p53 discovery
would be fully comprehended. The central importance of
p53 is further underscored by the fact that, in addition to
SV40 large T antigen, oncoproteins encoded by other DNA
tumor viruses, such as human papillomaviruses and human
adenoviruses, have similarly evolved to bind and inactivate
p53 in cells.
David P. Lane Arnold J. Levine Lane DP, Crawford LV. T antigen is bound to a host protein
in SV40-transformed cells. Nature 1979;278:261-263.
Linzer DI, Levine AJ. Characterization of a 54K dalton
cellular SV40 tumor antigen present in SV40-transformed
cells and uninfected embryonal carcinoma cells. Cell
1979;17:43-52.
DeLeo AB, Jay G, Appella E, Dubois GC, Law LW, Old LJ.
Detection of a transformation-related antigen in chemically
induced sarcomas and other transformed cells of the
p53 tetramer mouse. Proc Natl Acad Sci USA. 1979;76:2420-2424.
bound to Javier RT, Butel JS. The history of tumor virology. Cancer
the major Res. 2008;68:7693-7706.
groove of
DNA page 410

1979 David Lane, Arnold Levine, others discovery of p53 tumor suppressor protein in SV40-transformed cells
George R. Stark
The western blotting method originated in the laboratory of George Stark at Stanford University. It is used to detect specific proteins in a given sample, say a cell or tissue
homogenate or purified virus extract. It uses gel electrophoresis to separate native or denatured proteins by their length and their conformation. The proteins are then
transferred to a membrane (typically nitrocellulose, nylon or polyvinylidene fluoride - PVDF), where they are detected using antibodies specific for the target protein. The
detection step was at first autoradiography using radiolabeled secondary antibody, but today the most common methods involve use of (1) a horseradish peroxidase-linked
secondary antibody to cleave a bound chemiluminescent reagent, with the luminescent reaction product detected with a CCD camera (the image may be analyzed by
densitometry to estimate the amount of target protein present newer software allows further data analysis such as molecular weight analysis); or (2) a fluorescently labeled
secondary antibody, with the reaction product detected by a CCD camera. Fluorescence is considered to be among the most sensitive detection methods for blotting analysis.
The name western blot was given to the technique by W. Neal Burnette and is a play on the name Southern blot, a technique for DNA detection developed earlier by Edwin
Southern. Detection of RNA by similar means is termed northern blotting.
Renart J, Reiser J, Stark GR. Transfer of proteins from gels to diazobenzyloxymethyl-paper and detection with antisera: a method for studying antibody specificity and antigen
structure. Proc Natl Acad Sci USA. 1979;76: 3116-3120.
Towbin H, Staehelin T, Gordon J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc Natl Acad Sci
USA. 1979;76:4350-4354.
Burnette WN. Western blotting: electrophoretic transfer of proteins from sodium dodecyl sulfate-polyacrylamide gels to unmodified nitrocellulose and radiographic detection
with antibody and radioiodinated protein A. Anal Biochem 1981;112:195-203. (Citation Classic)
page 411

1979 George Stark, others western blotting: transfer of electrophoresed proteins from gel and detection with antibody
Robert A. Swanson (1947-1999) & Herbert W. Boyer Sculpture at Genentechs South San Francisco headquarters
allegorically depicting the first meeting of Robert Swanson
and Herbert Boyer at Churchills Bar in 1976

In 1976, Robert Swanson and Herbert Boyer created the biotechnology industry over a couple of beers at Churchills Bar in San Francisco. Swanson, then just 29, was a venture
capitalist who saw the promise in recombinant DNA technology. Boyer, a 40-year-old biochemistry and biophysics professor at the University of California San Francisco,
had co-developed the technology with Stanley Cohen. After being put off by Cohen, Swanson cold-called Boyer, stopped by his lab, and the two retreated to the bar to sketch
out a business plan. To turn recombinant DNA technology into a business, Boyer and Swanson incorporated under the name Genentech, Inc. Putting down $500 apiece, they
capitalized their new business, to seek practical uses for Boyer and Cohens engineered protein technology. Swanson raised money for staff and labs, and in a two-year, round-
the-clock effort, Genentech staff successfully created their first product: human insulin, which won Food & Drug Administration approval in 1982. In 1980, Genentech went
public and was listed on the New York Stock Exchange its stock leapt from $35 a share to a high of $88 after less than an hour on the market. The offering raised a spectacular
$38.5 million and made its founders multi-millionaires The event was one of the largest stock run-ups ever. Today, Genentech, which has more than 11,000 employees, is a
wholly-owned subsidiary of Roche it was purchased in 2009 for approximately $46.8 billion.
page 412

1980 Robert Swanson, Herbert Boyer Genentech, the first biotech company listed on the NY Stock Exchange
Adult T-cell leukemia (ATL) was identified as a distinct clinical entity by Kiyoshi
Takatsuki, Junji Yodoi and Takashi Uchiyama and their colleagues in Japan in
1976; this was based on the unique geographic distribution of the disease as
well as its distinctive clinical, immunological and histopathological features.
Isao Miyoshi established cell lines from peripheral lymphoid cells of ATL
patients, which exhibited extreme chromosomal abnormalities. In 1980, its
causative agent, human T-lymphotropic virus type I (HTLV-I) was identified
by Bernard Poiesz, Robert Gallo and their colleagues in a cell line derived from
a patient with cutaneous T cell lymphoma they used reverse transcriptase
assays and electron microscopy to prove the presence of the virus. In addition,
the close linkage between ATL and HTLV-I was demonstrated in 1981 by Yorio
Hinuma and his colleagues, by the presence of antibody against viral antigens
in the sera of patients with ATL. Thus, HTLV-I (now HTLV1) was the first
retrovirus shown to be associated with human disease. Today, HTLV1 is known
to infect 10 to 20 million individuals worldwide, and the virus is known to be
associated not only with ATL, but also with an inflammatory disease termed
HTLV-associated myelopathy (HAM) or tropical spastic paraparesis (TSP) as
Bernard J. Poiesz Robert C. Gallo well as other neurologic diseases. In 1981, David Golde described an unusual
immortalized T-lymphocyte cell line grown from spleen tissue of a patient with
hairy-cell leukemia. Gallo suggested that a new retrovirus could be present,
but because the cell line was producing a valuable lymphokine and had been
patented, access was denied. Later, Golde decided to collaborate anyway and
HTLV2 was isolated.
Takatsuki K, Uchiyama T, Sagawa K, Yodoi J. Adult T cell leukemia in Japan. In:
Seno S, Takaku F, Irino S, editors. Topics in hematology. Amsterdam: Excerpta
Medica; 1977. p. 73-77.
Poiesz BJ, Ruscetti FW, Gazdar AF, Bunn PA, Minna JD, Gallo RC. Detection
and isolation of type C retrovirus particles from fresh and cultured lymphocytes
of a patient with cutaneous T-cell lymphoma. Proc Natl Acad Sci USA. 1980;77:
7415-7419.
Hinuma Y, Nagata K, Hanaoka M, Nakai M, Matsumoto T, Kinoshita KI,
Shirakawa S, Miyoshi I. Adult T-cell leukemia: antigen in an ATL cell line and
detection of antibodies to the antigen in human sera. Proc Natl Acad Sci USA.
1981;78:6476-6480.
Mitsuaki Yoshida David Golde (1940-2004) Yoshida M, Miyoshi I, Hinuma Y. Isolation and characterization of retrovirus
from cell lines of human adult T-cell leukemia and its implication in the disease.
Proc Natl Acad Sci USA. 1982;79:2031-2035.
Kalyanaraman VS, Sarngadharan MG, Robert-Guroff M, Miyoshi I, Golde D,
Gallo RC. A new subtype of human T-cell leukemia virus (HTLV-II) associated
with a T-cell variant of hairy cell leukemia. Science. 1982;218:571573.

page 413

1980 Bernard Poiesz, Robert Gallo, Mitsuaki Yoshida, David Golde, others Human T-lymphotropic viruses 1 & 2
The National Center for Infectious
Diseases was created at CDC in 1980
by Walter Dowdle, who served as its
founding director. For the first time, it
brought together all resources needed to
support the mission of infectious disease
prevention and control: lab sciences,
epidemiology, field programs, etc. It was
very successful, but was terminated in
2004 as the CDC director reorganized,
forming smaller, easier to manage units.
The Board of Scientific Counselors, National Center for Infectious
Diseases, Centers for Disease Control and Prevention (1987)
Front row L to R): Joseph Jones (chairman, The Robert Woodruff Foundation), Jeff
Davis (state epidemiologist, Wisconsin Department of Health), J. Mehsen Joseph
(director of laboratories, Maryland Department of Health), Mary Ann Danello
(assistant commissioner, FDA), Walter Bowie (dean, School of Veterinary Medicine,
Tuskegee University), Bernard Fields (chairman, Department of Microbiology and
Molecular Genetics, Harvard Medical School). Back row: Frederick A. Murphy
(director, National Center for Infectious Diseases, CDC), David Fraser (president,
Swarthmore College), John Bennett (director, Clinical Mycology Laboratory, NIAID,
NIH), Lee Hand (director, Division of Infectious Diseases, Emory University School
of Medicine), Rebecca Rimel (executive director, The Pew Charitable Trusts), Scott
Halstead (director, Health Sciences, The Rockefeller Foundation), Carlos Lopez
(infectious disease specialist, Northside Hospital, Atlanta), D. A. Henderson (dean,
Johns Hopkins School of Hygiene and Public Health), Joseph Losos, director
general, Laboratory Centre for Disease Control, Canada), John LaMontagne
(deputy director, NIAID, NIH), Gail Cassell (chair, Department of Microbiology,
University of Alabama, Birmingham). Not present for this photo: Glenn Close
(actor, and president, Trillium Production Company), Stanley Falkow (professor,
Stanford University School of Medicine), Harlyn Halvorson (president, Marine
Biological Laboratory, Woods Hole), George Hill (associate dean for research and
graduate studies, Meharry Medical College), John Maupin (executive vice president,
Morehouse School of Medicine), Phillip Russell (commander, U.S. Army Medical
Research and Development Command), Laurence Foster (state epidemiologist,
Oregon Department of Health).
James M. Hughes Walter R. Dowdle Frederick A. Murphy
The only three directors of the NCID / CDC
page 414

1980 Walter Dowdle, Frederick Murphy James Hughes National Center for Infectious Diseases at CDC
Mule deer brain, immunohistochemical
Elizabeth S. Williams (1951-2004) Stuart Young (1925-2003) stain for PrP protein
Chronic wasting disease (CWD) was first identified as a clinical syndrome of unknown etiology
in mule deer (Odocoileus hemionus) in 1967 in the wildlife facility of Colorado State University.
Biologists conducting natural history and nutritional studies on captive deer observed clinical signs
(progressive weight loss, behavioral changes, listlessness, lowering of the head, repetitive walking in
set pattern, death) and thought these were associated with stresses of captivity, nutritional deficiencies,
or intoxication. Only after 13 years of substantial disease incidence were the veterinary pathologists
Elizabeth Williams and Stuart Young invited to seek a pathological diagnosis. They immediately
identified the disease as a spongiform encephalopathy, that is a prion disease. Shortly thereafter, CWD
was recognized among captive mule deer and in Rocky Mountain elk (Cervus elaphus) in a facility in
Wyoming. The first CWD case in a free-ranging cervid was found in 1981 in a Rocky Mountain elk in
Rocky Mountain National Park, Colorado. The first affected mule deer diagnosed in a wild population
was found in 1984 very near the deer pens where the disease was first observed in 1967. It has since
also been recognized in several other species in zoos and wild animal parks: greater kudu (Tragelaphus
strepsiceros), Arabian oryx (Oryx leucoryx), white-tailed deer (Odocoileus virginianus), black-tailed deer
Williams ES, Young S. Chronic wasting disease of captive mule deer:
(Odocoileus hemionus columbianus), et al. CWD in the game farm industry has become a significant
a spongiform encephalopathy. J Wildl Dis. 1980;16:89-98.
problem in several regions of Canada and the United States (map) and control (test and slaughter)
programs are in place in many affected areas, so far without any real sense of success. Williams ES, Young S. Spongiform encephalopathy of Rocky
page 415 Mountain elk. J Wildl Dis. 1982;18:465-471.

1980 Elizabeth Williams, Stuart Young discovery that chronic wasting disease of deer and elk is a prion disease
In 1967, Carl Woese, Francis Crick, and Leslie Orgel
suggested that RNA could act as a catalyst or enzyme.
This idea was based upon the discovery that RNA can
form complex secondary structures. The first ribozymes
were discovered in the 1980s by Thomas Cech and his
colleagues (especially Benjamin Stark) at the University of
Colorado, who were studying RNA splicing in the ciliated
protozoan Tetrahymena thermophila, and Sidney Altman
and his colleagues (especially Ryszard Kole), working at Yale
University, who were working on a bacterial RNase complex.
Cechs ribozyme was found when trying to purify an enzyme
responsible for a very complex self-splicing reaction they
found that an intron was spliced out during the maturation
of the enzyme in the absence of any added cell extract. As
much as they tried, Cech and his colleagues could not identify
any protein associated with the splicing reaction. Eventually,
they found that the intron portion of the immature RNA was
excised by a complex process self-accomplished by the RNA.
Altmans ribozyme, located in the RNA component of the
RNase P complex, was found to be responsible for conversion
of a precursor tRNA into an active tRNA. They found that
the enzyme that facilitated this was RNase-P and that it
contained RNA in addition to protein and that the RNA was
an essential component of the active enzyme in fact, the
RNA subunit could catalyze the cleavage of precursor tRNA
into active tRNA in the absence of any protein component.
The discovery of an enzymatic role of RNA came as a
complete surprise, falling outside the central dogma of
DNA>RNA>protein, in which RNAs were thought to only Sidney Altman Thomas R. Cech
act in transcription and translation. Among other things, the
discovery reinvigorated the hypothesis of the RNA World, Stark BC, Kole R, Bowman EJ, Altman S. Ribonuclease
that is that the earliest forms of life may have relied solely on P: an enzyme with an essential RNA component. Proc
RNA to store genetic information and to catalyze chemical Natl Acad Sci USA. 1978;75:3717-3721.
reactions. After Cechs and Altmans discovery, many other
Kole R, Altman S. Properties of purified ribonuclease
examples of self-cleaving RNA or catalytic RNA molecules
Extreme oversimplification of the action of P from Escherichia coli. Biochem. 1981;20:1902-1906.
were found today, about 100 RNA ribozymes are known. It
is now possible to make ribozymes that will specifically cleave a ribozyme cleaving an RNA molecule at a Kruger K, Grabowski PJ, Zaug AJ, Sands J, Gottschling
RNA molecules of interest. For example, a ribozyme has been specific sequence site DE, Cech TR. Self-splicing RNA: autoexcision and
designed to cleave the RNA of HIV. If such a ribozyme was autocyclization of the ribosomal RNA intervening
made by a cell, all incoming virus particles would have their sequence of Tetrahymena. Cell 1982;31:147-157.
RNA genome cleaved by the ribozyme, which would prevent Cech TR. RNA splicing: three themes with variations.
infection this idea is being actively pursued in regard to Cell 1983;34:713-716.
other viral pathogens as well.

page 416

1980 Thomas Cech, Sidney Altman, colleagues discovery of the catalytic properties of RNA, the ribozyme concept
James Curran Donald Francis Harold Jaffe Kevin De Cock

Paul Volberding Helene Gayle Jay Levy


These physician-epidemiologists, and many others, did the first outbreak
investigations and natural history studies on the disease that emerged in 1981,
eventually called acquired immunodeficiency syndrome (AIDS)
page 417

1981 MMWR reports of Pneumocystis carinii pneumonia in Los Angeles and Kaposi sarcoma in New York AIDS
Vincent Racaniello David Baltimore Eckard Wimmer
In 1981, in two separate sets of experiments conducted in two laboratories, a poliovirus
infectious clone was constructed and the complete genomic RNA sequence was determined
both were firsts for a virus of vertebrates. The infectious clone was made by Vincent Racaniello
and David Baltimore at MIT and the sequencing was done by the same individuals and
independently by Eckard Wimmers team at the State University of New York, Stony Brook
the team included Naomi Kitamura, Bert Semler, Paul Rothberg, Glenn Larsen, Cheryl Adler,
Andrew Dorner, Emilio Emini, Ronnie Hanecak, James Lee, Sylvie van der Werf and Carl
Anderson.
Racaniello Racaniello V, Baltimore D. Cloned poliovirus complementary DNA is infectious in mammalian
and cells. Science 1981;214:916-919.
Baltimores
poliovirus Kitamura N, Semler BL, Rothberg PG, Larsen GR, Adler CJ, Dorner AJ, Emini EA, Hanecak R,
cloning Lee JJ, van der Werf S, Anderson CW, Wimmer E. Primary structure, gene organization and
strategy polypeptide expression of poliovirus RNA. Nature 1981;291:547-553.
Racaniello VR, Baltimore D. Molecular cloning of poliovirus cDNA and determination of the
Structural model of poliovirus
complete nucleotide sequence of the viral genome. Proc Natl Acad Sci USA. 1981;78:4887-4891.
page 418

1981 Vincent Racaniello, David Baltimore, Eckard Wimmer infectious clone and complete sequence of poliovirus
Gerd Binnig and Heinrich Rohrer
The scanning tunneling microscope (STM) is an instrument for
imaging surfaces at the atomic level. Its development in 1981
earned its inventors, Gerd Binnig and Heinrich Rohrer, the
Nobel Prize in Physics in 1986. For an STM, good resolution
is considered to be 0.1nm lateral resolution and 0.01nm depth
resolution. With this resolution, individual atoms within materials
are routinely imaged and manipulated. A literature search of
papers published over the past 10 years reveals that most uses of
STM have been in materials sciences, with only a few innovative
approaches having been applied to biological materials: for Scanning tunneling electron microscope
example, quite a few STM images of various bacteriophages and
tobacco mosaic virus have been published. Further, some viral
proteins have been imaged, such as the reverse transcriptases of
human immunodeficiency virus (HIV1) and of Moloney murine Binnig G, Rohrer H, Gerber Ch,
leukemia virus. However, even though very high resolution has Weibel E. Surface studies by
been achieved, it has been difficult to interpret images obtained scanning tunneling microscopy.
by STM relative to other high resolution methods such as negative Phys Rev Lett. 1982;49:57-61.
contrast transmission electron microscopy and cryoelectron
microscopy and even harder to interpret findings relative to X-ray Scanning tunneling
crystallography data. Seemingly, more work is needed. microscopy image of
page 419 tobacco mosaic virus

1981 Gerd Binnig, Heinrich Rohrer development of the scanning tunneling electron microscope
Don C. Wiley (1944-2001) John J. Skehel Ian A. Wilson
In 1981, Ian Wilson, Don Wiley and John Skehel
published the atomic structure of the influenza
hemagglutinin to a resolution of 3 this was the
first depiction of the structure of a viral peplomer
protein. Two years later, the structure of influenza virus
neuraminidase was solved to a resolution of 2.9 by
Joseph Varghese, Graeme Laver and Peter Coleman.
Wilson IA, Skehel JJ, Wiley DA. Structure of the
haemagglutinin membrane glycoprotein of influenza virus
at 3 resolution. Nature 1981;289:366-373.

Various depictions of the ectodomain of


the influenza virus hemagglutinin trimer
page 420

1981 Don Wiley, John Skehel, Ian Wilson, others visualization of the structure of the influenza virus hemagglutinin
The beginning of research on the ras (RAt Sarcoma) oncogene family can be
traced back to 1964 when Jennifer Harvey observed that a preparation of the
rat leukemia virus that now bears her name induced sarcomas in new-born
rodents. Werner Kirsten found the same thing with a similar virus preparation
that now bears his name. Three additional viruses were later shown to carry
ras oncogenes. By the late 1970s, it was known that retroviral oncogenes could
rapidly transform cells, and that specific retroviruses had acquired these genes
from the genomes of the mammalian and avian cells that they had infected.
It was therefore proposed that mutations in the cellular homologues of these
genes could transform cells in the absence of any viral involvement, and that
this occurred in a substantial proportion of human cancers. In 1982, Robert
Weinberg, Michael Wigler and Mariano Barbacid each independently cloned
ras, the first oncogene, from bladder carcinoma cell lines. This cloned cellular
gene (c-ras) had the same transforming properties as the oncogene from the
viruses (v-ras). It was found that the oncogenes in question were the cellular
homologues of ras genes from Harvey and Kirsten rat sarcoma viruses. In 1982,
Weinberg, Wigler and Barbacid discovered that a single amino-acid change
in the Ras protein glycine to valine at position 12 was responsible for the
oncogenic effect. Subsequent research has shown that this change alters the
structure of the Ras protein to make it constitutively active. The family of
normal Ras proteins are involved in cellular signal transduction, and mutations
in the gene that encoded them in turn causes cell growth, differentiation,
dysregulated cell division and long-term cell survival. ras oncogene signaling
can lead to oncogenesis and cancer. Activating mutations in ras are found in 20-
25% of all human tumors and up to 90% in certain specific tumor types.
Shih C, Weinberg RA. Isolation of a transforming sequence from a human
bladder carcinoma cell line. Cell 1982;29:161-169.
Goldfarb M, Shimizu K, Perucho M, Wigler M. Isolation and preliminary
characterization of a human transforming gene from T24 bladder carcinoma
cells. Nature 1982;296:404-409.
Robert A. Weinberg and Michael H. Wigler
Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nature
Rev|Cancer 2003;3:7-13.
Mutations in
ras signaling
pathways
associated
with human
diseases

Ribbon model of
H-Ras protein
structure
page 421

1981 Robert Weinberg, Michael Wigler discovery and cloning of the first human tumor-derived oncogene, h-ras
Fred Murphy Milt Zaitlin Dorothy Horstman
Council Council (1911-2001) Council
Bill Joklik
President

David Bishop
Secretary-Treasurer

Max Summers Bob Haselkorn Priscilla Schaeffer


Council Council (1943-2010) Council
The Founding Officers
and Council Members
1981
Harry Ginsberg
(1917-2003)
Vice President

page 422

1981 Bill Joklik, Harry Ginsberg, others founding of the American Society for Virology
Founding of the American Society for Virology
[abridged from: Joklik WK, Grossberg SE. How the American Society for Virology was founded. Virology 2006;344:250-257.]:
1966: Ninth International Congress of Microbiology (ICM), Moscow dissatisfaction with virology venues at ICMs led to planning for a virology congress.

1968: First International Congress for Virology, Helsinki a grand success.

<1980: Virology was one of the Divisions of the American Society for Microbiology (ASM) dissatisfaction with virology venues at ASM meetings.

1980: Ad hoc discussions were held as to whether to found a free-standing American Society for Virology. Requests to the ASM by its Virology Division for travel funds to
the Fifth International Congress of Virology in Strasbourg in 1981 were denied.

1981: Bill Joklik sent an organizational planning letter to Peter Vogt, Purnell Choppin, Bob Wagner, David Baltimore, Tom Merigan, Juli Youngner, Norton Zinder, Harry
Ginsberg, Al Kaplan, Lee McLaren, Fred Murphy, Fred Rapp and Walter Schlesinger, laying out general planning ideas. The reaction to this letter was
overwhelmingly positive.

1981: A public meeting of 40 virologists was held on 9 June 1981 in Chicago at which a resolution was passed overwhelmingly to found a virology society 9 June 1981 is
the birthday of the American Society for Virology.

1981: An Organizing Committee was constituted both to design an administrative structure and to organized the first annual meeting of the society in August 1982 at
Cornell University. More than 650 virologists were invited to become charter members and attend the Ithaca meeting. The members of the interim Organizing
Committee were: David Baltimore, Purnell Choppin, Harry Ginsberg, Bob Haselkorn, Dorothy Horstman, Bill Joklik, Tom Merigan, Fred Murphy, Bernard
Roizman, Max Summers, Peter Vogt, Bob Wagner, Julius Youngner, Milt Zaitlin and Norton Zinder.

1982: The first annual meeting of the American Society for Virology was held at Cornell University, locally organized by Milt Zaitlin, and attended by more than 500
virologists (with the society already having about 1,000 paid-up members). The scientific program, organized by the interim president Bill Joklik, consisted of four
symposia and 27 workshops: the topics of the symposia were: Genome Structure and Expression (Norton Zinder, David Baltimore, Jan Kaper); Transformation and
Persistence (Mike Bishop, George Miller, Mark Ptashne); Mechanisms of Infection (Abner Notkins, Bill Robinson, Tom Monath); and Epidemiology and Ecology
(Peter Palese, Max Summers, Karl Johnson).

At the first business meeting of the Society the following were elected:
President Bill Joklik
President-Elect Harry Ginsberg
Councilors Fred Murphy, Milton Zaitlin, Dorothy Horstman, Max Summers, Bob Haselkorn, Priscilla Schaeffer
Secretary-Treasurer David Bishop

At the first business meeting the following committee appointments were made:
Meetings & Program Committee Ken McIntosh, Chairman, Skip Carmichael, Bob Goodman, Bill Joklik, Jonathan King, Bill Rawls, Max Summers
Membership Review Committee Al Wood, Chairman, Brian Derbyshire, Harry Ginsberg, Michael Gottesman, Nancy Hopkins, Tom Merigan, Andy
Nahmias, Helen Revel, Hugh Robertson, Hans Storz, Milt Zaitlin
Charter and Bylaws Committee Bernard Fields, Chairman, Bob Haselkorn, Dorothy Horstman, Jack Morris
Finance Committee George Miller, Chairman, David Bishop, Fred Murphy, Fred Rapp, Ed Scolnick

>1982: By 2010, the Society had 3,543 members, 614 of whom were from 44 countries outside the USA, including 183 from Canada. Successful annual meetings have been
held every summer since 1982.
page 423

1981 Bill Joklik, Harry Ginsberg, others founding of the American Society for Virology
The development of the first recombinant vaccine for humans, hepatitis B surface antigen (HBsAg) expressed
in yeast, began with the discovery by Baruch Blumberg, Barbara Werner, Manfred Bayer, and Lawrence Loeb of
particles smaller than whole virions in HBsAg-positive blood. Experiments showed that these particles could
induce protective immunity. Blumberg and Irving Millman proposed that a vaccine could be made from HBsAg
particles obtained from the blood of hepatitis B carriers. A patent for this concept was filed in 1969. Maurice
Hilleman and colleagues at Merck recognized the importance of the concept and by 1971 they obtained a license to
develop a hepatitis B vaccine made from HBsAg purified from blood. In 1980, Wolf Szmuness and others showed
that the vaccine provided more than 90 percent protection against hepatitis B and had no adverse effects. In 1981,
the serum-derived vaccine was made available for general use. Production of the hepatitis B subunit vaccine in large
quantities was hampered by the need for the blood of hepatitis B carriers and the realization that such blood could
be contaminated with other viruses. In 1977, William Rutter, Pablo Valenzuela and their colleagues at the University
of California San Francisco obtained material containing hepatitis B virus from Merck. After cloning the hepatitis
B virus and obtaining the genetic sequence of HBsAg, Rutter and his colleagues explored a variety of different
biological systems in which to produce the particles using recombinant techniques. They were unsuccessful using
bacteria. Then, in 1980 and 1981, Rutter collaborated with Benjamin Hall and colleagues, of the University of
Washington, who had developed a model system using yeast cells. Rutter and Hall successfully produced pure
HBsAg particles from recombinant yeast cells. Rutter and colleagues then founded Chiron Corporation, in part
to develop the HBsAg vaccine through a contractual relationship with Merck and also to develop other medical
therapies using recombinant techniques. At Merck, Hilleman used the recombinant yeast-derived HBsAg to make
an improved version of a hepatitis B vaccine. This vaccine was the first recombinant vaccine licensed for use in
humans by the FDA this occurred in 1986. There is another side to this story, not covered here: there have been
many precedent-setting law suits involving the recombinant yeast technology and its ownership, and beyond this,
legal questions over the patenting of products, processes and concepts, each of which has fundamentally changed
the entire biotechnology industry.

William Rutter (center) and his former students,


Edward E. Penhoet of the University of California
Berkeley (left) and Pablo Valenzuela of the
University of California San Francisco (right).
They developed the first recombinant vaccine
for humans, hepatitis B surface antigen (HBsAg),
expressed in yeast. They then founded Chiron
Corporation, now part of Novartis International AG.

Edman JC, Hallewell RA, Valenzuela P, Goodman HM, Rutter WJ. Synthesis of
hepatitis B surface and core antigens in E. coli. Nature 1981;291:503-506.
Valenzuela P, Medina A, Rutter WJ, Ammerer G, Hall BD. Synthesis and assembly
of hepatitis B virus surface antigen particles in yeast. Nature 1982;298:347-350. HBsAg as vaccine substrate, produced in yeast
(Saccharomyces cerevisiae)
Hilleman MR. Yeast recombinant hepatitis B vaccine. Infection 1987;15:3-7. negative contrast electron microscopy page 424

1981 William Rutter, Pablo Valenzuela, colleagues first recombinant human vaccine, Hepatitis B (HBsAg in yeast)
The term prion (pronounced pree-on) was
coined in 1982 by Stanley Prusiner; it is derived
from protein and infection. Tikvah Alper and her
colleagues in London had earlier suggested that
the scrapie agent of sheep, which had been studied
for many years, might lack nucleic acid, which
usually can be degraded by ultraviolet or ionizing
radiation. When extracts of scrapie-infected brains
were irradiated, the extracts retained their ability to
transmit the disease. Prusiner and his colleagues at
the University of California San Francisco purified
the scrapie infectious agent and showed that it
consisted solely of a specific protein prion protein
(PrP, a 35kD membrane glycoprotein). Normal prion Bovine spongiform encephalopathy, bovine
protein (PrPC) consists primarily of alpha helices brain, vacuoles in neurons, progressing to
where the amino acid backbone twists into a specific florid spongiform degeneration and neuronal
degeneration. H&E.
kind of spiral; the abnormal form (PrPSc, for scrapie),
however, contains beta sheets where the amino acid
backbone is fully extended. The prion diseases of
humans include: several types of Creutzfeldt-Jakob
disease (CJD); kuru, a now-extinct disease of New
Guinea natives; and several familial neurologic
diseases. The most common animal prion diseases
are: scrapie, an important disease of sheep; bovine
spongiform encephalopathy (BSE); chronic wasting
disease (CWD) of deer and elk; and others that
follow cross-species transmission from scrapie or
BSE. All of the prion diseases are invariably fatal,
always with a very long incubation period, always
with characteristic brain lesions (vacuoles in Mode of production of PrPC in neurons and the
accumulation of PrPSc as scrapie-associated
neurons, progressing to spongiform degeneration fibrils and plaques
usually with associated with amyloid plaques. The
most amazing thing about the prion diseases is that
the infectious agents, the prions, are propagated
Stanley B. Prusiner and transmitted solely by misfolded proteins
when the abnormal form of prion proteins (PrPSc)
Prusiner SB. Novel proteinaceous infectious particles cause scrapie. enter a healthy animal they induce a refolding of
Science 1982;216:136-144. normal prion proteins (PrPC), which in turn act as a
Committee on Transmissible Spongiform Encephalopathies. template causing the misfolding of more and more
Advancing prion science: guidance for the national prion research normal protein in a chain reaction. The abnormal
program. Washington: National Academies Press; 2003. prion proteins are extremely stable and resistant
to normal proteolysis. They accumulate in neural
Colby DW, Prusiner SB. Prions. Cold Spring Harb Perspect Biol. tissue and cause cellular damage and death by as yet
2011;3:a6833. unknown means. Molecular models of normal prion protein
page 425
(PrPC) and the abnormal prion protein (PrPSc)

1982 Stanley Prusiner concept of the prion the etiologic agents of the spongiform encephalopathies
Goad WB. GenBank - and
its promise for molecular
genetics. Los Alamos
Science 1983;9(Fall):52-61.

GenBank Overview: http://


www.ncbi.nlm.nih.gov/
genbank/

Walter Goad (1925-2000)

In 1979, 30 molecular biologists and computer scientists meeting at the Rockefeller University agreed on the necessity to create a national, computerized database. The impetus
was the explosive growth in the number of known DNA sequences and the promise of producing biological knowledge by analyzing and comparing them a database seemed
indispensable. It took almost three years for the NIH to come up with a funding scheme; Walter Goad and his colleagues were awarded a $2 million, 5-year grant in 1982 and
began building the Los Alamos National Laboratory (LANL) Sequence Library, which was re-christened GenBank, the Nucleic Acid Sequence Data Bank. Goad was a physicist
who had spent 1970-1971 with Francis Crick at the MRC Laboratory of Molecular Biology at Cambridge University; Crick lent strong support to the idea of an international
data resource. From Goads paper in Los Alamos Science in 1983: It was clear that just accumulating the data in a computer was not particularly interesting. What was
interesting were questions about organizing, managing, and analyzing the data. The first job we faced was collecting the sequence data. This was straightforward although
physically very demanding. We worked very hard just to get caught up with the data that was in existence when we started. Our estimate of what needed to be added was
based on scanning some journals and looking through lists of titles. Unfortunately, the estimate was low because the titles of many articles with sequences in them didnt reveal
much. Most of the sequences were on the order of a thousand bases long, but the longest the entire genome of the lambda bacteriophage contains about 50,000 bases.
My outlook was that mysticism about life was being crowded out by the greater joy of knowledge thanks to molecular genetics and molecular biology in general. There is,
after all an immense difference between speculating about the way things might work and knowing how they do work. Today, GenBank, which is managed by NIH and its
National Library of Medicine, in collaboration with the DNA Data Bank of Japan and the EMBL Nucleotide Sequence Database in the United Kingdom, supports some 40,000
users searches per day, accessing its 22 billion item library for many purposes, not the least of which concerns virology. page 426

1982 Walter Goad, others development of GenBank


Stunting of growth (a) and death (b) of newborn mice infected with LCM
virus (strain Armstrong), and reversal of stunting by transplantation of cells
secreting growth hormone. (b) Without hormone replacement the mice die.
From Michael Oldstone and colleagues paper in Nature, 1984.
Oldstone MBA, Sinha YN, Blount P, Tishon A, Rodriguez M, von Wedel R, Lampert PW. Virus-induced alterations in
homeostasis: Alterations in differentiated functions of infected cells in vivo. Science 1982;218:1125-1127.
Rodriguez M, von Wedel RJ, Garrett RS, Lampert PW, Oldstone MBA. Pituitary dwarfism in mice persistently
infected with lymphocytic choriomeningitis virus. Lab Invest. 1983;49:48-53.
Oldstone MBA, Rodriguez M, Daughaday WH, Lampert PW. Viral perturbation of endocrine function: Disorder of
Michael B.A. Oldstone cell function leading to disturbed homeostasis and disease. Nature 1984;307:278-280.
Oldstone MBA. Viral alteration of cell function. Scientific Amer. 1989;260:42-48.
Early in his career, Michael Oldstone and his colleagues showed that, contrary to established dogma, in persistent virus infections the host was not tolerant but made specific
antiviral immune responses usually ineffective in clearing infection but nevertheless affecting its course, outcome and pathogenesis. This immune response often caused
tissue damage and disease, the phenomenon of immunopathology. Discoveries in this area were made originally with lymphocytic choriomeningitis (LCM) virus and murine
retroviruses, but later were extended to measles virus and other infections including several in humans. Buildings on this work, they showed that certain antibodies to some
viruses recognized similar amino acid sequences or motifs found in host/cell proteins this cross-reactivity, referred to as molecular mimicry, has been shown to be important
in several viral diseases. They also explored the details of how viruses cause immunosuppression and abrogate normal host immunologic surveillance, again determining
the pathogenetic role of this phenomenon in viral persistence and chronic disease. Another mechanism discovered by Oldstone and his colleagues by which persistent virus
infection can produce disease involves infection effects upon the differentiation or functions of cells, thereby altering physiological homeostasis and normal functioning of key
organs and tissues. This was termed interference with cellular luxury functions, but many of the functions found to be affected are hardly luxuries. For example, they showed
that persistent noncytopathic LCM virus infection of the anterior lobe of the murine pituitary gland affects the production of growth hormone, causing growth retardation,
hypoglycemia and death. This finding of a noncytopathic virus causing endocrine dysfunction has led to exploration of the possible viral etiology of other endocrine,
neurotransmitter, cytokine and inflammatory diseases this subject, as applied to human diseases, especially neurologic diseases, is an important work-in-progress.
page 427

1982 Michael Oldstone, colleagues concept of virus damage to cellular homeostasis and luxury functions
Gerald N. Woode (1934-2013) Marian C. Horzinek Bovine torovirus
negative contrast electron microscopy
In 1979, Gerald Woode and his colleagues visualized an unusual virus in feces from a calf with acute enteritis its morphology was distinct from that of known viruses. The
calf was from a farm near Breda, Iowa (hence the name Breda virus now, bovine torovirus), where >50% of calves developed diarrhea during the first 20 days of life. In
following years, similar viruses were identified in calves from other states. After Woodes initial report, morphological and antigenic similarities of Breda virus and an equine
virus from Berne, Switzerland (Berne virus, now, equine torovirus), were noted. The latter virus was first described by Marianne Weiss, Franz Steck and Marian Horzinek in
1983. Because of their similarities, the two viruses were grouped together as the toroviruses, in a provisional family, the Toroviridae. The term torovirus refers to the unique
kidney-shape of the viral nucleocapsid and often the virion itself torus is Latin for the doughnut-shaped molding that some columns have in their bases. Today, based also on
common genomic organization and replication strategy, the toroviruses form a genus in the family Coronaviridae. The genus now includes toroviruses of other animals, and
in 1984 torovirus-like particles were detected in feces from children with gastroenteritis by Graham Beards and his colleagues. The human virus(es) have since been found in
many countries.
Woode GN, Reed DE, Runnels PL, Herrig MA, Hill TH. Studies with an unclassified virus isolated from diarrheic calves. Vet Microbiol. 1982;7:221-240.
Weiss M, Steck F, Horzinek MC. Purification and partial characterization of a new enveloped RNA virus (Berne virus). J Gen Virol. 1983;64:1849-1858.
Horzinek MC, Weiss M. Toroviridae: a taxonomic proposal. Zentralbl Veterinarmed B. 1984;31:649-659.
Beards GM, Green J, Hall C, Flewett TH, Lamouliatte F, du Pasquier P. An enveloped virus in stools of children and adults with gastroenteritis that resembles the Breda virus
of calves. Lancet 1984;1:1050-1052. page 428

1982> Gerald Woode, Marian Horzinek, Graham Beards, others discovery of bovine, equine, human toroviruses
Hepatitis E virus, negative contrast electron microscopy

The self-sacrifice of Russian virologist, Mikhail Balayan, bordered on the extreme. In 1983, Balayan was
investigating an outbreak of non-A, non-B hepatitis in a central Asian part of the Soviet Union. Though
he wanted to bring samples back to his Moscow laboratory (in the Institute of Poliomyelitis and Viral
Encephalitides, Russian Academy of Medical Sciences), he lacked refrigeration. So, he made a shake of
yogurt and an infected patients stool, drank it, went back to Moscow, and waited. When he became
Mikhail Surenovich Balayan seriously ill a few weeks later, he started collecting and analyzing his own stool samples. In these he found
a new virus that produced liver injury in laboratory animals and could be seen by electron microscopy.
It looked like hepatitis A virus, but he showed that it was not, because he already had antibodies against
hepatitis A virus and these did not react with the new virus. Subsequently, in 1990, Gregory Reyes and his
colleagues at Genelabs Technologies, Inc. cloned and sequenced the genome of the virus in collaboration
with Balayan and Daniel Bradley and his colleagues at the Centers for Disease Control and Prevention.
The virus was named hepatitis E virus. It was found in succeeding years to be the cause of major epidemics
of severe hepatitis with high mortality in pregnant women. Further, it was found to be zoonotic with
reservoirs in swine and other animals.
Balayan S, Andjaparidze AG, Savinskaya SS, Ketiladze ES, Braginsky DM, Savinov AP, Poleschuk
VF. Evidence for a virus in non-A, non-B hepatitis transmitted via the fecal-oral route. lntervirology
1983;20:23-31.

page 429

1983 Mikhail Balayan discovery of hepatitis E virus


Francoise Barr-Sinoussi Luc Montagnier Jean-Claude Chermann
Much has been written about the discovery of human immunodeficiency virus 1 (HIV1); the full story cannot be repeated here, but the discovery of the virus itself at the
Institut Pasteur and how this was made public can be. After AIDS was first clinically recognized in Los Angeles and New York, several research groups sought its etiology, most
without success. Clinicians in Paris asked for etiologic research to be done and in 1982 Jean-Claude Chermann and Franoise Barr-Sinoussi working with Luc Montagnier
and other colleagues responded. Tissues from patients were cultured and then searched for viruses, including retroviruses [using a reverse transcriptase (RT) assay]. One RT
assay turned positive after three weeks of culture, but this was associated with the death of the cells, not characteristic of the retroviruses of the day. The virus was named
lymphadenopathy-associated virus (LAV); it was studied by electron microscopy and seen to be a lentivirus (now a genus in the family Retroviridae). At first it was not known
if the virus was the cause of AIDS; nevertheless, in 1983 its description was published in Science. For some time it was said that the team led by Montagnier had discovered
the virus, but the team led by Robert Gallo of NIH had proven that the virus was the cause of AIDS. Later, it came out that a highly selected set of human sera from AIDS
patients together with controls, put together by Donald Francis and his colleagues at the U.S. Centers for Disease Control and Prevention (CDC), had been tested in 1983 by
Montagnier and his colleagues and found to strongly support an etiologic association between the virus and the disease. Sets of sera from lymphadenopathy patients in France
reacted similarly. Early in 1984, Chermann was invited to speak at a retrovirus conference in Park City, Utah Chermann presented serologic data based on antigen produced
from LAV virus-infected human leukocytes. After a few minutes, the room went silent. For the first time it was realized that the virus had been found. Afterward, the audience
cheered and also welcomed the news that a blood-test would soon be available. Two day later, Chermann presented the same data at CDC in Atlanta the largest auditorium
was jammed, and again silence was followed by cheers. The name HIV1 was chosen in 1986 by third parties to end the dispute between the French teams name, LAV, and
American teams name, HTLV3. Patent rights disputes and discovery primacy issues went on far into the 1990s.
Barr-Sinoussi F, Chermann J-C, Rey F, Nugeyre MT, Chamaret S, Gruest J, Dauguet C, Axler-Blin C, Brun-Vezinet F, Rouzioux C, Rozenbaum W, Montagnier L. Isolation of a
T-lymphotropic retrovirus from a patient at risk for acquired immunodeficiency syndrome (AIDS). Science 1983;220:868-871.
Brun-Vezinet F, Barr-Sinoussi F, Saimot AG, Christol D, Montagnier L, Rouzioux C, Klatzmann D, Rozenbaum W, Gluckman JC, Chermann J-C. Detection of IgG antibodies
to lymphadenopathy-associated virus in patients with AIDS or lymphadenopathy syndrome. Lancet 1984;1:1253-1256. page 430

1983 Francoise Barr-Sinoussi, Luc Montagnier, Jean-Claude Chermann human immunodeficiency virus 1 (HIV1)
In 1955, the first chemical synthesis of DNA was
accomplished by A.M. Michelson and Alexander Todd.
Subsequently, this contribution was recognized by a Nobel
Prize to Todd. Next, Har Gobind Khorana and his colleagues
showed how a DNA sequence could be assembled via
chemical means, now known as the phosphodiester method.
In 1976, Khorana with his 19 co-workers reported on the
synthesis of a 126-residue long DNA. This project took
8 years; today the same product can be made in an hour
using an automated DNA synthesizer. In the mid-1970s,
the first solid-phase synthesis of DNA was performed in
the laboratories of Hubert Kster, Michael Gait and Keiichi
Itakura. Solid-phase synthesis is the dominant method used
today. The specific chemistry used today, employing amidite
chemistry, was developed in 1981 by Marvin Caruthers and
Mark Matteucci. DNA strands are formed by the sequential
addition of activated nucleotides to an immobilized starter
DNA molecule. The product remains attached to an
insoluble support until the final release step. This solid-
phase approach was key to the development of automated
oligonucleotide synthesis machines. In 1981, two engineers
from Hewlett Packard, Andre Marion and Sam Eletr,
commenced operations of Applied Biosystems Inc. (ABI);
their first instrument was a peptide synthesizer, but by 1983
their second instrument, the Model 380A DNA Synthesizer,
Marvin Carruthers Andre F. Marion and Sam H. Eletr entered production and was an instant success. It was a
crucial factor in the advance of technologies requiring
DNA probes and primers, the latter especially for use in
polymerase chain reaction (PCR) technology, which in turn
was crucial for large DNA sequencing projects. By 1986, ABI
marketed its 394 DNA Synthesizer, which could make four
different oligonucleotides simultaneously. Advances were
made continuously, such that today instruments, such as
the ABI 3948 with 96 parallel reaction capillaries, can run
continuously in automated fashion yielding the very large
numbers of oligonucleotides that are needed to support
huge genome sequencing programs, such as the Human
Genome Project. Several other companies make competing
machines.
Matteucci MD, Caruthers MH. Studies on nucleotide
chemistry IV. Synthesis of deoxyoligonucleotides on a
polymer support. J Amer Chem Soc. 1981;103; 3185-3191.
ABI 380A, 1983 ABI 394 (4-columns), 1986 ABI 3948, 1998
Caruthers MH. Gene synthesis machines: the DNA
Applied Biosystems, Inc., Oligonucleotide (DNA) Synthesizers chemistry and its uses. Science 1985;230:281-285.
page 431

1983 Marvin Carruthers, others, Applied Biosystems, Inc. development of commercial DNA synthesis technology
Andrew W. Murray Jack W. Szostack Construction of a yeast artificial chromosome (YAC)

Yeast (Saccharomyces cerevisiae) artificial chromosomes (YACs) are vectors used to clone DNA fragments larger than 100 kb, up
to a maximum size of 1Mb to 3Mb. YACs are useful for the physical mapping of complex genomes and for the cloning of large
genes. First described in 1983 by Andrew Murray and Jack Szostak, YACs are artificially constructed so that they are rather like
chromosomes they contain (1) telomeres, located at each chromosome end to protect the DNA from degradation by nucleases;
(2) a centromere, the attachment site for mitotic spindle fibers, required to duplicate the chromosome when yeast cells divide;
(3) replication origin sequences needed for chromosome replication in yeast cells; and (4) other specific sequences such as
selectable markers that allow the easy isolation of yeast cells that have taken up the artificial chromosome and recognition sites
for restriction enzymes. YACs are built using an initial circular plasmid, which is typically broken into two linear molecules using
restriction enzymes; DNA ligase is then used to ligate a sequence or gene of interest between the two linear molecules, forming
a single large linear piece of DNA. In some instances, this DNA is re-circularized. YACs and other yeast plasmid vectors have
another advantage in that they can be used to express eukaryotic proteins that require posttranslational modification.
Murray AW, Szostak JW. Construction of artificial chromosomes in yeast. Nature 1983;305:189-193.
The Nobel Prize in Physiology or Medicine of 2009 was awarded to Elizabeth H. Blackburn, Carol W. Greider and Jack W.
Szostak for the discovery of how chromosomes are protected by telomeres and the enzyme telomerase.
page 432

1983 Andrew Murray, Jack Szostack yeast artificial chromosomes (YACs) vectors to clone large DNA fragments
Some Vaccinia Virus Recombinant Vectored Virus Vaccine Candidates
Pathogen Experimental Animal Antigen
Species Protected
Rabies virus Raccoons, foxes, dogs G, N
Measles virus Mice, rats, dogs HA, F, N
Hepatitis B virus Chimpanzees HBsAg
Herpes simplex virus Mice, guinea pigs GB, GD
Murine cytomegalovirus Mice NS pp89
Epstein-Barr virus Cottontop tamarins G 340
Human influenza viruses Hamsters, ferrets, mice HA, NA
Dengue viruses Mice PreM, E, NS1, NS2
Yellow fever virus Mice PreM, E, NS1, NS2ab
Japanese encephalitis virus Mice, swine PreM, E, NS1
Respiratory syncytial virus Cotton rats, chimpanzees M2 ,G
Human papillomaviruses Mice, rats E6, E7 oncoproteins
Human parainfluenza viruses Monkeys HA, F, N
Lassa virus Guniea pigs G, N
Vesicular stomatitis viruses Mice, cattle G, N
Rinderpest virus Rabbits, cattle HA, F
Peste d petits ruminants virus Goats HA, F
Equine herpesviruses Hamsters G13, G14
Pesudorabies virus Mice, swine G50, GII, GIII
Equine influenza viruses Horses HA
Bovine leukemia virus Sheep E, G51
Bovine papillomavirus Rats Early proteins
Classical swine fever virus Swine G55, G33
Bernard Moss Venezuelan encephalitis virus Mice, monkey, horses E1, E2, N
Sendai virus Mice HA, F, N, M
Avian Influenza viruses Chickens HA
Friend leukemia virus Mice Gag
HIV 1 and 2 Macaques, chimpanzees G, gag, pol, protease Basic scheme for vaccinia vectored vaccines
Despite having first been developed in 1983 by Bernard Moss of NIH, Enzo Paoletti of the New York State Department of Health and their colleagues, there are no viral
vectored vaccines currently on the market for use in humans. However, several are in various stages of development and approval for use. One impediment has been the
conservatism of the regulatory authorities in several countries, especially the FDA in the U.S. Recombinant vaccinia viruses, that is vaccinia vectored vaccine viruses, have been
shown to protect animals against diseases of veterinary importance, including vesicular stomatitis virus and rinderpest in cattle, pseudorabies virus in swine, and influenza
viruses in chickens. A recombinant vaccinia virus expressing rabies virus glycoprotein has been successfully administered in bait form as a wildlife vaccine in both the United
States and Europe. Other poxviruses have also been tested as vectors for veterinary applications. Examples include a raccoon poxvirus vector to protect raccoons against rabies
virus; a capripoxvirus vector for cattle against rinderpest virus; a swinepox virus vector for pigs against pseudorabies virus; a fowlpox vector for chickens against influenza
viruses, Newcastle disease virus, and infectious bursal disease virus; and canarypox virus for cats against feline leukemia virus.
Smith GL, Mackett M, Moss B. Infectious vaccinia virus recombinants that express hepatitis B virus surface antigen. Nature 1983;302:490-495.
Paoletti E, Lipinskas BR, Samsonoff C, Mercer C, Panicali D. Construction of live vaccines using genetically engineered poxviruses: biological activity of vaccinia virus
recombinants expressing the hepatitis B virus surface antigen and the herpes simplex virus glycoprotein D. Proc Natl Acad Sci USA. 1984;81:193-197.
Quinnan GV Jr. Vaccinia viruses as vectors for vaccine antigens. Proceedings of a workshop, 1984, Chevy Chase, Maryland. New York: Elsevier North Holland; 1985.
Draper SJ and Heeney JL. Viruses as vaccine vectors for infectious diseases and cancer. Nature Reviews Microbiology 2010;8:62-73.
page 433

1983 Bernard Moss, Enzo Paoletti, others recombinant vaccinia virus as vector for viral vaccines (hepatitis B virus)
The concept underlying the polymerase chain reaction (PCR), which allows
the nearly limitless amplification of specific DNA sequences, was described
by Kjell Kleppe and Har Gobind Khorana in 1971. The major improvements
made by Kary Mullis allowed PCR to become a (perhaps the) central technique
in molecular biology, such that it was described by The New York Times as
virtually dividing biology into two epochs, before PCR and after. In 1983,
Mullis was working for the Cetus Corporation when he had the idea to use a
pair of primers to bracket a desired DNA sequence and to copy the sequence
over and over using DNA polymerase Mullis spent more than a year before
making his idea work, first in late 1983. Other Cetus scientists (Randall Saiki
and Henry Erlich) worked on developing AIDS and other tests utilizing PCR
Saiki was first author on the first paper (1983), while Mullis took longer to finish
the paper that described PCR itself (1984). A major shortcoming of the first PCR
was that the DNA polymerase was destroyed by the high heat used and had to
be replaced in each cycle. In 1986, Mullis started to use Thermophilus aquaticus
DNA polymerase (Taq polymerase) the enzyme from this bacterium, which
lives happily in Yellowstone hot springs, is heat resistant and needs to be
added only once, thus making the technique more affordable and amenable to
automation. In 1991, Cetus was sold to Chiron Corporation, its value largely
based upon its patent rights to PCR. PCR patent rights were then sold for $300
million to Hoffman-La Roche and its subsidiary, Roche Molecular Systems.
Saiki RK, Scharf S, Faloona F, Mullis KB, Horn GT, Erlich HA, Arnheim N.
Kary B. Mullis and Sydney Brenner Enzymatic amplification of beta-globin genomic sequences and restriction site
at the Nobel Prize Celebration, Stockholm, 1993 analysis for diagnosis of sickle cell anemia. Science. 1985;20:230:1350-1354.
Mullis K, Faloona F, Scharf S, Saiki R, Horn G, Erlich H. Specific enzymatic
amplification of DNA in vitro: the polymerase chain reaction. Cold Spring Harb
Symp Quant Biol. 1986;51 Pt 1:263-273.

Kary
Mullis
first
thermocycler
Scheme for the first three of usually 20-30 PCR cycles
page 434

1985 Kary Mullis, colleagues, Cetus, Perkin-Elmer invention of the polymerase chain reaction (PCR)
Julio I. Maiztegui (1931-1993) Julio G. Barrera Oro George French
Candid #1 vaccine for Argentine hemorrhagic fever was developed
in 1985 by Julio Barrera Oro and Julio Maiztegui, in collaboration
with Gerald Eddy and George French of USAMRIID. The vaccine
was shown to be very effective in preventing disease in high risk farm
workers in the endemic region of Argentina. The vaccine was first
manufactured by the Salk Institute in the U.S. and became available in
Argentina in 1990. In 2006, the Instituto Nacional de Enfermedades
Virales Humanas Dr. Julio I. Maiztegui started production of the
vaccine in Argentina, with a goal of producing the 5 million doses to
vaccinate the entire population of the endemic area.
Sculpture, Instituto
Nacional de
Enfermedades Junin Virus Vaccine Development Team
Virales Humanas L-R: Francisco Pinheiro, C.J. Peters,
Julio Maiztegui, Gerald Eddy, Neal Halsey, Nick Tauraso,
Pergamino, Argentina Julio Barrera Oro, George French, Julio
page 435 Maiztegui, Alexis Shelokov, Kelly McKee

1985 Julio Maiztegui, Julio Barrera Oro, George French development of Argentine hemorrhagic fever vaccine
Abstracted from various reviews of Fields Virology over the years:
...Fields Virology was conceived by Bernard Fields, a distinguished
Harvard virologist of the 1980s and 1990s, as a means of bringing
fundamental and medical aspects of virology together in a more
integrated manner than previously available in contemporary
textbooks of virology. The project fulfilled the need and,
notwithstanding the early death of Bernard Fields, has progressed to
become one of the classic resources of virology.
Samuel Johnsons Dictionary defines Bible as the sacred volume
in which are contained the revelations of God. A current dictionary
has provided me with an additional definition: any book, reference
work, periodical, etc. accepted as authoritative, informative, or
reliable. When our virology graduate students were asked about their
use of Fields Virology, they said it was their virology Bible.
The Editors, many of them previous co-editors of Bernard Fields,
have taken particular care in entrusting the individual chapters to
internationally recognized experts in their fields; many of them have
contributed to previous editions, others have been recently recruited.
This is the most comprehensive book in virology, tying together David M. Knipe
detailed accounts of the molecular biology, molecular pathology and
medical features of individual viruses in the broad context of general
virology. Not in vain is this opus regarded as the bible of virology
apart from all its other values, it is great fun to study.

Editors-in-Chief over the years: Bernard N. Fields,


David M. Knipe, Peter M. Howley
Associate Editors over the years: Robert M. Chanock, Joseph
L. Melnick, Bernard Roizman, Robert E. Shope, Thomas P.
Bernard Nathan Fields (1938-1995) Monath, Stephen E. Strauss, Diane E. Griffin, Robert A. Lamb,
Malcolm A. Martin

Peter M. Howley
Fields Virology:
1st ed. 1985
2nd ed. 1990
3rd ed. 1996
4th ed. 2001
5th ed. 2007 page 436

1985 Bernard Fields, others publication of Fields Virology, now five editions
Acquired immunodeficiency syndrome (AIDS) first
appeared in 1981 and was first seen in hemophiliacs
and transfusion recipients in 1982. Subsequent studies
established the transmissibility of the causative agent
(HIV) by various routes, including blood transfusions,
untreated coagulation factor concentrates and
blood components needed by hemophiliacs. Once
transmissibility was established, some preventive
measures were put into place, although for some years
important aspects of the natural history of HIV infection
remained unknown. The first preventive measures
were to protect the blood supply. In 1983, a detailed
blood donor history questionnaire with deferral of
high-risk blood donors was implemented. Antibody
testing for HIV1 began in 1985, first for use in blood
banks, then for clinical diagnostics and epidemiologic
studies. The three pioneering firms that developed the
first commercially available enzyme immunoassays
(EIAs) were Abbott Laboratories, bioMerieux, Inc.
and Bio-Rad Genetic Systems. At first the antigen
used in these EIAs was a crude cell culture extract; in
recent years, synthetic peptides derived from highly
conserved, immunodominant regions of the HIV env
(envelope) and pol (polymerase) gene products have
been used. For clinical purposes, usually, one screening
test (often run more than once if the first test is positive
or indeterminate) have been employed, followed by a
confirmatory test where needed. EIA tests have been the
most widely used for screening, with the confirmatory
test now usually utilizing a western blot assay. The EIA
HIV tests have undergone many iterations that have
improved their sensitivity, specificity and reproducibility.
Additional methodologies have reduced the diagnostic
window, that is the period after infection but before
the appearance of detectable antibodies. PCR testing
for viral RNA or proviral DNA is now widely used
to confirm EIA tests its use has also narrowed the
diagnostic window considerably. Modern HIV serologic
and nucleic acid testing is extremely accurate. The
chance of a false-positive result in multi-step testing
protocols is estimated to be 0.0004% to 0.0007% in the
general U.S. population.

page 437

1985 bioMrieux, Bio-Rad Genetic Systems, Abbott development of first FDA approved HIV antibody tests (EIAs)
Norman L. Letvin (1949-2012) Ronald C. Desrosiers Phylogeny of the many SIVs,
each associated with a particular nonhuman primate species

Shortly after AIDS was recognized in humans a similar syndrome was observed in macaques housed at the New England and California National Primate Research Centers
(NENPRC and CNPRC). Although initially called simian AIDS, the syndrome was found in most instances to be caused by a type D retrovirus (now called simian retrovirus
1 (SRV1), related to Mason Pfizer monkey virus. Only after human AIDS emerged were transmission studies done; it was in these studies, done in 1983, that the first simian
immunodeficiency virus (SIV) was discovered at the NENPRC; this was the macaque SIV virus (SIVmac). Experimentally, this virus caused an AIDS-like disease that was fatal
in a rather short time (6 months to 2 years). SIVmac was shown to be closely related to HIV1 and virtually identical to HIV2 [which was even more closely related to the SIV
isolated from sooty mangabeys (SIVsmm)]. Many diverse SIV variants were then isolated from many species of seropositive, captive and wild African nonhuman primates,
including chimpanzees (SIVcpz, the variant most closely related to HIV1). Unlike HIV1 and HIV2 infections in humans, most SIV infections in their natural hosts appear to be
non-pathogenic, despite high levels of circulating virus the exceptions are important: SIVsmm infection in rhesus macaques causes simian AIDS (SAIDS) and SIVcpz causes
in chimpanzees an AIDS-like disease similar to HIV1 infection in humans. Indeed, further genomic/phylogenetic studies strongly suggest that HIV1 first infected humans via
zoonotic transmission of a SIVcpz-like virus and HIV2 of a SIVsmm-like virus. The presence of many of the variant SIVs in several of the National Primate Research Centers
was later traced back to animal transport in the 1960s and 1970s.
Daniel MD, Letvin NL, King NW, Kannagi M, Sehgal PK, Hunt RD, Kank PJ, Essex M, Desrosiers RC. Isolation of T-cell tropic HTLV-III-like retrovirus from macaques.
Science 1985;228:1201-1203.
Letvin NL, Daniel MD, Sehgal PK, Desrosiers RC, Hunt RD, Waldron LM, Mackey JJ, Schmidt DK, Chalifoux LV, King NW. Induction of AIDS-like disease in macaque
monkeys with T-cell tropic retrovirus STLV-III. Science 1985;230:71-73.
Gardner MB. Simian AIDS: an historical perspective. J Med Primatol. 2003; 32:180-186.
page 438

1985 Norman Letvin, Ronald Desrosiers, colleagues discovery of simian immunodeficiency viruses (SIVs)
Francis Barin Max Essex Phyllis Jean Kanki Ronald Hunt
Although HIV1 and HIV2 are closely related, they maintain distinct epidemiologic and biologic
characteristics. HIV2 is largely confined to West Africa, while HIV1 infection is prevalent worldwide
and accounts for approximately 95% of all HIV infections globally. Importantly, disease progression to
AIDS occurs much more slowly in HIV2 in one way, HIV2 infection appears like an attenuated HIV
infection. The discovery of HIV2 in Senegal in the mid-1980s prompted numerous studies to determine
its geographic distribution and biologic significance. Through the use of type-specific serologic assays
it was found that HIV2 is present in most West African countries, but infection with HIV1 is more
prevalent than infection with HIV2, ranging from a 3- to 24-fold ratio (HIV1 versus HIV2). In fact, from
more recent serosurvey data it seems that HIV2 prevalence rates are diminishing and that HIV1 may
competitively displace HIV2 in the long term. Outside West Africa, sporadic cases of HIV2 infection
have been found, mostly related to human movement, with particular foci stemming from former
Portuguese colonies. It is now recognized that HIV2 and the SIV variant (SIVsmm) from the sooty
mangabey are so closely related that the human virus must have originated from zoonotic transmission
from the nonhuman primates of a SIVsmm-like virus to humans. It has been estimated that HIV1 and
HIV2/SIV viruses may have diverged from each other as recently as 50-60 years ago.
Barin F, MBoup S, Denis F, Kanki P, Allan JS, Lee TH, Essex M. Serological evidence for virus related to
simian T-lymphotropic retrovirus III in residents of west Africa. Lancet 1985;2:1387-1389.
Clavel F, Gutard D, Brun-Vzinet F, Chamaret S, Rey MA, Santos-Ferreira MO, Laurent AG, Dauguet
C, Katlama C, Rouzioux C, Klatzmann D, Champalimaud JL, Montagnier L. Isolation of a new human
retrovirus from West African patients with AIDS. Science 1986;233:343-346.
page 439
Phylogeny of HIV2 and related viruses

1985 Francis Barin, Phyllis Kanki, Max Essex, colleagues discovery of human immunodeficiency virus 2 (HIV2)
HSV genomic DNAs digested with
SalI and electrophoresed in
Alec J. Jeffreys Bernard Roizman agarose gel

DNA fingerprinting has had more than one definition seemingly, the first was the method of analyzing isolates of herpes simplex virus (HSV), by digestion of their DNA
with restriction endonucleases and visualization of fragments by gel electrophoresis, which originated in the laboratory of Bernard Roizman at the University of Chicago.
In 1978, he and his colleagues discriminated between HSV isolates in clusters of severe hospital infections, showing in one instance that there were two independent
introductions of HSV1 into a pediatric intensive care unit resulting in two clusters of epidemiologically related, but virologically independent chains of infection. The method
was subsequently used for distinguishing variants of other large DNA viruses and double-stranded RNA viruses (e.g., rotaviruses); the methodology was employed widely until
being replaced by various partial and complete viral genome sequencing methods. However, DNA fingerprinting is otherwise associated with Alec Jeffreys and his discovery in
1984 of a means of distinguishing the DNA of individuals. He had a eureka moment after looking at the X-ray film image of isotopically-labeled restriction enzyme fragments
of the DNA of different members of his technicians family. He realized that DNA fingerprinting could be used in forensics to identify individuals and also to resolve paternity
and immigration disputes. DNA fingerprinting was first used to identify the rapist and killer of two teenagers in 1983 and 1986. In 1985, DNA profiling, based on typing
individual highly variable minisatellites [also known as short tandem repeats (STRs)] in the human genome, was also developed by Jeffreys and his team by focusing on just
a few of these highly variable minisatellites, DNA profiling became sensitive, reproducible, and amenable to computer databasing; it has become the standard forensic DNA
system used worldwide. Jeffreys and his team also were also among the first to describe restriction fragment length polymorphism (RFLP) technology and the use of single
nucleotide polymorphisms (SNPs), which have had great value in viral pathogenesis research.
Linnemann CC, Light IJ, Buchman TG, Ballard JL, Roizman B. Transmission of herpes-simplex virus type 1 in a nursery for the newborn identification of viral isolates by DNA
fingerprinting. Lancet 1978;1:964-966.
Halperin SA, Hendley JO, Nosal C, B. DNA fingerprinting in investigation of apparent nosocomial acquisition of neonatal herpes simplex. J Pediatr. 1980;97:91-93.
Jeffreys AJ, Wilson V and Thein SL. Hypervariable minisatellite regions in human DNA. Nature 1985;314: 67-73.
Jeffreys AJ, Wilson V, Thein SL. Individual-specific fingerprints of human DNA. Nature 1985 316:76-79.
page 440

1985 Alec Jeffreys development of DNA fingerprinting (and detection of single nucleotide polymorphisms SNPs)
The first person to recognize that the sheep disease, scrapie, may have infected cows and caused bovine
spongiform encephalopathy (BSE) was Carol Richardson, a pathologist at the MAFF (Ministry of Agriculture,
Fisheries & Food, now DEFRA, the Department for Environment, Food and Rural Affairs) Central Veterinary
Laboratory (CVL) in the UK. In December 1984, a dairy cow was identified that had been behaving strangely,
showing tremors, and not responding to treatment. Soon other cows were identified with the same clinical signs;
tissues from the 10th cow were submitted to the CVL, where in September 1985 brain specimens were examined
by Richardson. She observed vacuolation in the neuriopil, similar to those she had seen many times before in
sheep affected with scrapie. Her colleagues agreed that this looked like a case of bovine scrapie, something
never seen before. But, the significance of the moment was lost it was only when more cases were referred to
the laboratory that it was realized that this was something new and significant. The first official MAFF record
of BSE in the UK was a memo from June 1987, although a confidential memo on the subject signed by the head
of pathology department of the CVL circulated in MAFF in December 1986. It took until October 1987 before
Richardsons colleagues published the first description of the clinical syndrome and pathology of BSE in The
Veterinary Record. The epidemic surged even as control activities were initiated, reaching a peak in 1993. In a
2004 study, Roy Anderson and his epidemiology research group, first at Oxford University and then at Imperial
College London, estimated that 1.9 million cattle had been infected, 1.6 million of which had entered the food
chain. The overall cost of the epidemic to the British economy has been estimated at more than 4 billion.
Wells GA, Scott AC, Johnson CT, Gunning RF, Hancock RD, Jeffrey M, Dawson M, Bradley R. A novel
progressive spongiform encephalopathy in cattle. Vet Rec. 1987;121:419-420.
Carol Richardson
Left: MAFF Official data, BSE incidence,
UK, 1986 2009 (monthly plot)
(total of 181,123 confirmed cases on 36,191
farms, as of 2011) MAFF has always only
counted cases confirmed by histologic /
immuno-histochemical methods.
Right: The Economist, Aug 1996
[from: Anderson RM, Donnelly CA,
Ferguson NM, Woolhouse ME, et al.
Nature 1996;29;382:779-788.], showing that
1.9M cattle had been infected, not 181,000.

Carol Richardsons pathology case card, MAFF Central MAFF, Official first record of BSE in the UK, MAFF, confidential memo of BSE in the UK,
Veterinary Laboratory, Weybridge, 19 September 1985 17 June 1987 [but, November 1986 is the date given by the 19 December 1986, six months before public
page 441 UK government when BSE was first identified in UK] announcement: advise keeping an open mind

1985 Carol Richardson discovery of bovine spongiform encephalopathy in the United Kingdom
Saul Kits thymidine kinase deletion mutant pseudorabies vaccine
The UL23 gene-encodes thymidine kinase, which phosphorylates
deoxythymidine into deoxythymidine-triphosphate. This enzyme is not
essential for virus growth in cell culture but is important for the replication
of the virus in vivo in differentiated cells

Herpesvirus infection, brain,


thin section electron microscopy
Saul Kit (1928-2008)
photo courtesy of Baylor College of Medicine Archives
Abstracted from the New Scientist and the New York Times, 1986: The first field test of a genetically altered virus vaccine took place in 1985 in the U.S. without the knowledge
of the public or a review by scientists appointed to approve such experiments. The Animal and Plant Health Inspection Service (APHIS), a division of the U.S. Department of
Agriculture, was accused of sidetracking the federal regulatory system in order to get a genetically altered virus vaccine onto the market quickly. When the permit for the first
release was suspended the controversy threatened to paralyze industrial biotechnology. The vaccine (Omnivac; TechAmerica, Omaha, Nebraska), for pseudorabies in swine,
was constructed by Saul Kit of Baylor College of Medicine as a thymidine kinase deletion mutant. The mutant virus replicates well in cell culture, but not in differentiated cells
in vivo. APHIS allowed the company to test the vaccine (which previously had only been tested in laboratory animals) in three states, but many scientists within the agency
were not aware of this, nor that the vaccine was approved for sale. The departments Agricultural Recombinant-DNA Research Committee, which was set up in 1976 to assess
proposals for biotechnology in agriculture, did not review the proposal prior to its release. The APHIS chief veterinarian stated that the proposal was mentioned informally
to the expert panel and there was no concern raised. He said that because only a single gene had been deleted, the virus was not considered worthy of special treatment. He
also argued that vaccination does not constitute release into the environment. Clearly, regulatory approval of novel vaccines, for animal or human use, has come a long way
since this episode. Even then, activist groups opposed to genetic engineering immediately filed law suits. In the end, years later, the vaccine was re-licensed.
Kit S, Kit M, Pirtle EC. Attenuated properties of thymidine kinase negative deletion mutants of pseudorabies virus. Am J Vet Res. 1985;46:1359-1367.
page 442

1985 Saul Kit, colleagues development of the first recombinant live-virus vaccine (pseudorabies)
The Development of Modern Viral Vaccinology Based Upon Innovative Technologies
Date People Virus Technology
Attenuated Live-Virus Vaccines
1789 E Jenner Variola virus Vaccines produced from naturally occurring attenuated viruses
1935 M Theiler Yellow fever virus Vaccines produced by serial passage of viruses in heterologous host animal or embryonating eggs
1959 A Sabin, others Polioviruses Vaccines produced by serial passage of viruses in cultured cells and animals
1970s H Maassab, others Influenza viruses Vaccines produced by selection of cold-adapted mutants and reassortant viruses
Non-Replicating Native Antigen Vaccines
1885 L Pasteur Rabies virus Vaccines produced from inactivated whole virions
1980s G Laver, R Webster, others Influenza viruses Vaccines produced from purified native viral proteins
1981 M Hilleman Hepatitis B virus Vaccines produced from native viral subunits
Vaccines Produced by Recombinant-DNA and Other Innovative Technologies
1981 W Rutter, P Valenzuela, Hepatitis B virus Vaccines produced by recombinant-DNA technology, expression in eukaryotic (yeast, mammalian, insect, plant) cells
M Hilleman, others
1982 B Moss, E Paoletti Vaccinia virus Vaccines produced by recombinant-DNA technology, by the expression of viral antigens in viral vectors
(e.g., poxvirus vectors, rhabdovirus vectors, adenovirus vectors, picornavirus vectors, herpesvirus vectors)
1985 S Kit Pseudorabies virus Vaccines produced by recombinant-DNA technology, by the deletion of a gene(s) associated with pathogenicity
1990s D Markowitz, I Verma, Several viruses Vaccines produced by recombinant-DNA technology, by the expression of viral antigens in defective viral vectors
A Stewart, M Hitt, others (e.g., adenovirus vectors, AAV vectors, alphavirus vectors, flavivirus vectors, herpesvirus vectors)
1990s A Charbit, M Hofnung, Several viruses Vaccines produced by recombinant-DNA technology, by the expression of viral antigens in bacterial vectors
R Curtiss, D OCallaghan
1990s R Kennedy, H Kunkel, others Hepatitis B virus Vaccines produced by recombinant-DNA technology, by anti-idiotypic antibodies
1993 J Ulmer, others Influenza viruses Vaccines produced by recombinant-DNA technology, by the injection of viral DNA or plasmid DNA or replicons
containing viral gene(s)
2002 I Frazer, others Human Vaccines produced by recombinant-DNA technology, by viral proteins self-assembling in to virus-like particles (e.g.,
papillomaviruses HBV, HPV, parvovirus B19, BTV) or pseudoparticles with/without heterologous epitopes
2002 T Monath, others Yellow fever virus, Vaccines produced by recombinant-DNA technology, by the formation of viral chimeras (viruses with the replicative
West Nile virus machinery of one virus and the protective antigens of another)
This is not a place for coverage of the huge subject of modern, innovative vaccinology and the diverse methodologies involved in research, development and commercial
production of new vaccines, especially vaccines for those viral diseases where conventional vaccines have not succeeded (HIV/AIDS, hepatitis C virus, herpes simplex
virus, respiratory syncytial virus, Coxsackie A viruses, et al.). By the 1960s, there was a decline in the sort of innovation that had led to many successful vaccines, but after
the introduction of hepatitis B virus vaccine in the 1980s (recombinant HBsAg produced first in yeast and then in various hosts), a change for the better was seen, and now
at least in the areas of basic science and preclinical development, there is much ongoing innovation. It remains to be seen if the regulatory aspects and commercial (profit-
making) aspects of vaccinology will follow. Optimism stems from ongoing scientific, technological and manufacturing advances, most but not all grounded in molecular
and cell biology, molecular immunology, information technologies and the concept of platform technologies. In this context, the introduction of influenza vaccines made in
cell culture rather than in eggs, seemingly a prosaic change and not reflective of any really innovative technology, is nevertheless an important symbol. The table here might
have been placed in association with several key discoveries, but upon reflection it is the work of William Rutter, Pablo Valenzuela, Maurice Hilleman and their colleagues on
recombinant hepatitis B (HBsAg) vaccine in 1981 and of Saul Kit on thymidine kinase deletion mutant pseudorabies vaccine in 1985 that seem to stand as the benchmarks.
page 443

1981> The development of modern viral vaccinology based upon innovative technologies
Feline immunodeficiency virus (FIV) was first isolated from cats in
Petaluma, California in 1986 by Niels Pedersen, Janet Yamamoto
and their colleagues from the School of Veterinary Medicine at the
University of California Davis. The virus was shown to be a typical
lentivirus it replicates preferentially but not exclusively in feline
T-lymphoblastoid cells, where it causes a characteristic cytopathic
effect. Kittens experimentally infected with FIV manifest a transient
(several days to 2 weeks) fever and neutropenia beginning 4 to
8 weeks after inoculation. This is associated with a generalized
lymphadenopathy that persists for up to 9 months. Most cats
recover from this initial phase of the disease but complete recovery
is rare in nature or in the laboratory setting. The terminal AIDS-
like phase of the illness has been seen mainly in naturally infected
cats. It appears a year or more following the initial infection in an
unknown proportion of infected animals. Similarities between FIV
infection of cats and HIV infection of humans are striking, making
FIV infection of cats a useful model for human AIDS. In spite of the
similarities between FIV and HIV, there is no evidence that FIV is a
health hazard to humans or other species of animals. FIV has been
identified in cats from all parts of the world. It is most prevalent in
high density populations of free roaming cats (feral and pet), with
prevalence rates of 1-12%.
Pedersen NC, Ho EW, Brown ML, Yamamoto JK. Isolation of a
T-lymphotropic virus from domestic cats with an immunodeficiency-
like syndrome. Science. 1987;235:790-793.
Yamamoto JK, Sparger E, Ho EW, Andersen PR, OConnor TP,
Mandell CP, Lowenstine L, Munn R, Pedersen NC. Pathogenesis of
Niels C. Pedersen Janet K. Yamamoto experimentally induced feline immunodeficiency virus infection in
cats. Am J Vet Res. 1988;49:1246-1258.
Pedersen NC, Yamamoto JK, Ishida T, Hansen H. Feline
immunodeficiency virus infection. Vet Immunol Immunopathol.
1989;21:111-129.

A simplified phylogenetic tree


of the lentiviruses, based upon
multiple sources of genomic
data and analysis page 444

1986 Niels Pedersen, Janet Yamamoto, colleagues discovery of feline immunodeficiency virus
Automated DNA-sequencing output from a capillary
electrophoresis machine, where after size separation,
detection and recording of dye fluorescence is
visualized as shown, while the data are sent directly for
computer-based analysis and storage
Leroy E. Hood Lloyd M. Smith
Shortly after dideoxynucleotide chain-terminating DNA sequencing was introduced by Fredrick Sanger, Leroy Hood, Lloyd Smith, Tim and Mike Hunkapiller and their
colleagues, working in a Caltech basement, attempted to develop an automated sequencer it turned out to be a difficult job, taking several years. The difficulty was the
underlying chemistry and the means for detecting reaction products the chemistry for linking dyes to DNA did not exist and neither did a way to measure the different
colored fluorescent dyes (fluors). In a paper published in Nature in 1986, the group announced their prototype automated DNA sequencer, ushering in a new era in genomics
and in virology. The key to their success was fluorescence detection of enzymatically degraded DNA fragments using fluorophores (developed by DuPont), which were
covalently attached to oligonucleotide primers. A different colored fluorophore was used for each of the reactions specific for the four bases A, C, G and T. The reaction
mixtures were combined and co-electrophoresed down a single polyacrylamide gel tube, the separated fluorescent bands of DNA being detected near the bottom of the tube
as the electrophoresis continued and fragments ran out of the gel. The sequence information was fed directly into a computer. After the prototype was complete, Hood sold
the rights to Applied Biosystems, Inc., but he continued to work with the company to refine the original concept and machine. The invention became the engine of the global
effort to sequence the genomes of all prototypic viruses, as well as the engine of the Human Genome Project. Today, the electrophoresis is usually done in capillaries, thereby
allowing massively parallel runs there are also many different newer technologies in use today, supporting even more massive sequencing projects.
Smith LM, Fung S, Hunkapiller MW, Hunkapiller TJ, Hood LE. The synthesis of oligonucleotides containing an aliphatic amino group at the 5 terminus: synthesis of
fluorescent DNA primers for use in DNA sequence analysis. Nucleic Acids Res. 1985;13: 2399-2412.
Smith LM, Sanders JZ, Kaiser RJ, Hughes P, Dodd C, Connell CR, Heiner C, Kent SB, Hood LE. Fluorescence detection in automated DNA sequence analysis. Nature
1986;321:674-679.
page 445

1986 Leroy Hood, Lloyd Smith, Applied Biosystems, Inc. development of automated DNA sequencing
Frank Fenner (1914-2010) Authors of Veterinary Virology, Third Edition
E. Paul J. Gibbs, Marian C. Horzinek, Michael J. Studdert, Frederick A. Murphy
In 1987, Frank Fenner, David White (1931-2004) and their
colleagues published a companion to the book, Medical Virology,
the first editions of which had been published in 1970 and 1976.
The first edition of Veterinary Virology was a success, so a second
edition was published in 1993, a third in 1999 and a fourth in
2010. Each edition followed Fenners unique style, and attempted,
in the words of M. Anthony (Tony) Epstein in his review in
Nature, to mimic Fenners seeming ease with which extremely
complicated topics are reduced to an orderly survey of the
Frank Fenner, Peter A. Bachmann, E. Paul J. Gibbs, Frederick A.
basic facts involved, together with suitable explanations of their
Murphy, Michael J. Studdert, David O. White, First Edition, 1987
significance. Fenners incomparable standard of scholarship, seen
in Veterinary Virology, was grounded in his memorable personal Frank Fenner, E. Paul J. Gibbs, Frederick A. Murphy, Rudolf Rott,
qualities, his warm, generous, unpretentious character, matched Michael J. Studdert, David O. White, Second Edition, 1993
with his great fortitude, insight and understanding of his students. Frederick A. Murphy, E. Paul J. Gibbs, Marian C. Horzinek,
Fenners vision was so broad as to extend from virologys historic Michael J. Studdert, Third Edition, 1999
roots as a microbiological / pathological / clinical infectious N. James Maclachlan, Edward J. Dubovi (Editors); Stephen W.
disease science, and later a molecular and cell biologic science, to Barthold, Richard A. Bowen, Ronald P. Hedrick, Donald P.
its present role as a major contributor to the overall advance of Knowles, Michael D. Lairmore, Colin R. Parrish, Linda J. Saif,
our understanding and stewardship of life on earth. David E. Swayne (Chapter Authors), Fourth Edition, 2010
page 446

1987> Frank Fenner, others publication of book, Veterinary Virology (currently four editions)
AZT - Zidovudine, structural model

Marty St. Clair Samuel Broder


Azidothymidine (AZT) (Zidovudine, INN) (trade-names: Retrovir, Retrovis) was the first drug approved for the treatment of HIV infection /AIDS. It is an analog of thymidine.
Jerome Horwitz of Wayne State University School of Medicine first synthesized AZT in 1964; it was originally intended to treat cancer, but failed to show efficacy. In 1984,
shortly after HIV was shown to be the cause of AIDS, scientists at Burroughs Wellcome Co. (now GlaxoSmithKline) began searching for compounds to treat the disease.
Burroughs Wellcome had expertise in viral diseases, led by Gertrude Elion, George Hitchings David Barry, Phil Furman, Marty St. Clair, Janet Rideout, Sandi Lehman
and others. Their efforts focused on the viral reverse transcriptase; they identified and synthesized compounds and developed a screening test for activity against murine
retroviruses. One compound coded BW A509U showed potent activity against the murine viruses. At the same time, Samuel Broder, Hiroaki Mitsuya, and Robert Yarchoan
at the National Cancer Institute initiated a program to develop drugs active against HIV they soon developed a collaboration with Janet Rideout and others at Burroughs
Wellcome and confirmed that BW A509U had activity against HIV. A few months later, they started a phase 1 clinical trial of the drug, now named AZT, at the NCI and Duke
University. This trial showed that the drug could be safely administered to patients with HIV and that it could increase CD4 counts in AIDS patients. A placebo-controlled
randomized trial was subsequently conducted by Burroughs-Wellcome, in which it was shown that AZT could prolong the life of patients with AIDS. The FDA approved the
drug (via a then-new FDA accelerated approval system) in 1987. The time between the first demonstration that AZT was active against HIV in cell culture and its approval was
25 months, one of the shortest periods for drug development and licensing in history.
Mitsuya H, Weinhold K, Furman P, St Clair M, Lehrman S, Gallo R, Bolognesi D, Barry D, Broder S. 3-Azido-3-deoxythymidine (BW A509U): an antiviral agent that inhibits
the infectivity and cytopathic effect of human T-lymphotropic virus type III/lymphadenopathy-associated virus in vitro. Proc Natl Acad Sci USA. 1985;82: 7096-7100.
Broder S. The development of antiretroviral therapy and its impact on the HIV1/AIDS pandemic. Antiviral research 2009;85: 1-2.
page 447

1987 Marty St. Clair, Samuel Broder, others, Burroughs Wellcome/NCI first anti-HIV drug approved by FDA (AZT)
Mario R. Capecchi Oliver Smithies Martin J. Evans
Mario Capecchi and Oliver Smithies were seeking ways of specifically altering the mammalian genome, Capecchi with a
view to inserting new genes into cells and Smithies in the hope of correcting genetic defects. They independently discovered
that they could use a natural mechanism, discovered decades before by Joshua Lederberg in bacteria, to introduce short
sequences of manipulated DNA into the chromosomes of cultured mammalian cells. Their techniques allowed them to
target individual genes, producing the genetic alterations they sought, but only at the cellular level. The embryonic stem cell
cultures that Martin Evans was then developing provided the necessary vehicle for taking such gene manipulations from cell
culture into whole animals. By modifying genes in embryonic stem cells and then injecting these cells into fertilized mouse
eggs, it became possible to rear mice with discrete inheritable genetic modifications knock-out mice.
Evans MJ, Kaufman MH. Establishment in culture of pluripotential cells from mouse embryos. Nature. 1981;292:154-156.
Doetschman T, Maeda N, Smithies O. Targeted mutation of the hprt gene in mouse embryo stem cells Proc Natl Acad Sci
USA. 1988;85:8583-8587.
Mansour SL, Thomas KR, Capecchi MR. Disruption of the proto-oncogene Int-2 in mouse embryo-derived stem cells a
general strategy for targeting mutations to non-selectable genes. Nature 1988;336:348-352.
Evans MJ. Potential for genetic manipulation of mammals. Mol Biol Med. 1989;6:557-565. Mouse in which a gene affecting hair
growth has been knocked out
Capeccci MR. Altering the genome by gene targeting. Trends Genet. 1989;5:70-76. page 448

1988 Mario Capecchi, Oliver Smithies, Martin Evans knockout and other genetically manipulated mice
Exanthem subitum
(roseola infantum)
in a young child
A virus called human herpesvirus 6 (HHV-6) was first isolated from adult patients
with lymphoproliferative disorders in 1986; however, soon it became evident that
there were two viruses, now named human herpesvirus 6A and 6B. The two viruses
are closely related, but show consistent differences in biological, immunological,
epidemiological and molecular properties. HHV-6B is the major causative agent
of exanthem subitum, but no clear association has been made between any clinical
disease and infection with HHV-6A. Exanthem subitum is also called roseola
infantum and sixth disease (as the sixth rash-causing childhood disease), but these
clinical terms are vague in that they encompass infections caused by other viruses
for example, HHV-7 can also cause the same clinical syndrome. HHV-6A, 6B
and HHV-7 are ubiquitous, with most infections occurring in infancy and with
more than 90% of adults having antibody to the three viruses. Typically, in infants
Koichi Yamanishi Takeshi Kurata infected with HHV-6B there is sudden fever, which lasts for a few days, and a rash
which appears on the trunk and face and spreads to the lower extremities as the
fever subsides. More severe cases present diagnostic and treatment complexities
(thrombocytopenia, granulocytopenia, hepatitis, encephalopathy with seizures).
In adults, primary infections can cause a mononucleosis like disease. Since like
other herpesviruses, HHV-6B establishes latency following primary infection, it
is an important pathogen in immunocompromised hosts. Reactivation occurs in
patients after bone marrow transplantation, solid organ transplantation such as
liver, renal and heart transplantation, and AIDS.
Yamanishi K, Okuno T, Shiraki K, Takahashi M, Kondo T, Asano Y, Kurata T.
Identification of human herpesvirus-6 as a causal agent for exanthem subitum.
Lancet 1986;1:1065-1067.
Lopez C, Pellett P, Stewart J, Goldsmith C, Sanderlin K, Black J, Warfield D,
Feorino P. Characteristics of human herpesvirus-6. J Infect Dis. 1988;157:1271-
1273.
Pellett PE, Lindquester GJ, Feorino P, Lopez C. Genomic heterogeneity of human
herpesvirus 6 isolates. Adv Exp Med Biol. 1990;278:9-18.
Yamanishi K. Human herpesvirus 6. Microbiol Immunol. 1992;36:551-561.
Mori Y, Yamanishi K. HHV-6A, 6B, and 7: pathogenesis, host response, and clinical
disease. In: Arvin A, Campadelli-Fiume G, Mocarski E, Moore PS, Roizman B,
Whitley R, Yamanishi K, editors. Human herpesviruses: Biology, therapy, and
page 449 Philip E. Pellett Carlos Lopez immunoprophylaxis. Cambridge: Cambridge University Press; 2007. Chapter 46.

1988 Koichi Yamanishi, Takeshi Kurata, Philip Pellett, Carlos Lopez human herpesvirus 6B and exanthem subitum
Internal ribosomal initiation sites (IRES) are
nucleotide sequences that allow for translation
initiation in the middle of a messenger RNA
(mRNA) sequence with consequent protein
synthesis. Usually, in eukaryotes, translation
can be initiated only at the 5 end of the mRNA
molecule, since 5 cap recognition is required.
IRES sequences were first discovered in 1988 in
encephalomyocarditis virus RNA and poliovirus
RNA in the labs of Eckard Wimmer and Nahum
Sonenberg, respectively. They were described
as distinct regions in mRNA molecules that
are able to attract the eukaryotic ribosome. It
was hypothesized and later proven that IRES
elements have a distinct secondary or even
tertiary structure within the mRNA, leading to
their correct spatial orientation with ribosomes.
IRES-ribosomal interactions also require the
recognition of viral mRNA by host factors.
Poliovirus IRES elements are the most complex
elements described to date, but they are very
efficient in driving internal translation initiation.
IRES elements have also been described in other
RNA viruses, including flaviviruses, retroviruses,
and several plant viruses. The IRES regions of
viral mRNAs also provide possible targets for
antiviral therapies.
Pelletier J, Sonenberg N. Internal initiation of
translation of eukaryotic mRNA directed by a
sequence derived from poliovirus RNA. Nature
Eckard Wimmer Nahum Sonnenberg 1988;334:320-325.
Jang SK, Krusslich HG, Nicklin MJ, Duke GM,
Palmenberg AC, Wimmer E. A segment of the
5 nontranslated region of encephalomyocarditis
virus RNA directs internal entry of ribosomes
during in vitro translation. J Virol. 1988;62:2636-
2643.
Fernndez-Miragalla O, Lpez de Quinto S,
Martnez-Salas E. Relevance of RNA structure
for the activity of picornavirus IRES elements.
Virus Res. 2009;139:172-182.
Cartoon of poliovirus genome with its complex internal ribosomal entry site (IRES, blue)

page 450

1988 Eckard Wimmer, Nahum Sonnenberg, colleagues internal ribosomal entry sites (IRES) in picornavirus mRNA
The National Center for Biotechnology Information (NCBI) is
part of the National Library of Medicine (NLM), a branch of
the National Institutes of Health (NIH). The NCBI was founded
in 1988 and houses several resources: (1) genome sequencing
data in GenBank; (2) an index of biomedical research articles
in PubMed; and (3) online databases through the Entrez search
engine. NCBI also houses a multi-disciplinary intramural
research program. Initially, the mission of the NCBI was to
aid in understanding the molecular mechanisms that affect
human health and disease. It was to create and maintain
public databases, develop software to analyze genomic data,
and to conduct research in computational biology. In time,
and through widespread use of the Internet, NCBI became
increasingly involved in supporting basic biological research,
that is molecular and cellular biology research. NCBI evolved
further to: (1) develop new methods to deal with the volume
and complexity of the data being accumulated faster and faster;
(2) develop methods to analyze the structure and function of
macromolecules; (3) develop computerized systems for storing
data in more accessible form; and (4) develop better means to
access analytic and computational tools by researchers and the
public. In the process of database development, NCBI formed
database standards such as the database nomenclature that is
also used by other databases. The NCBI database GenBank, the
nucleic acid sequence database, contains sequence information
from over 200,000 different organisms; it is the most popular
database in use today. The foundation of GenBank predates
the NCBI, having been formalized in 1982; it is linked to
the genomic database of the European Molecular Biology
Laboratory (EMBL) and the DNA Database of Japan (DDBJ), all
part of the International Nucleotide Sequence Database project.

PubMed:
Since 1997 free access to the entire
MEDLINE dataset (plus others)
20 million articles, 10.4 million
with abstracts
2 million articles with full text in
PubMed Central
21,000 journals
16 million requests per day

page 451

1988 National Library of Medicine National Center for Biotechnology Information (NCBI) and its public databases
H. Fred Clark (1937-2012) and Paul A. Offit Albert Kapikian Richard Ward
Pentvalent human-bovine reassortant rotavirus vaccine With colleagues. Human-bovine With David Bernstein. Attenuated
(RotaTeq - Merck) reassortant rotavirus vaccine (several human rotavirus vaccine (Rotarix -
international manufacturers) GlaxoSmithKline)
In 1998, a rotavirus vaccine (RotaShield, Wyeth) containing an attenuated rhesus-human reassortant virus was licensed for use in the United States. Clinical trials in the
United States, Finland, and Venezuela had found it to be 80 to 100% effective at preventing severe diarrhea caused by the most common rotavirus variants, The vaccine,
however, was withdrawn from the market in 1999 after it was discovered that it may have contributed to a risk of intussusception in one of every 12,000 vaccinated infants.
Today, two newer vaccines (Rotarix, GlaxoSmithKline; RotaTeq, Merck) have been approved by FDA and another is in advanced development for use in developing countries
(Albert Kapikian, colleagues, NIH). Before the introduction of vaccine in the U.S. in 2006, rotavirus was the most common cause of severe gastroenteritis in children <5 years
of age. Although mortality is uncommon in the U.S., with an estimated 20 to 60 deaths annually, rotavirus gastroenteritis results in 55,000 to 70,000 hospitalizations, 200,000
emergency department visits, and 400,000 outpatient visits each year among children <5 years of age. Rotavirus disease in developing countries is much more significant:
worldwide, diarrhea is the second most common cause of fatal childhood disease, estimated to cause ~1.4 million deaths among children aged <5 years. Clinical trials in
developed countries have shown high efficacy for the presently licensed vaccines, reducing hospitalizations due to rotavirus gastroenteritis by 96% and office visits by 86% it
is anticipated that efficacy will be similar in developing countries as rotavirus vaccination programs expand globally.
Clark HF, Offit PA, Ellis RW, Eiden JJ, Krah D, Shaw AR, Pichichero M, Treanor JJ, Borian FE, Bell LM, Plotkin SA. The development of multivalent bovine rotavirus (strain
WC3) reassortant vaccine for infants. J Infect Dis. 1996;174(Suppl 1):S73-80.
Kapikian AZ, Simonsen L, Vesikari T, Hoshino Y, Morens DM, Chanock RM, La Montagne JR, Murphy BR. A hexavalent human rotavirus-bovine rotavirus (UK) reassortant
vaccine designed for use in developing countries and delivered in a schedule with the potential to eliminate the risk of intussusception. J Infect Dis. 2005;192(Suppl 1):S22-29.
Patel MM, Steele D, Gentsch JR, Wecker J, Glass RI, Parashar UD. Real-world impact of rotavirus vaccination. Pediatr Infect Dis J. 2011;30(Suppl):S1-5. page 452

1988> Paul Offit, H. Fred Clark, Albert Kapikian, Richard Ward, others development of human rotavirus vaccines
Since 1988, morbilliviruses have been increasingly recognized as the
cause of mass mortality, first among harbor seals (Phoca vitulina)
and gray seals (Halichoerus grypus) (phocid distemper virus - PDV),
and then several other marine mammals: Baikal seals (Phoca sibirica)
(canine distemper viruses - CDV), bottlenose dolphins (Tursiops
truncatus) and striped dolphins (Stenella coeruleoalba) (both dolphin
morbillivirus - DMV), and harbor porpoises (Phocoena phocoena)
(porpoise morbillivirus - PMV). Morbillivirus antibody and isolates
have also been made and in some cases sequenced from: killer whales
(Orcinus orca), manatees (Trichechus manatus latirostris), polar
bears (Ursus maritimus), walruses (Odobenus rosmarus), long-finned
pilot whales (Globicephala melas) and fin whales (Balaenoptera
physalus). Clinical signs of morbillivirus infection in these animals
are rarely observed severe viral disease is likely to be incompatible
with life in the harsh environments in which these animals live;
however, infected animals have been found in poor condition and
in some instances with distemper-like lesions (oculonasal discharge,
conjunctivitis, keratitis, respiratory distress - pneumonia, diarrhea
and abortion).
Osterhaus AD, Groen J, Niesters H, van de Bildt M, Martina B,
Vedder L, Vos J, van Egmond H, Abou Sidi B, Ould Barham ME.
Morbillivirus in monk seal mass mortality. Nature 1997;388:838-839.
Osterhaus AD, Vedder EJ. Identification of virus causing recent seal
deaths. Nature 1988;335:20.

Albert D.M.E. Osterhaus Morbillivirus, ruptured virion showing Mahy BWJ, Barrett T, Evans S, Anderson EC, Bostock CJ.
herringbone nucleocapsid strands. Characterization of a seal morbillivirus. Nature 1988;336:115.
Negative contrast electron microscopy
Kennedy S, Kuiken T, Jepson PD, Deaville R, Forsyth M, Barrett T,
Unrooted phylogenetic tree van de Bildt MWG, Osterhaus AD, Eybatov T, Duck C, Kydyrmanov
(hemagglutinin gene) of A, Mitrofanov I, Wilson S. Mass die-off of Caspian seals caused by
the morbilliviruses (marine canine distemper virus. Emerg Infect Dis. 2000;6:637-639.
morbilliviruses in yellow)
An epizootic caused by dolphin
morbillivirus (DMV) killed
thousands of striped dolphins in the
Mediterranean Sea in 1990-1992.
The first victims were detected in
the Gulf of Valencia off the coast of
Spain; in the next two years, large
foci of infection were found further
east. Here, bottlenose dolphins
were also affected. By 1944, the
virus had moved into the Black
Sea, where it caused many deaths
among the subspecies of bottlenose
page 453
dolphin that inhabits that region.

1988> Albert Osterhaus, colleagues discovery of phocine distemper virus, marine mammal morbilliviruses
Manfred Eigen Esteban Domingo Peter Schuster John Holland

Juan de la Torre Eugene Koonin Adrian Gibbs Luis Villarreal page 454

1988 Manfred Eigen, others development of modern concepts of the origin of viruses, virus evolution, quasispecies
Development of modern concepts of the origin of viruses evolving into another via Darwinian mutation/selection processes, it is widely
presumed, on the basis of all evidence, that most of the families of viruses originated
From the earliest International Congresses for Virology, sessions on the origin and and evolved separately that the families of viruses that relate to the archaea, to the
evolution of the viruses were very popular, but most of the content was entirely bacteria and to eukaryotic organisms originated and evolved separately, as suggested
speculative and ungrounded in observational or experimental proofs lots of fun, but in the diagram below, where a pre-virus appears from the ancient mist and enters one
not too serious. In the past decade or so this has changed: only data-based concepts of the three virospheres, coincident with the formation of the three domains of life.
now have credibility and these are largely grounded in genomics and the rise of Eugene Koonin and his colleagues have envisioned the emergence of the eukaryotic
evolutionary biology, overall. The subject has gotten bigger entire conferences are cell as another melting pot of virus evolution, from which the major groups of
devoted to it, proceedings are published and biologists from outside the world of eukaryotic viruses originated as a result of extensive recombination of genes from
virology contribute. Nevertheless, data-based or not, the subject is full of strongly various bacteriophages, archaeal viruses, plasmids, and bits of evolving eukaryotic
held opinions. A decade ago, students were taught that there were three hypotheses genomes. As remarkable as this early end of the evolutionary spectrum is, it is
to explain the origin of viruses: (1) the progressive, or escape, hypothesis stated that equaled at the other end of the spectrum, where today we are still concerned with the
viruses arose from genetic elements (DNA, RNA) that escaped from cellular control evolution of new viral genotypes and consequent phenotypes (pathotypes, topotypes,
and gained the ability to move between cells; (2) the regressive, or reductionistic, etc.), which explain, say, the emergence of variant viruses with importantly altered
hypothesis asserted that viruses are remnants of cellular organisms or organelles; transmission and/or pathogenicity characteristics.
and (3) the virus-first hypothesis stated that viruses originated in a pre-cellular
world and predate or coevolved with their current hosts. The first hypothesis was Miller SL, Urey HC. Organic compound synthesis on the primitive Earth. Science
usually given most credibility, based on the notion that bacteriophages originated 1959;130: 245-251; and Origin of Life. Science 1959;130:1622-1624.
from bacterial genomes and eukaryotic viruses from eukaryotic genomes, etc. The Koonin EV, Senkevich TG, Dolja VV. The ancient virus world and evolution of cells.
discovery that virus genes came from diverse sources, as well as unique sources, Biol Direct. 2006;1:29.
and the discovery of the archaea and viruses of archaea confounded this, but better
Prangishvili D, Forterre P, Garrett RA. Viruses of the archaea: a unifying view. Nat Rev
notions arose only after further progress was made in discerning very distant
Microbiol. 2006;4:837-848.
viral genomic relationships and in discerning molecular structure/function
relationships among viral and cellular proteins. Slowly, it came to be realized
that viruses form a world of their own, and that their molecular features
strongly suggest that they are ancient, predating the Last Universal Cellular
Ancestor (LUCA). The main debate has shifted to those who suggest a long
period of acellular evolution (up to the actual emergence of the archaea and
bacteria), versus those who favor an early appearance of cells (or something
very similar to cells). Those who suggest the former have revived the virus-
first hypothesis. That is, viruses emerged from an assemblage of self-
replicating elements thriving in special environments (say, hydrothermal
vents or such), using inorganic compartments as their hosts. For those who
favor an early emergence of cellular organisms, viruses are usually con-
sidered to have originated in cells in which RNAs played the part of enzymes
(ribozymes) and were also the stuff of inheritance. This has been called the
RNA world. The literature on the evolution of life (whether viruses are alive
or not is another subject!) from the point of self-replication is large and
diverse the literature on the initial steps transforming inorganic molecules
(the primordial soup) into the constituents of living organisms is small, not
having moved much from the famous experiments of Stanly Miller and
Harold Urey in 1952, in which they simulated hypothetical conditions
thought to be present on the early Earth and synthesized organic compounds
from inorganic precursors. Once amino acids were formed, the rest was easy
but hardly explained. At another level of evolution, that of one virus
page 455

1988 Manfred Eigen, others development of modern concepts of the origin of viruses, virus evolution, quasispecies
Development of the concept of viral quasispecies
Every virus species, as defined by conventional phenotypic properties, exists as a genetically
dynamic, diverse population of virions in which individual genotypes have only a fleeting
existence. Most individual viral genomes differ in one or more nucleotides from the consensus
or average sequence of the population and over relatively short times genotypic drift occurs as
particular variants gain advantage. Genotypic drift over longer times leads to the evolution of
substantially different viruses. Manfred Eigen, John Holland and their colleagues introduced the
term quasispecies to describe such diverse, rapidly evolving and competing viral populations. If
viral nucleic acid replication were without error all progeny would be the same and there would
be no evolution of phenotypes. If the error rate was too high, mutants of all sorts would appear
and the viral population would lose its integrity. However, at an intermediate error rate, such as
is seen with most or all RNA viruses, the viral population becomes a coherent, self-sustaining
entity that resembles a metaphorical cloud of variants centered around a consensus sequence, but
capable of continuous expansion and contraction in different directions as new mutants continue
to emerge and others disappear within the population. Darwinian selection limits the survival of
the most extreme mutants extreme outliers do not survive and favors variants near the center
of the cloud since these best achieve fit in the environmental niche. Just as the center of a cloud
is unclear, so the consensus sequence at the heart of the quasispecies is considered inscrutable.
Any published viral genomic nucleotide sequence reflects a random choice of starting material,
one biological clone among many, more or less representative of the consensus sequence of the
genome of the population as a whole. Using new technologies, such as deep sequencing, it is
hoped that the kinds of population structures built around consensus sequences will become
discernable. Do population structures radiate outward from consensus in a stochastic manner,
creating a symmetrical cloud focused around one single master genome? Or do they radiate out
from a multiple number of dominant genotypes. How do these populations differ in different hosts
or different cell types or tissues? How do small adaptive changes, mutations that become fixed
in the consensus sequence, alter the surrounding mutant cloud? The evolution of quasispecies
would be expected to be most conspicuous in viruses with large RNA genomes, where non-
lethal changes might accumulate rapidly. Indeed, for example, the genomes of coronaviruses, Depiction of the quasispecies concept of Manfred Eigen
with the largest RNA viral genomes known, are fraught with genetic defects. At the mutation The box represents sequence space, that is the confines of all possible
rate commonly seen in single-stranded RNA viruses, that is 10-3 per nucleotide per replication genetic variants that might occur when a virus replicates through
cycle (i.e., 1 out of every 1,000 nucleotides in every coronavirus genome in every round of many infection cycles. The central spot represents the lack of variance
replication), would be changed. Since coronavirus genomes contain about 30,000 nucleotides, that would follow if the replication process was perfectly accurate and
every genome must differ from the next by at least one nucleotide. From this, one might wonder environmental selective pressures were constant. The cloud represents
how coronaviruses or other RNA viruses can maintain their identities as pathogens over any the viral population diversity that actually follows upon an intermediate
evolutionarily significant period of time; why have these viruses not mutated out of existence? error rate in replication. The population, overall, becomes a coherent,
The answer lies in the quasispecies concept, with all its stabilizing influences. self-sustaining entity that metaphorically resembles a cloud in that its
Eigen M. Viral quasispecies. Sci Am. 1993;269:42-49. center, the original consensus sequence, is inscrutable, while its edges
represent probes pushing into the environment seeking better-and-better
Prangishvili D, Forterre P, Garrett RA. Viruses of the archaea: a unifying view. Nat Rev Microbiol. fit. It has been found that the evolution of many viruses operates at the
2006;4:837-848. level of the quasispecies, not at the level of individual virion genotypes,
Vignuzzi M, Andino R. Picornaviruses fidelity mutants and biological implications. In: Domingo and the result is the continuing emergence of new viral phenotypes,
E, Ehrenfeld E, Roos R, editors. Picornaviruses: molecular biology, evolution and pathogenesis. some of which cause new and/or more severe disease.
Washington DC. ASM Press; 2010.
page 456

1988 Manfred Eigen, others development of modern concepts of the origin of viruses, virus evolution, quasispecies
In the early 1980s, Kary Mullis at Cetus Corporation
invented the polymerase chain reaction (PCR). In its
first iteration, a new aliquot of the thermolabile DNA
polymerase from E. coli had to be added after each
cycle, because the enzyme was denatured by the >90C
temperature used to dissociate the strands of the newly
formed DNA hence, PCR was slow and laborious.
Switching to the thermostable DNA polymerase, Taq
polymerase, was key to the evolution of PCR into its
present form, using fully automated thermal cyclers
and other streamlining instruments. Because of these
improvements over the initial invention, PCR became the
most widely applicable technique in molecular biology.
Taq polymerase was isolated from the thermophilic
bacterium, Thermus aquaticus, which was discovered
in 1966 in Mushroom Spring in the Lower Geyser Basin
of Yellowstone National Park by Thomas Brock and
Hudson Freeze, then of Indiana University. The optimum
temperature for Taq polymerase activity is 75-80C
and it can survive 97C for some time; it can replicate a
1,000 base pair strand of DNA in less than 10 seconds
at 72C. At Cetus, purifying the Taq polymerase was
achieved by David Gelfand and Susanna Stoffel and proof
Thomas D. Brock, Yellowstone National Park of the fidelity of its enzymatic activity was done by Ray
Saiki. Gelfand and Stoffel then cloned and successfully
over-expressed the enzyme in E. coli so that large
amounts of extremely pure product became available.
The refined PCR technique was seen to be so important
that Hoffmann-LaRoche paid more than $300 million
to acquire rights to it. Essentially, the company was
acquiring the rights to two patents, that of Gelfand, et al.
(1989) for purified Taq polymerase and that of Mullis, et
al. (1990) for the PCR technique itself. In 1989, Science
magazine established a new award called The Molecule
of the Year. Taq polymerase was the first awardee.
Brock TD, Freeze H. Thermus aquaticus, a
nonsporulating extreme thermophile. J Bact. 1969;98:289-
297.
Saiki RK, Gelfand DH, Stoffel S, Scharf SJ, Higuchi
R, Horn GT, Mullis KB, Erlich HA. Primer-directed
The Taq polymerase molecule
is shaped like a hand. The DNA enzymatic amplification of DNA with a thermostable
fits into the palm (between DNA polymerase. Science. 1988;239:487-491.
Thermus aquaticus, scanning electron microscopy the green and magenta residues)
Mullis KB. The unusual origin of the polymerase chain
when the enzyme is active
page 457
reaction. Sci Am. 1990;262:56-65.

1988 Thomas Brock discovery of Taq polymerase


Choo QL, Kuo G, Weiner AJ, Overby
LR, Bradley DW, Houghton M.
Isolation of a cDNA clone derived
from a blood-borne non-A, non-B
viral hepatitis genome. Science
1989;244:359-362.
Kuo G, Choo QL, Alter HJ, Gitnick
GL, Redeker AG, Purcell RH,
Miyamura T, Dienstag JL, Alter MJ,
Stevens CE, Tegtmeier GE, Bonino
F, Colombo M, Lee W-S, Kuo C,
Berger K, Shuster JR, Overby LR,
Bradley DW, Houghton M. An
assay for circulating antibodies to
a major etiologic virus of human
non-A, non-B hepatitis. Science.
1989;244:362-364.
Houghton M. The long and winding
road leading to the identification
of the hepatitis C virus. J Hepatol.
2009;51:939-948.

George Kuo, Qui-Lim Choo, Michael Houghton Daniel W. Bradley The approach that led to the
isolation and identification of HCV
The effort to discover the etiologic agent of non-A, non-B hepatitis, that is hepatitis C virus
(HCV), which started in 1982, took more than five years and was based in techniques that were
novel and unproven at the time. Daniel Bradley, at the Centers for Disease Control and Prevention,
showed that the agent could be transmitted to chimpanzees and that plasma from these animals
was infectious. However, despite heroic efforts the agent could not be grown in cell cultures. Then,
Michael Houghton and his colleagues George Kuo and Qui-Lim Choo at Chiron Corporation (now
part of Novartis International AG), entered into collaboration with Bradley; in 1985, they cloned
all the DNA and RNA they could find in samples of infected chimpanzee plasma. They carried out
large-scale immunoscreening of bacterial cDNA libraries derived from the chimpanzee plasma (this
was prior to the development of PCR) using sera from patients presumed to have post-infection
antibodies. Eventually, this approach led to the isolation of a single small cDNA clone that was proven
to be derived from the HCV genome by Harvey Alter at NIH. The clone was expressed and used to
develop an initial serologic test; Alter then ran this antigen against a coded reference set of human
sera and proved that the virus had been found. The clone also provided a handle allowing the entire
genome to be isolated and sequenced. Next, a blood test was developed and by the early 1990s all
blood donors were tested in a successful program to eliminate the virus from the blood supply the
yearly rate of new infections in the U.S. dropped from ~250,000 to ~25,000. The development of a
HCV vaccine has proven to be far more difficult the high mutation rate of the virus, the production
of quasispecies variation, and the frequent establishment of chronic infection and long-term shedding
has confounded vaccine development, which is just now showing promise of success. page 458

1989 Michael Houghton, Qui-Lim Choo, George Kuo, Daniel Bradley discovery of hepatitis C virus
Several methods for generating large peptide and oligonucleotide
libraries through what was called combinatorial chemistry came on
the scene in the mid-1980s. The field got its start in 1963, when R. Bruce
Merrifield introduced solid phase synthesis, whereby polypeptide or
oligonucleotide chains could be made in assembly-line fashion using
automated synthesizers. Microarrays and gene chips then grew out of
efforts by a team of scientists concerned with developing novel chemical
approaches to automate drug discovery. This team, at the time a unique
collaboration between academic and industrial scientists, was assembled
by Alex Zaffaroni in 1988 and had members from the company
Affymax (Stephen Fodor and his colleagues), Stanford University
(Patrick Brown and his colleagues), and others. In 1989, Affymax staff
members Leighton Read and Michael Pirrung invented VLSIPS (Very
Large Scale Immobilized Polymer Synthesis); this was based on the
analogy of semiconductor chip manufacture by photolithography it
was called light-directed spatially addressable chemical synthesis
and involved depositing onto a glass microscope slide peptides that
could be photoactivated and photoprotected in a complex system using
fluorescent markers to control the composition of the forming peptide
chain. Stephen Fodor became the overall project leader and advanced
the construction of high-density arrays of peptides and oligonucleotides
substantially he also invented a scanner employing fluorescent labels
Stephen P. A. Fodor Patrick O. Brown and confocal laser microscopy to measure each individual binding event
on the surface of the chip with extraordinary sensitivity and precision. At
this time, 1993, Fodor co-founded Affymetrix, Inc., first as a division of
Affymax, then as an independent entity. The first commercial microarray
system, the Affymetrix GeneChip, was developed in 1994, and by 2000
there was an explosion of technological advances and biomedical
applications, mostly following upon hundreds of millions of dollars of
investment by biotechnology and pharmaceutical companies. The initial
microarray consisted of 1,024 peptides in a 1.6 cm2 area generated in
a ten-step process [today, resolution is so good that >250,000 sites per
square centimeter is possible]. The Affymetrix groups 1991 paper in
Science became a citation classic and received the Newcomb-Cleveland
Award as the outstanding paper published in Science that year. About
the same time the Affymetrix group was developing the GeneChip,
several academic teams were developing alternative microarray systems;
of particular importance has been spotted microarray technology
Fodor SPA, Read JL, Pirrung MC, Stryer L, Lu AT, Solas D. Light-directed, spatially addressable developed at Stanford by Patrick Brown, Dari Shalon, Stephen J.
parallel chemical synthesis. Science 1991;251:767-773. Smith, Mark Schena and Ron Davis. In this system, the probes are
Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns oligonucleotides; i.e., cDNA or PCR products that correspond to
with a complementary DNA microarray. Science 1995;270:467-470. mRNAs. The probes are synthesized beforehand and then spotted
onto glass slides. [By convention the term probe is the ligand bound to
Lenoir T, Giannella E. The emergence and diffusion of DNA microarray technology. J Biomed Disc
the solid substrate, that is the glass slide, and the target is the sample of
Collab. 2006;1:11.
interest in the liquid phase.]
page 459

1989 Stephen Fodor, Patrick Brown, others development of microarray technology


van Regenmortel MHV. Virus species, a much overlooked
but essential concept in virus classification. Intervirology
1990;31:241-254.
van Regenmortel MHV. Viruses are real, virus species
are man-made taxonomic constructions. Arch Virol.
2003;148:2481-2488.
Gibbs A. Virus nomenclature descending into chaos. Arch
Virol. 2000;145:1505-1507.
Condit RC. Principles of virology. In: Knipe DM, Howley
PM, editors. Fields virology, fifth edition. Philadelphia:
Lippincott, Williams & Wilkins; 2007. p. 25-27.

Marc H. V. van Regenmortel


The science of virology had grown so much and so many viruses were recognized by the early 1960s that Andr Lwoff led the first comprehensive efforts to build a taxonomy
(and consistent nomenclature); this evolved by 1966 into the International Committee on Nomenclature of Viruses (soon changed to the International Committee on
Taxonomy of Viruses, ICTV). The ICTV wrestled with the fundamental problem of developing a taxonomic system that would accommodate the unique properties of viruses
and which could also anticipate advancements in the identification and characterization of viruses. One critical issue was whether the system should consider virus properties
in a monothetical, hierarchical fashion (a system based on a single characteristic, modeled after the Linnean system), or a polythetic, hierarchical fashion (a system based on
more than one shared common characteristic, without any one characteristic being essential for membership in the group). The taxonomy system chosen was monothetical
at the family and genus levels, but for many years the species level was left in a rather informal, vernacular status. One reality behind this was that virologists dealing with the
various virus families and genera, that is the ICTV Study Groups, used different characteristics in distinguishing viruses and groups of viruses below the genus level. From the
beginning, the system differed from that used by any other taxonomists (e.g., for bacteria or any higher organisms), but it was useful, it was used to the exclusion of competing
systems, and it stayed close to the interests of those virologists more interested in the product than the process (e.g., diagnostic virology, pathology). By 1970, the ICTV had
established two virus families each containing two genera, 24 floating genera, and 16 plant groups. It was inevitable that the species level should be addressed and in 1989
Marc van Regenmortel proposed that a virus species be defined as a polythetic class of viruses that constitutes a replicating lineage and occupies a particular ecological niche.
This definition was accepted by the ICTV in 1991 and work began to apply it to the entire system. By 2010, 6 orders, 87 families, 19 subfamilies, 348 genera and 2,285 species
of viruses had been formally recognized. Even though there had hardly ever been any disagreement over the use of the system at the order, family or genus level, there has
been considerable confusion at the species level, partly based in misunderstanding over the difference between the man-made taxonomic construction, the species, and the
real entity, the virus. This was exacerbated by controversial efforts to change the formal nomenclature for virus species, and by disagreements between the lumpers-and-the-
splitters, especially when important pathogenic viruses were reduced to sub-species levels in the hierarchy based solely on genomic sequence relationships. van Regenmortel
and a few colleagues have spent much effort trying to educate the community and resolve the differences of opinion and the widespread indifference that still leaves many
venues of virologic communication using various vernaculars that stand in the way of the kind of precision that the universal taxonomy system was meant to fix. page 460

1989 Marc van Regenmortel definition of virus species


When the first marine virus was described in 1955, it was largely ignored; however,
when in 1989 ivind Bergh and his colleagues recognized that viruses were
abundant in marine environments others entered the field and made astounding
discoveries. An amazing diversity of virion morphologies were discovered, but
more importantly, marine viruses have been recognized as having an important
role in ocean ecology and energy and nutrient cycles. These viruses infect
bacterioplankton, phytoplankton, microzooplankton and other unicellular
organisms, which constitute 90% of the living biomass of the oceans and produce
50% of the Earths oxygen it is estimated that viruses kill approximately 20%
of this biomass per day. This viral shunt moves organic matter from living
organisms into the particulate and dissolved pools of organic matter, where
some of it is converted into CO2 by degradative processes, but the rest is reused
in new organismal growth without being recycled to the bottom of the ocean
where reuse cycles are much slower. It is estimated that as much as one-quarter
of the primary organic synthesis in the ocean ultimately flows through the viral
shunt. The level of viral infection in the oceans is not at a steady state, so it has an
influence on temporal changes in CO2 levels in surface water and air. The mineral
elements that are liberated during viral lysis (iron, nitrogen, phosphorous) are
rapidly re-assimilated, also greatly influencing the abundance of microorganismal
communities. Each recent estimate of the abundance of marine viruses has been
higher than the preceding one (each being based upon newer methods): surface
seawater abundances routinely are found to be greater than 106 virus particles per
ivind Bergh Curtis A. Suttle ml. In marine sediments, abundances are even higher, with 108-109 particles per
ml typical in nearshore sediments. Electron microscopic studies had revealed a
plethora of virion morphologies, many absolutely bizarre, but genomic sequencing
of isolates and metagenomic sequencing of marine viral communities has revealed
an even more amazing diversity of species. Among marine virus genomes,
typically 60-80% of open reading frames show no similarity to any sequences in
GenBank. Furthermore, 65-95% of marine viral metagenomic sequences are not
similar to previously described sequences, suggesting that we have only begun to
scratch the surface of the true diversity of this econiche.
Bergh , Brsheim KY, Bratbak G, Heldal M. High abundance of viruses found in
aquatic environments. Nature 1989;340: 467-468.
Suttle CA, Chan AM, Cottrell MT. Infection of phytoplankton by viruses and
reduction of primary productivity. Nature 1990;347;467-469.
Fuhrman JA. Marine viruses and their biogeochemical and ecological effects.
Nature 1999;399:541-548.
Venter JC, Remington K, Heidelberg JF, Halpern AL, Rusch D, Eisen JA, Smith
HO, et al. Environmental genome shotgun sequencing of the Sargasso Sea. Science
2004;304:66-74.
Venters Sorcerer II Global Ocean Suttle C. The virosphere: the greatest biological diversity on Earth and driver of
Sampling Expedition: relative global processes. Environ Microbiol. 2005;7:481-482.
proportion of viral sequences
page 461 J. Craig Venter by virus type

1989 ivind Bergh, Curtis Suttle, Craig Venter, others high concentrations of diverse viruses in the ocean
Primary Human Genome Project Sequencing Sites
(Members of the International Human Genome Sequencing Consortium):
U.S.DOE Joint Genome Institute, Walnut Creek, California, integrating three
genome centers at Department of Energy national laboratories.
Baylor College of Medicine Human Genome Sequencing Center,
Department of Molecular and Human Genetics, Houston, Texas
The Wellcome Trust Sanger Institute, The Wellcome Trust Genome Campus,
Hinxton, Cambridgeshire, United Kingdom
Washington University School of Medicine Genome Sequencing Center,
St. Louis, Missouri
Whitehead Institute/MIT Center for Genome Research,
Cambridge, Massachusetts
Automated Sample Preparation, Whitehead Institute
page 462

1990 International Human Genome Sequencing Consortium launching of the Human Genome Project
Herpesvirus
scanning electron Human herpesvirus 7 (HHV7) was identified serendipitously in the cells of
Nitza Frenkel microscopy a healthy adult in 1990 by Nitza Frenkel and her colleagues. In infants and
children the virus often acts together with HHV6B causing exanthem subitum
(roseola), but it is the latter virus that is the primary cause of this childrens
disease. Like HHV6B, HHV7 infection is nearly universal antibody prevalence
for HHV7 reaches 75% in 3- to 6-year-old children and 98% in adults. In adults,
clinical manifestations of HHV7 infection have been primarily associated
with reactivation in immunocompromised persons the virus becomes latent
in peripheral blood mononuclear cells, particularly mature CD4+ T cells.
Although HHV7 antigen expressing cells are detectable in a wide number
of other sites, including skin, lungs, salivary glands, mammary glands, liver
and kidney, it is not know if all these tissues are involved in reactivation.
HHV7 DNA has been found in 93% of lesions from pityriasis rosea patients,
although often diagnosis in this adult disease is confounded by the close
genetic relationship between HHV6B and 7. In immunocompromised hosts,
particularly transplant recipients (and in particular bone marrow transplant
recipients), there is increased risk for symptomatic primary or reactivation
Herpesvirus capsid model disease associated with HHV6B as well as HHV7 infection interstitial
pneumonitis and encephalitis has been recognized, but are not common.
Frenkel N, Schirmer E, Wyatt L, Katsafanas G, Roffman E, Danovich R, June C.
Isolation of a new herpesvirus from human CD4+ T cells. Proc Natl Acad Sci
Case of exanthem subitum USA. 1990;87:748-752.
caused by human herpesvirus 7 BlackJ, PellettP. Human herpesvirus 7. Rev Med Virol. 1999;9:245-262.
page 463

1990 Nitza Frenkel, colleagues discovery of human herpesvirus 7 (HHV7)


PRRS virus
thin section electron microscopy

Cathrinus Terpstra James E. Collins David A. Benfield


Veterinary clinicians in the U.S. in the late 1980s noted the appearance in swine herds of a previously unrecognized disease with a clinical presentation of severe reproductive
losses in late gestation, increases in the number of weak live-born piglets, severe pneumonia in neonatal and nursery pigs, and poor growth and increased mortality (porcine
reproductive and respiratory syndrome, PRRS). A similar clinical syndrome was initially described in Germany in 1990 and spread to other countries in Europe over the
following years. The causative agent remained unknown, leading to the name mystery swine disease in the U.S. and several other names in Europe. Gert Wensvoort,
Cathrinus Terpstra and their colleagues at the Central Veterinary Institute (Lelystad), The Netherlands, were the first to describe a small, enveloped RNA virus as the causative
agent. A similar virus was isolated in the U.S. by James Collins, David Benfield and their colleagues in South Dakota and Minnesota. Comparison of the genomic sequence
of the isolates from Europe and the U.S. indicated that they belonged to the family Arteriviridae, genus Arterivirus, but differed genomically by ~40%. Since then, PRRS
has emerged as the most important disease of swine in the world; in the U.S. annual losses are estimated at $560 million. The virus continues to evolve, producing varying
pathotypes with varying mortality rates. For example, in recent years quite virulent strains have become a major problem in China.
Wensvoort G, Terpstra C, Pol JMA, Lask EA, Bloemraad M, de Kluyver EP, Kragten C, van Butten L, den Besten A, Wagenaar F, Broekhuijsen JM, Moonen PJM, Zetstra T, de
Boer EA, Tibben AhJ, de Jong MF, vanr Veld P, Groenland GJR, van Gennep JA, Voets MTh, Verheijden JHM, Braamkamp J. Mystery swine disease in the Netherlands: the
isolation of Lelystad virus Vet Q. 1991;13:121-130.
Terpstra C, Wensvoort G, Pol JM. Experimental reproduction of porcine epidemic abortion and respiratory syndrome (mystery swine disease) by infection with Lelystad virus:
Kochs postulates fulfilled. Vet Q. 1991;13:131-136.
Benfield DA, Nelson E, Collins JE, Harris L, Goyal SM, Robison D, Christianson WT, Morrison RB, Goryca D, Chladek D. Isolation of swine infertility and respiratory
syndrome virus (isolate ATCC VR-2332) in North America and experimental reproduction of the disease in gnotobiotic pigs. J Vet Diagn Invest. 1992;4:117-126.
Benfield DA, Nelson E, Collins JE, Harris L, Goyal SM, Robison D, Christianson WT, Morrison RB, Goryca D, Chladek D. Characteristics of swine infertility and respiratory
syndrome (SIRS) virus (isolate ATCC VR-2332) J Vet Diagn Invest. 1992;4:127-133. page 464

1990 Cathrinus Terpstra, others discovery of porcine reproductive and respiratory syndrome virus (PRRS virus)
The World Wide Web (WWW, the Web) is a system of interlinked hypertext documents accessed via the
Internet. The term is often mistakenly used as a synonym for the Internet itself, but the Web is a service
that operates over the Internet, as e-mail does. Web pages may contain text, images, videos, or multimedia;
navigation between pages is via hyperlinks. In 1984 at CERN (the European Organization for Nuclear
Research), engineer and computer scientist Tim Berners-Lee considered the problem of information sharing:
to link and access information of various kinds as a web of nodes in which the user can browse at will. He
was concerned that physicists from around the world who needed to share data had no common hardware or
software with which to do so. He wrote a proposal in March 1989 for a large hypertext database with typed
links, but it generated little interest. He then collaborated with Robert Cailliau, who rewrote the proposal in
a more persuasive style and sought resources within CERN to implement it. It worked. By 1990, Berners-Lee
had built all the tools necessary for a working Web: the HyperText Transfer Protocol (HTTP), the HyperText
Markup Language (HTML), the first Web browser (named WorldWideWeb, which was also a Web editor), the
first HTTP server software (CERN httpd), the first web server (http://info.cern.ch), and the first Web pages that
described the project itself and accessed the CERN telephone directory. However, the system could run only on
NeXT computers; Nicola Pellow created a simple text browser that could run on almost any computer. With all
this at hand, CERN made major investments in the expansion and refinement of the Web and it took off as the
Internet expanded. Berners-Lee is now Director of the World Wide Web Consortium.
Berners-Lee T, Fischetti M. Weaving the web: the original design and ultimate destiny of the World Wide Web
by its inventor. SanFrancisco: Harper; 1999.

Tim Berners-Lee

Visualization of various routes through a portion of the Internet


page 465

1991 Tim Berners-Lee development of the World Wide Web (WWW)


Since the chain termination method of DNA sequencing only yields
short products, longer sequences are obtained by fragmentation,
sequencing and re-assembly of fragment sequences to give the overall
J. Craig Venter Hamilton Smith sequence. One way this is done is by shotgun sequencing, which is fast
but requires multiple overlapping reads of the target DNA, obtained
by performing several rounds of fragmentation and sequencing.
Complex computer programs are then used to assemble the overlapping
ends of the reads forming the continuous sequence. This process
uses enormous amounts of computing power to deal with errors and
repetitive sequences; for example, to complete the Human Genome
Project, most of the human genome was sequenced at 12X or greater
coverage. In 1995, shotgun cloning/sequencing was adapted to larger
targets and was used by the Institute for Genomic Research (TIGR) to
sequence the genome of the bacterium Haemophilus influenzae and
then in 2000 by Celera Genomics to sequence the genome of Drosophila
melanogaster. It was then used in the Human Genome Project.
Although shotgun sequencing was the most advanced technique for
sequencing genomes from about 1995-2005, other technologies have
surfaced since then, called next-generation sequencing. These are very
Sequencing Lab, J. Craig Venter Institute fast but use even more computer power to assemble fragments.
page 466

1991 Craig Venter, Hamilton Smith (TIGR), others invention of shotgun cloning/sequencing methods
SISPA RDA

Gregory Reyes Michael H. Wigler


Virus discovery has been aided greatly by the development of sequence independent methodologies for the generation of genomic data and, indeed, quite a few important
viruses have been discovered in recent years solely by the use of such methods (table). The most prominent of these methodologies include representational difference analysis
(RDA) and sequence-independent, single-primer amplification (SISPA), each with several variations. The SISPA method, first developed by Gregory Reyes and Jungsuh Kim in
1991, entails the directional ligation of an asymmetric primer at either end of a DNA molecule. Following several cycles of denaturation, annealing and amplification, minute
amounts of the initial DNA are enriched and then cloned, sequenced, PCR amplified and analyzed. SISPA is especially useful when no nucleotide sequence data are available
and the nucleic acid of interest is present in only very small amounts these are the conditions present in the initial isolation and cloning of previously uncharacterized viral
genomes. When first developed, SISPA was incredibly tedious and took weeks to run, but, again, advances made it much more practicable. The same was the case for RDA
when first developed by Michael Wigler and his colleagues, but again technical improvements over the years have made it more practicable. This technique is based upon
amplifying sequences that are different in two genomic or cDNA samples. Genomes or cDNA sequences from two samples (i.e., a known and unknown virus or an unknown
virus in a clinical specimen) are PCR amplified and differences
analyzed using subtractive DNA hybridization. Both methods also Some Examples of Viral Genomes Initially Discovered or Characterized by Molecular Methods
employ high-throughput equipment, virus concentration methods Virus Disease Know* Genome Method Date
such as ultracentrifugation, and modern assays for detection and Parvovirus B19 Fifth disease Yes ssDNA Molecular cloning and DNA hybridization 1984
further characterization of products of interest. Both methods also Hepatitis C virus
Hepatitis E virus
Non-A, non-B hepatitis No ssRNA
Non-A, non-B hepatitis No ssRNA
Transmission in primates; molecular cloning and immunoscreening
Transmission in primates; molecular cloning and sequence similarity
1989
1990
are tied to downstream production of large amounts of such Retroviruses Various Yes ssRNA/dsDNA Degenerate or consensus primer PCR and sequencing 1990>
Rotavirus Gastroenteritis Yes Seg dsRNA SISPA 1992
products for diagnostic antigens, nucleic acid probes, etc. Astrovirus Gastroenteritis Yes ssRNA SISPA and immunoscreening 1993
HHV-8, KSHV Kaposis sarcoma No dsDNA RDA 1994
Reyes GR, Kim JP. Sequence-independent, single-primer GB viruses A and B Not known No ssRNA Transmission in primates and RDA 1995
GB virus C Not known Yes ssRNA SISPA, cloning and sequencing 1996
amplification (SISPA) of complex DNA populations. TTV (Torque teno) Not known No ssDNA RDA 1997
Bovine parvovirus Not known No ssDNA SISPA, cloning and sequencing 2001
Mol Cell Probes 1991;5:473-481. Coronavirus V-NL63 Respiratory disease No ssRNA Cell culture and VIDISCA 2004
Bocavirus Respiratory disease? No ssDNA Random PCR, cloning, sequencing 2005
Lisitsyn N, Lisitsyn N, Wigler M. Cloning the differences Parvovirus 4 Not known No ssDNA SISPA, cloning and sequencing 2005
between two complex genomes. Science 1993;259: 946-951. *Were the investigators searching for a specific type of virus genome?
SISPA, Sequence Independent Single Primer Amplification; VIDISCA, Virus Discovery cDNA AFLP (a SISPA variant);
RDA, Representational difference analysis
page 467

1991> Gregory Reyes, Michael Wigler, others development of novel techniques to find uncultivable viruses
Rebecca Rico-Hesse Robert Ellis Shope (1929-2004)

Short-tailed cane mouse


(Zygodontomys brevicauda)
Robert Tesh, Rosalba Salas
In 1989, physicians in the state of Portuguesa, Venezuela, became aware of an outbreak of a severe febrile disease, mainly among rural inhabitants of the southern part of
the state, which was characterized by fever, headache, myalgia, sore throat, weakness, anorexia, nausea, vomiting and occasionally convulsions. Many of the patients were
hospitalized because of unremitting fever, weakness, dehydration and hemorrhagic manifestations (epistaxis, bleeding gums, hematemesis, melena and menorrhagia). Between
1989 and 1997, 220 cases were reported with a fatality rate of 33%. The disease is now called Venezuelan hemorrhagic fever (VHF). All age groups and both sexes are affected,
but the highest incidence is in males 15-44 years of age mostly agricultural workers. A new arenavirus, Guanarito virus, was identified as the etiologic agent of the disease.
This was done by Rosalba Salas and her colleagues at the Venezuelan National Institute of Hygiene, working with international colleagues. Its natural reservoirs are the rodent
Zygodontomys brevicauda (cane mouse) and the cotton rat Sigmodon alstoni.
Salas RA, de Manzione N, Tesh RB, Ricco-Hesse R, Shope RE, Betancourt A, Godoy O, Bruzual R, Pacheco ME, Ramos B, Taibo ME, Tamayo JG, Jaimes E, Vasquez C, Araoz F,
Querales J. Venezuelan hemorrhagic fever. Lancet 1991;338:1033-1036.
de Manzione N, Salas RA, Paredes H, Godoy O, Rojas L, Araoz F, Fulhorst CF, Ksiazek TG, Mills JN, Ellis BA, Peters CJ, Tesh RB. Venezuelan hemorrhagic fever: clinical and
epidemiological studies of 165 cases. Clin Infect Dis. 1998;26: 308-313. page 468

1991 Rosalba Salas, Robert Tesh, others discovery of Guanarito virus, the cause of Venezuelan hemorrhagic fever
Rabies Virus Variants in Terrestrial Reservoir Host Species, United States

Phylogenetic relationship of
Jean S. Smith rabies virus variants in U.S.
(N gene, 1350 bp)
Rabies virus (genotype 1) is distributed worldwide and is endemic throughout the tropical, subtropical, and temperate regions
of Africa, North and South America, Asia, Europe, and Australia. Within each region virus genotypic variants exhibit different
degrees of host specialization and geographic compartmentalization. North American virus variants in terrestrial carnivores
show significant species-specific geographic distributions (map). The restriction of distinct variants of the virus in single or a few
related mammalian reservoir host species has been only recently appreciated it was made possible through the work of Jean
Smith and her collegues at the U.S. Centers for Disease Control and Prevention (CDC) and Charles Rupprecht and his colleagues,
first at the Wistar Institute and then with Smith at CDC, first by the use of monoclonal antibodies and then more recently by
genomic sequencing of large numbers of virus isolates. Genomic sequence analysis suggests that geographic variants of major
terrestrial carnivore hosts cluster phylogenetically within specific host lineages (skunk-bites-skunk-bites-skunk), without any
regard to the phylogenetic relatedness of the hosts, per se. This has made genotyping and the mapping of geographic spread and
temporal distribution of virus variants valuable in support of wildlife rabies control by bait-delivered vaccination. Genotyping and
the mapping of variants may have started with the U.S. and Canada, but today it is being done all around the world, again yielding
valuable information for prioritizing control programs. It is being done with bat rabies isolates as well, where control may be far off
but understanding the complex natural history in so many bat species is also proving valuable.
Rupprecht CE, Glickman LT, Spencer PA, Wiktor TJ. Epidemiology of rabies virus variants. Differentiation using monoclonal
antibodies and discriminant analysis. Am J Epidemiol. 1987;126:298-309.
Smith JS, Orciari LA, Yager PA, Seidel HD, Warner CK. Epidemiologic and historical relationships among 87 rabies virus isolates as
determined by limited sequence analysis. J Infect Dis. 1992;166:296-307.
Real LA, Childs JE. Spatial-temporal dynamics of rabies in ecological communities. In: Collinge SK, Ray C, editors. Disease ecology
Charles E. Rupprecht - community structure and pathogen dynamics. Oxford; Oxford University Press; 2006. p. 170-187.
page 469

1992 Jean Smith, Charles Rupprecht, others rabies virus genotyping and mapping of wildlife species variants
Joshua Lederberg Stephen Morse Robert Shope Margaret Hamburg Mark Smolinski
(1925-2008) (1929-2004)
It was the perfect storm a tempest that may happen only once in a century a noreaster created by so
rare a combination of factors that it could not possibly have been worse... (The Perfect Storm: A True Story
of Men Against the Sea, Sebastian Junger) A transcendent moment nears upon the world for a microbial
perfect storm. Unlike the meteorological perfect storm happening just once in a century the microbial
perfect storm will be a recurrent event. The two events share a common feature; a combination of factors is
the driving force behind each (Microbial Threats to Health, Emergence, Detection and Response, 2003)

The Convergence Model


Convergence of factors leading to the emergence of an infectious disease.
Microbial Threats to Health, Emergence, Detection and Response.
1992 1996 2003 Washington DC: National Academies Press; 2003. page 470

1992> Joshua Lederberg, Stephen Morse, others the concept of New, Emerging and Re-emerging Infectious Diseases
The online Program to Monitor Emerging Diseases (ProMED and ProMED-Mail, the name first suggested by Robert Shope)
was established in 1994 under the auspices of the Federation of American Scientists. It was launched largely by the initiative
of Stephen S. Morse, following meetings with an advisory group. Early on, Morse was joined by Jack Woodall and Barbara
Hatch Rosenberg, the three becoming ProMEDs steering committee. Its principal intent was to assist local, national, and
international organizations in disseminating, as rapidly as possible, reports of outbreaks of infectious diseases wherever
they occur; these reports are taken from sources such as media reports, online summaries, local observers, official reports,
and others. ProMED circulates reports on animal, plant and human diseases and toxins from all corners of the globe.
This is presently done through 26 expert moderators, who add guidance and context to the reports. As volunteers, the
moderators are free to act independently, enabling ProMED to play a silent but important role in furthering more government
transparency and even gently coercing governments and international organizations into action. With minimum funding,
the program started with 40 subscribers in 1994. Challenges that confronted the system early on, such as e-mail access in
remote locations, have proven less daunting than anticipated. That seems clear: the service now has over 50,000 subscribers
in more than 180 countries. Since 1999, the service has been housed at the International Society for Infectious Diseases, at
the Harvard School of Public Health. The online system is run by Oracle. In 2009, collaboration with HealthMap.org added
automated map-based geographic information to online reports. Further upgrades in the technology have been underwritten
by Google.org.
Morse SS, Rosenberg BH, Woodall J. Global monitoring of emerging diseases: design for a demonstration program. Health
Policy 1996;38:135-153.
Morse, SS. Global infectious disease surveillance and early warning systems: ProMED nd ProMED-Mail. In: Global infectious
disease surveillance and detection: assessing the challengesfinding solutions, workshop summary, Institute of Medicine,
Forum on Microbial Threats. Washington DC: National Academies Press; 2007.

Stephen S. Morse

ProMEDs
original
Steering
Committee,
1994 (L-to-R):
Stephen Morse,
Jack Woodall,
Barbara Rosenberg
page 471

1993> Stephen Morse, others founding of the Program for Monitoring Emerging Diseases (ProMED)
Stuart T. Nichol C.J. Peters Thomas G. Ksiazek, Pierre E. Rollin
Cynthia S. Goldsmith
In 1993, a new hantavirus disease, an acute respiratory distress syndrome, was recognized in the southwestern region of the USA. Clinical signs included hypoxia, shock and,
in many cases, death. The story of how the etiology of the disease was determined and the virus characterized presents a lesson in modern virologic and epidemiologic science:
as the first patients were identified by clinicians and pathologists in New Mexico, specimens were sent to the U.S. Centers for Disease Control and Prevention. All test results
were negative except for low level serologic signals for the hantavirus, Puumala virus a most unexpected result, given that Puumala virus, from Finland, had never been
associated with such a disease. PCR primers were synthesized to Puumala virus genomic sequences and used as probes on tissues from patients results were positive and new
primers were made specifically to the virus present in the tissues. With new probes, tissues from 10 of 10 patients were positive. As more and more viral genomic sequence
was determined, a bacterial expression system was used to synthesize viral proteins, which were used as antigens for testing humans and animals for the presence of antibody.
Without a virus isolate, within 30 days of the first reported death, the virus had been identified as a new hantavirus, 42 cases had been confirmed, the principal virus reservoir
host had been identified (the deer mouse, Peromyscus maniculatus) and the public was informed on how to minimize risk. Months
later, the virus, named Sin Nombre virus, was isolated from deer mouse tissues by tedious, patient cell culture efforts. In 1996, it
was recognized that many other hantaviruses are also present in rodent populations throughout the western hemisphere, some
proven to be human pathogens, others not.
Nichol ST, Spiropoulou CF, Morzunov S, Rollin PE, Ksiazek TG, Feldmann H, Sanchez A, Childs J, Zaki S, Peters CJ. Genetic
identification of a hantavirus associated with an outbreak of acute respiratory illness. Science. 1993;262:914-917.
Childs JE, Ksiazek TG, Spiropoulou CF, Krebs JW, Morzunov S, Maupin GO, Gage KL, Rollin PE, Sarisky J, Enscore RE, Frey JK,
Peters CJ, Nichol ST. Serologic and genetic identification of Peromyscus maniculatus as the primary rodent reservoir for a new
hantavirus in the southwestern United States. J Infect Dis. 1994;169:1271-1280.
Elliott LH, Ksiazek TG, Rollin PE, Spiropoulou CF, Morzunov S, Monroe M, Goldsmith CS, Humphrey CD, Zaki SR, Krebs JW,
Maupin G, Gage K, Childs JE, Nichol ST, Peters CJ. Isolation of the causative agent of hantavirus pulmonary syndrome. Am J Trop
Sin nombre virus by Cynthia Goldsmith
Med Hyg. 1994;51:102-108. page 472

1993 Stuart Nichol, C.J. Peters, Thomas Ksiazek, others Sin Nombre virus and hantavirus pulmonary syndrome
Encyclopedia of Virology, Third Edition, five volumes, 2008, 3,234 pages, list price US$1,600.00.
The largest single reference work in virology, organized mini-review articles, covering all
biological, molecular, and medical topics concerning viruses of animals, plants, bacteria
and insects. [This work has also been issued by the publisher as four separate volumes: Desk
Encyclopedia of General Virology, Desk Encyclopedia of Human and Medical Virology, Desk
Encyclopedia Animal and Bacterial Virology, and Desk Encyclopedia of Plant and Fungal Virology,
Mahy BWJ, van Regenmortel MHV, editors, 2009.]
Paraphrased from reviews: ...The encyclopedia has the intention of being all-encompassing at a
high level of quality. This is reflected in a large number of specialists (over 640; almost a Whos
Who of virology), coordinated by the two Editors-in-Chief and 12 Associate Editors, who have
all contributed in their areas of special expertise. ...It is amazing that viruses are found as obligate
parasites in cells of practically all branches of the tree of life (Eubacteria, Archaea and Eukarya). The
Encyclopedia presents an enormous body of knowledge in this respect in a very comprehensive way.
Robert G. Webster Allan Granoff (1923-2012) ...The chapters are alphabetically ordered, frequently according to the names of individual viruses or
virus species, genera, families or orders, interspersed with articles on particular diseases or on general
virology topics. Classification issues have received close attention, following the ICTV 8th Report
on Virus Taxonomy (2005). Glossaries preceding some of the chapters are useful in explaining
terms of specific significance for the chapter. ... This magnum opus represents a tremendous effort in
providing a synopsis of present knowledge in all branches of virology.

Brian W. J. Mahy Marc van Regenmortel

page 473 Granoff photo courtesy St. Jude Childrens Hospital

1994> Robert Webster, Alan Granoff, Brian Mahy, Marc van Regenmortel Encyclopedia of Virology (three editions)
With the onset of the AIDS epidemic in the early 1980s,
there was a sudden epidemic resurgence of Kaposi
sarcoma (KS) affecting primarily gay AIDS patients, with
up to 50% of AIDS patients having this tumor. Analysis of
epidemiologic data by Valerie Beral, Thomas Peterman,
Ruth Berkelman and Harold Jaffe led them to propose
that KS was caused by an unknown sexually transmitted
virus that only causes tumors when the person becomes
immunosuppressed, as in AIDS. As early as 1984, it was
reported that by electron microscopy herpesvirus-like
structures were evident in KS tumors from this clue,
more than 20 agents were subsequently described as the
cause of KS, including cytomegalovirus and HIV itself.
The etiologic agent of KS, human herpesvirus 8 (HHV8),
was discovered by Yuan Chang, Patrick Moore, a wife
and husband team then at Columbia University, and their
team, in 1994. They did this by isolating fragments of the
genomic DNA of the virus from a KS tumor in an AIDS
patient. They used representational difference analysis
(RDA), a method based upon comparing DNA fragments
in the KS tumor tissue to unaffected normal tissue from
the same patient. In their initial RDA experiment, they
isolated two small DNA fragments that represented
less than 1% of the viral genome. These fragments
were similar to but distinct from known herpesvirus
genomic sequences, indicating the presence of a new
virus. Starting from these fragments, and working for
two years, they sequenced the entire genome of the virus
and using expression systems made immunoreagents.
Since this discovery, HHV8 has also been associated
with other diseases: primary effusion lymphoma,
Yuan Chang Patrick S. Moore multicentric Castlemans disease, angioendotheliomatosis,
angiosarcomas and dermatofibroma.
Beral V, Peterman TA, Berkelman RL, Jaffe HW.
Kaposis sarcoma among persons with AIDS: a sexually
transmitted infection?. Lancet 1990;335: 123-128.
Chang Y, Cesarman E, Pessin MS, Lee F, Culpepper J,
Knowles DM, Moore PS. Identification of herpesvirus-like
DNA sequences in AIDS-associated Kaposis sarcoma.
Science. 1994;266:1865-1869.
Moore PS, Chang Y. Detection of herpesvirus-like DNA
sequences in Kaposis sarcoma in patients with and
Kaposi sarcoma in an AIDS patient; tissue stained with H&E (left) and with a monoclonal antibody directed without HIV infection. N Engl J Med. 1995;332:1181-
at latent nuclear antigen, a protein consistently expressed in HHV8 infected cells (right) 1185. page 474

1994 Yuan Chang, Patrick Moore, colleagues discovery of human herpesvirus 8 Kaposi sarcoma herpesvirus
Directed genetic manipulation of RNA virus genomes depends on the ability to produce recombinant RNAs which
are accepted as templates by particular viral RNA-dependent RNA polymerases. Transcripts generated by the DNA-
dependent RNA polymerases most commonly used (e.g., phage T7 RNA polymerase and cellular RNA polymerase II)
are recognized by the polymerases of many positive-strand RNA viruses. This has allowed the recovery of infectious
viruses or replicons from cells transfected with cDNA transcripts this, in turn, has allowed the application of this kind
of recombinant DNA technology to the analysis of virus genomes, the production of mutants for pathogenesis studies,
the use of these viruses as as vectors for expressing foreign proteins, etc. However, the templates of the polymerases
of negative-strand RNA viruses are exclusively viral ribonucleoprotein complexes (RNPs), consisting of the genomic
RNA tightly encapsidated (within N or NP) and associated with a phosphoprotein (P). The RNPs apparently never
disassemble and RNA synthesis does not change the structure of the RNA-nucleoprotein template. That is, the RNP
proteins are actively required for viral gene expression and can be regarded as part of the template. Therefore, since the
genomic RNAs of these viruses do not function as mRNAs all the proteins involved in replication and transcription
have to be provided by other means. This hindered the application of recombinant DNA technology for study of these
viruses and the infections they cause. Success in developing reverse genetics of negative-strand RNA viruses with large,
non-segmented genomes (e.g., filo-, paramyxo- and rhabdoviruses) started with rabies virus and the work of Karl-Klaus
Conzelmann and his colleagues. Their approach was soon extended to many other nonsegmented negative-strand RNA
viruses and to the segmented negative-strand viruses as well. The key to recovering recombinant viruses was the use
of plasmids directing transcription of antigenome (positive-strand, messenger-sense) RNAs for each gene, rather than
genome RNAs. The use of positive-sense RNAs turned out to be successful for recovering infectious cDNA from every
rhabdovirus and paramyxovirus tried.
Schnell M.J., Mebatsion T., and Conzelmann K.K. Infectious rabies viruses from cloned cDNA. EMBO J. 1994;13:4195-
4203.
Conzelmann K.K. Nonsegmented negative-strand RNA viruses: genetics and manipulation of viral genomes. Annu Rev
Genet. 1998;32:123-162.
Gene order of nonsegmented negative-strand RNA viruses; comparison of representative virus genomes.
Karl-Klaus Conzelmann Equivalents of the five basic genes are drawn as filled boxes: N (NP in paramyxoviruses; nucleoprotein),
P (phosphoprotein), M (matrix protein), and L (Large; catalytic subunit of the RNA polymerase).
Rabies virus (red) was the first negative-strand RNA virus expressed from cDNA
Some Negative-Strand RNA Viruses
Recovered from cDNA:
Rabies virus
Vesicular stomatitis viruses
Measles virus
Rinderpest virus
Human respiratory syncytial virus
Sendai virus (PIV1)
Human parainfluenza virus 3 (HPIV3)
SV5 virus
Bunyamwera virus

page 475

1994 Karl-Klaus Conzelmann, colleagues development of reverse genetics for negative-strand RNA viruses
Only one known case of naturally
contracted Sabi virus infection
has been documented, yet
the virus remains important
due to at least two laboratory
infections that have occurred.
The original natural case of Sabi
virus infection occurred in a
woman in the village of Sabi,
outside of Sao Paulo Brazil, in
1990. In this instance, severe
liver damage led physicians to
an initial diagnosis of yellow
fever. Following the patients
death, the virus was identified as
a then unknown arenavirus. The
viriologist who was responsible
for this identification, however,
contracted the disease; he,
fortunately, survived. Four years
later, at the Yale Arbovirus
Research Unit a visiting research
scientist was exposed to the virus
when a centrifuge tube broke
while he was purifying virus from
cell culture fluid. He became very
ill, but recovered.
Coimbra TLM, Nassar ES,
Burattini MN, de Souza LT,
Ferreira I, Rocco IM, da Rosa
The team from the Instituto Adolfo Lutz in So Paulo, Brazil, that discovered Sabi virus AP, Vasconcelos PF, Pinheiro
From left to right: Front - Luiza T. M. de Souza, Danya Moyses Fialho, Terezinha Lisieux Moraes Coimbra, Iray Maria Rocco; FP, LeDuc JW, Rico-Hesse R,
Back - Luiz Eloy Pereira, Ivani Bisordi Ferreira, Elza da Silva Nassar, Esther L. Boccato Chamelet, Raimundo Nonato Gonzalez J-P, Tesh RB, Jahrling
PB. New arenavirus [Sabi]
isolated in Brazil. Lancet
1994;343:391-392.

Left to right:
Amelia Travassos da Rosa
Francisco Pinheiro
Rebeca Rico-Hesse
James LeDuc
Peter Jahrling
Robert Tesh page 476

1994 Terezinha Lisieux Moraes Coimbra, others Sabi virus and Brazilian hemorrhagic fever
Keith Murray Alex Hyatt Peter Hooper Hendra virus
director, Australian Animal Health negative contrast electron microscopy
Laboratory, 1994, with Fred Murphy
Hendra virus (HeV) was first identified during the first recorded outbreak of the disease that took place in the
Brisbane suburb of Hendra, Australia, in 1994. The index case, a mare, housed with 23 other horses, died after
two days of illness. Subsequently, 19 of the remaining horses became ill and 13 died. Both the trainer and a
stable hand who were involved in nursing the index case fell ill with an influenza-like illness within one week
of the horses death. The stable hand recovered, but the trainer died of respiratory and renal failure. A total of
13 outbreaks of Hendra virus infection have occurred since 1994, all confined to the east coast of Australia,
all involving infection of horses. The case fatality rate in horses has been approximately 75%. Four of these
outbreaks have spread to humans as a result of direct contact with infected horses. Horses have been identified
as intermediate hosts, transmitting infection from reservoir host fruit bats (flying foxes - family Pteropodidae,
genus Pteropus) to humans through close contact during care or necropsy of ill or dead horses. Symptoms of
HeV infection in humans range from mild influenza-like illness to fatal respiratory and neurological disease.
Neurologic disease has been associated with late sequelae (as long as 14 months after initial infection); this has
included persistent convulsions, personality changes and eventually death this resembles subacute sclerosing
panencephalitis caused by persistent measles virus infection.
Murray, K, Selleck P, Hyatt A, Gould A, Gleeson L, Westbury W, Hiley L, Selvey L, Rodwell B, Ketterer P.
A morbillivirus that caused fatal disease in horses and humans. Science 1995;268:94-97.
Investigating a Hendra virus outbreak Murray K, Rogers R, Selvey L, Selleck P, Hyatt A, Gould A, Gleeson L, Hooper P, Westbury H. A novel
Queensland, Australia
page 477 morbillivirus pneumonia of horses and its transmission to humans. Emerg Infect Dis. 1995;1:31-33.

1995 Keith Murray, Peter Hooper, Alex Hyatt, colleagues discovery of Hendra virus and its reservoir host fruit bats
Abridged from book reviews: Marburg virus first showed
up in 1967 in a vaccine factory in Germany, and was traced
to cells from African green monkeys. Seven people died, a
quarter of those infected. The first known Ebola outbreak
was in Sudan in 1976. The virus spread rapidly from
village to village, killing half of its victims. Two months
later, an even deadlier strain of Ebola hit Zaire, erupting
simultaneously in some 50 villages, killing nine out of ten
people it infected. Zaires president, Mobutu Sese Seko,
called out his army to seal off the entire zone of infected
villages, with orders to shoot anyone trying to come out.
Prestons account makes these events and the people
who investigated them unforgettable, tracing events back
to individuals with names and faces and stories, not only
the victims but the doctors and scientists willing to risk
their own lives to treat and investigate these mysterious
outbreaks. Centered on the events in Zaire, Preston opens
with a disturbingly graphic description of the meltdown
of a human body invaded by Ebola virus. He describes
the infection as essentially liquifying the body. The story
switches to the United States: in the fall of 1989, a monkey-
importing company with a primate quarantine unit in
Reston, VA (about ten miles from Washington, D.C.),
noted that monkeys were dying at an alarming rate and
with suspicious symptoms. When the cause was found to
be Ebola virus all hell broke loose. The Army quickly took
Random House, 1994 over, and a team was sent in to halt the spread of the virus.
The complicated and hazardous job of entering the monkey
Richard Preston house (the hot zone), killing each monkey, and retrieving
tissue samples is dramatically described. This operation had
to be conducted in secret to avoid public panic. That the
virus, now known as Ebola Reston, turned out not to affect
humans is small comfort: viruses mutate rapidly, and the
rain forests are only a plane ride away. ...A totally convincing
page turner, proving that truth is scarier than fiction.
Preston casts the story as a scientific thriller, which it is.
And he writes in the manner of such popular novelists as
Michael Crichton and Robin Cook, who have made the
strange virus outbreak into a literary convention of high-
tech, neo-Gothic horror. As a result, this book is hard to put
down, very scary, crammed with the detail that can make
fiction seem real or reality read like fiction. The genre
Richard Preston with New York Times #1 nonfiction Preston has inherited from the fiction writers draws you in
projected image of bestseller for more than a year, by amassing small, even trivial details, and he is a master at
Ebola virus in background published in more than 30 languages,
this.
sold more then 2.5 million copies. page 478

1995 Richard Preston publication of the influential book, The Hot Zone
Geographical distribution of patients
Robert Will James Ironside John Collinge at onset of symptoms of vCJD, United
Kingdom, through 2009 (n=172)

Variant Creutzfeldt-Jakob disease, human, left-to-right: March 20, 1996: announcement from the UK Spongiform Encephalopathy Advisory Committee
Spongiform change with single and multiloculated vacuoles (SEAC) that 10 people may have become infected with the BSE agent through exposure to beef: ...a
varying in size from one to 50 in diameter in the neuropil previously unrecognized and consistent disease pattern... ...although there is no direct evidence of a
of cortical gray matter. H&E. link, on current data and in the absence of any credible alternative, the most likely explanation is that
Florid plaques in the neuropil of cortical gray matter. H&E. these cases are linked to exposure to BSE before the bovine offal ban in 1989. This is a cause for great
concern.
Immunohistochemical staining for PrP; heavy staining of
focus in a lymphoid follicle. Nature, 1996: John Collinge and his colleagues report that PrPsc from patients with variant CJD
The crystallographic structure of the prion protein (PrP). has strain characteristics distinct from other types of CJD, but similar to PrPSc from mice, cats and
macaques infected with BSE. ...this is consistent with BSE being the source of this new disease [vCJD].
page 479

1996 Robert Will, James Ironside, John Collinge BSE prion, the cause of human variant Creutzfeldt-Jakob disease
Molecular Guidelines for Establishing
Microbial Disease Causation
A nucleic acid sequence belonging to a
putative pathogen should be present in most
cases of an infectious disease. Microbial
nucleic acids should be found preferentially
in those organs or gross anatomic sites known
to be diseased (i.e., with anatomic, histologic,
chemical, or clinical evidence of pathology)
and not in those organs that lack pathology.
Fewer, or no, copy numbers of pathogen-
associated nucleic acid sequences should
occur in hosts or tissues without disease.
With resolution of disease (for example,
with clinically effective treatment), the copy
number of pathogen-associated nucleic
acid sequences should decrease or become
undetectable. With clinical relapse, the
opposite should occur.
When sequence detection predates disease, or
sequence copy number correlates with severity
of disease or pathology, the sequence-disease
David N. Fredricks association is more likely to be a causal
relationship.
The nature of the microorganism inferred
from the available sequence should be
consistent with the known biological
characteristics of that group of organisms.
When phenotypes (e.g., pathology, microbial
morphology, and clinical features) are
predicted by sequence-based phylogenetic
relationships, the meaningfulness of the
sequence is enhanced.
Tissue-sequence correlates should be sought
at the cellular level: efforts should be made
to demonstrate specific in situ hybridization
Flow diagram demonstrating various approaches for the of microbial sequence to areas of tissue
detection and identification of viruses in clinical samples pathology and to visible microorganisms or to
using sequence-based technologies areas where microorganisms are presumed to
be located.
These sequence-based forms of evidence for
microbial causation should be reproducible.
Fredricks DN, Relman DA. Sequence-based
identification of microbial pathogens: A
reconsideration of Kochs postulates. Clin
David A. Relman Microbiol Rev 1996; 9:18-33. page 480

1996 David Fredricks, David Relman sequence-based criteria for proof of causation of viral diseases
Martin S. Hirsch David Ho Thomas C. Merigan
The concept and basic studies supporting combination antiretroviral therapy for HIV infection were developed in the laboratory of Martin Hirsch during the late 1980s, and
brought into clinical trials in the 1990s. Drug therapy has evolved ever since, embracing the concept of highly active antiretroviral therapy (HAART) and more recent iterations
of the concept. HAART has been defined in various ways, but it is commonly considered to consist of therapy using at least three anti-retroviral drugs, typically two nucleoside
or nucleotide reverse transcriptase inhibitors (NRTIs) plus a non-nucleoside reverse transcriptase inhibitor (NNRTI) or a protease inhibitor (PI) or a second NRTI. HAART
affords a potent way of suppressing viral replication while attempting to prevent the virus from developing resistance to individual drugs. HAART has been clearly shown to
delay progression to AIDS and prolong life. Presently, there are ~25 drugs and several fixed-dose drug combinations approved for use in HIV/AIDS patients: individual drugs
target many of the critical steps in the HIV replication cycle entry, reverse transcription, integration, and proteolytic processing. Choosing which drugs to use in which
clinical circumstance is complex and depends upon viral load, intercurrent infections, the practicality of each drugs use, etc. In the years following the introduction of HAART
therapy, the death rate from AIDS was reduced by 50 to 80% and changed from a nearly universally fatal illness to what is now often a manageable chronic illness.
Hammer SM, Katzenstein DA, Hughes MD, Gundacker H, Schooley RT, Haubrich RH, Henry WK, Lederman MM, Phair JP, Niu M, Hirsch MS, Merigan TC. A trial
comparing nucleoside monotherapy with combination therapy in HIV-infected adults with CD4 cell counts from 200 to 500 per cubic millimeter. AIDS Clinical Trials Group
Study 175 Study Team. N Engl J Med. 1996;335:1081-1090.

page 481

1996 Martin Hirsch, David Ho, Thomas Merigan, others development of HAART therapy for AIDS
In 1996, Jules Hoffmann and his colleagues discovered the main mechanism
by which fruit flies (Drosophila spp) combat infections. They used flies with
mutations in several different genes, including Toll genes, which encode
seven distinct families of inducible antimicrobial peptides. When they
infected flies with bacteria or fungi, they discovered that Toll mutants died.
They found that the product of the Toll genes were involved in sensing
pathogenic microorganisms and Toll activation was needed for successful
defense against invading pathogens. All were found to act via transmembrane
receptors, called Toll receptors. The discovery that Toll receptors are
essential for innate resistance to infection in flies led to the discovery of the
innate immune sensing functions of the mammalian Toll-like receptors, and
provoked a revolution in immunology. The mammalian Toll family, a dozen
invariant receptors, recognizes evolutionarily conserved microbial/viral
molecules, which activate expression of many different immune-response
genes, many of which regulate T cell responses. [Hoffmann is now studying
antiviral defenses in Drosophila. His findings point to RNA interference as a
potent defense mechanism, along with a STAT-JAK pathway (regarded as an
analogue of the mammalian interferon system)].
In 1998, Bruce Beutler and his colleagues were searching for a receptor that
could bind the bacterial product, lipopolysaccharide (LPS), which can cause
septic shock by overstimulating the immune system. They discovered that
Jules A. Hoffmann Bruce A. Beutler mice resistant to LPS had a mutation in a gene that was quite similar to the
Toll genes of the fruit fly. This Toll-like receptor (TLR) turned out to be the
Diagram from InvivoGen, 2011 elusive LPS receptor, which activates a cascade of mediators of inflammation.
These findings showed that mammals and fruit flies use similar molecules
to activate innate immunity. Beutler went on to discover the structure and
function of many of the Toll-like receptors, each detecting different signature
molecules that herald infection. These receptors also mediate severe illnesses,
including shock and systemic inflammation as it occurs in the course of septic
infection. They are also central to the pathogenesis of sterile inflammatory
and autoimmune diseases such as systemic lupus erythematosus.
Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. The
dorsoventral regulatory gene cassette sptzle/Toll/cactus controls the potent
antifungal response in drosophila adults. Cell 1996;86:973-983.
Beutler B, Eidenschenk C, Crozat K, Imler JL, Takeuchi O, Hoffmann JA,
Akira S. Genetic analysis of resistance to viral infection. Nature Reviews of
Immunology 2007;7:753-766.
Mueller S, Gausson V, Vodovar N, Deddouche S, Troxler L, Perot J, Pfeffer S,
Hoffmann JA, Saleh MC, Imler JL. RNAi-mediated immunity provides strong
protection against the negative-strand RNA vesicular stomatitis virus in
Toll-Like Receptors (TLRs) recognize pathogen-associated microbial patterns (PAMPs), Drosophila. Proc Natl Acad Sci USA. 2010;107:19390-19305.
which when stimulated initiate complex signaling cascades that lead to the activation of
transcription factors, such as AP-1, NF-B and IRFs, which in turn induce the secretion of
pro-inflammatory cytokines that direct the adaptive immune response. page 482

1996> Jules Hoffmann, Bruce Beutler, others discovery of Toll genes / Toll-like receptors, keys to innate immunity
The first indication that viruses could be used as transneuronal
markers came in the studies of Albert Sabin in 1938. He
studied the different routes of entry into the CNS of mice
following intranasal instillation by various neurotropic
viruses. Each virus studied produced a unique pattern: e.g.,
pseudorabies virus produced transneuronal infection in
multiple systems (sympathetic, parasympathetic, and sensory
systems); However, it was not until 1983 that Xavier Martin
and Michel Dolivo used pseudorabies virus to map trigeminal
nerve pathways. The current explosion of research in neuronal
pathway tracing started with the work of Henricus (Hans)
Kuypers (1925-1989)and Gabriella Ugolini, who in 1990
showed that herpes simplex virus could be used as a self-
amplifying transneuronal tracer. Since then, each neurotropic
virus used has had particular advantages: rabies virus has been
particularly valuable because after its injection into muscles or
nerves it is propagated exclusively by retrograde transneuronal
transfer without infection of unconnected neurons or
supporting glial cells, but it crosses many synapses enroute
to target sites in the brain. Recently, rabies virus mutants
Tracing monosynaptic inputs to single neurons: A
have been used; e.g., a glycoprotein gene deletion mutant has
single neuron in a slice of the cortex of a mouse was
been used which is only capable of infecting a single cell and
electroporated with plasmids encoding fluorophores
jumping across one synapse (first-order synaptic partner); this
and rabies glycoprotein (green). In a complex
Transneuronal tracers: (A) wheat germ has allowed investigation of local connectivity of neurons.
protocol, after six days the electroporated neuron
agglutinin-horseradish peroxidase (WGA- Other neurotropic viruses such vesicular stomatitis and mouse
expressed markers indicating successful transfection
HRP); (B) herpes simplex virus 1 (HSV 1) and hepatitis viruses have been used in mouse and rat models.
and virus infection. Thirty-six neurons were infected
pseudorabies virus (PrV); and (C) rabies virus. The advance of this technology to answer neuro-anatomic,
trans-synaptically by the virus (red). From the work of
With conventional tracers (A), only a small -physiologic and -pathologic questions has required incredible
James H. Marshel, Takuma Mori1, Kristina J. Nielsen
amount of the marker is transferred from attention to the details of virus strain and genotype/phenotype,
and Edward M. Callaway: Targeting single neuronal
directly labeled first-order neurons (1) to amount of virus used, timing, anatomic localization of entry
networks for gene expression and cell labeling in vivo.
second and third-order neurons (2, 3), thereby (in some instances via microinjection) and all specifics of the
Neuron 2010;67:562-574.
limiting usefulness. Viruses function as self- animal model employed. In the case of rabies, much is being
amplifying markers: however, herpesviruses learned about the neuropathogenesis of the infection as well,
(B) induce neuronal necrosis (X) and can also the infection that drives the host to the fury that leads to bite
propagate via cell-to-cell spread to local glial transmission.
cells and non-contacted neurons. In contrast,
rabies virus (C) propagates exclusively via
retrograde transneuronal transfer, regardless of Kuypers HG, Ugolini G. Viruses as transneuronal tracers. Trends Neurosci. 1990;13:71-75.
the dose and postinoculation time, and thereby Song CK, Enquist LW, Bartness TJ. New developments in tracing neural circuits with herpesviruses. Virus Res.
is particularly useful in tracing pathways. 2005;111:235-249.
Luo L, Callaway EM, Svoboda K. Genetic dissection of neural circuits. Neuron 2008;57:634-660.
Curanovic D, Enquist L. Directional transneuronal spread of alpha-herpesvirus infection. Future Virol. 2009;4:591-603.
Ugolini G. Rabies virus as a transneuronal tracer of neuronal connections. Adv Virus Res. 2011;79:165-202.
page 483

1996 Hans Kuypers, Gabriella Ugolini, others viruses as neuronal pathway tracers in viral pathogenesis research
PubMed was first released in 1996 as an
experimental database under the Entrez retrieval
system with full access to MEDLINE. The term
experimental was removed the next year. There
were approximately two million PubMed searches
per month by June 1997 current usage typically
exceeds three million searches per day. By 1998, the
PubMed database was redesigned to change the way
information is stored and retrieved, dramatically
increasing the speed of the system. Internet Grateful
Med started using the PubMed system to search
MEDLINE and MedlinePlus began linking to
PubMed for Health Topics. In 2000, PubMed began
linking to PubMed Centrals full-text articles and the
Bookshelf s initial book, Molecular Biology of the
Cell. PubMed continued to grow the 16 millionth
citation was added in 2005, the 20 millionth citation
was added in 2010 and at the same time the 2
millionth full-text article was added. Growth has
continued unabated ever since.

Inauguration of PubMed, 1996


National Center for Biotechnology Information
director David Lipman (left) coaches Vice President
Al Gore (seated) as he searches PubMed. NIH director
Harold Varmus (center) and National Library of
Medicine director Donald Lindberg (right) look on.
Gore inaugurated the PubMed search system at a
Capitol Hill press conference. With helpful prompts
from Lipman, Gore demonstrated the usefulness of
PubMed by performing the first three searches. At one
point, Gore typed in Should I get a flu shot?
page 484

1996 National Center for Biotechnology Information development of PubMed


Robert G. (Rob) Webster Kennedy F. Shortridge
A strain of H5N1 influenza virus (highly pathogenic avian influenza virus, HPAI A/H5N1) was
the causative agent of the epidemic that started in China and in 1997 spread to chicken farms
in the New Territories of Hong Kong. Surveillance of retail and wholesale poultry markets in
Hong Kong in late 1997 revealed that almost 20% of chickens and a small percentage of ducks
and geese were infected with this virus. The virus killed tens of millions of birds in China
and Hong Kong and hundreds of millions of others were culled to stem the spread of the
virus. It was considered to be an incipient pandemic situation, the chicken being the source
of virus for humans it was the first instance where a pandemic may have been averted. This virus was exceptional in that it infected humans directly; by 2010 there had been
510 human cases and 303 deaths. The case fatality rate was ~60%, but the bigger worry has been that this virus will continue to mutate and acquire increased transmissibility
characteristics. Indeed, over the period since 1997 the virus has continued to mutate, branching into five clades as it has moved around the world, but its capacity to infect
humans and cause severe disease and death has not changed. There is much speculation, but no consensus, about future virus evolution and its public health consequences.
Shortridge KF, Zhou NN, Guan Y, Gao P, Ito T, Kawaoka Y, Kodihalli S, Krauss S, Markwell D, Murti KG, Norwood M, Senne D, Sims L, Takada A, Webster RG.
Characterization of avian H5N1 influenza viruses from poultry in Hong Kong. Virology. 1998;252:331-342.
Zhou NN, Shortridge KF, Claas EC, Krauss SL, Webster RG. Rapid evolution of H5N1 influenza viruses in chickens in Hong Kong. J Virol. 1999;73:3366-3374.
Li KS, Guan Y, Wang J, Smith GJ, Xu KM, Duan L, Rahardjo AP, Puthavathana P, Buranathai C, Nguyen TD, Estoepangestie AT, Chaisingh A, Auewarakul P, Long HT, Hanh
NT, Webby RJ, Poon LL, Chen H, Shortridge KF, Yuen KY, Webster RG, Peiris JS. Genesis of a highly pathogenic and potentially pandemic H5N1 influenza virus in eastern
Asia. Nature 2004;430: 209-213.
page 485

1997 Robert Webster, others highly pathogenic H5N1 influenza virus and lethal disease in humans in Hong Kong
Torque teno virus 1 (TTV1, transfusion transmitted
virus) was discovered in 1997 in the plasma of a
Japanese patient suffering from posttransfusion
hepatitis by Tsutomu Nishizawa, Hiroaki Okamoto
and their colleagues. The virus was initially
discovered by means of representational difference
analysis (RDA) this technology allowed the
development of nucleic acid probes and expressed
proteins for use as immunological reagents.
Remarkably, given the notion that most human
viruses had been discovered by the 1990s, the virus
was found to be extremely common, reaching 100%
prevalence in some countries, and present in ~10%
of blood donors in the U.S. and U.K. Still more
remarkably, the virus was found to be the first of a
very large number of related viruses found in large
numbers in many animal species: chimpanzees,
several species of African monkeys, tamarins, cows,
pigs, cats, dogs, and tree shrews. These viruses
(there are at least 103 of them) have been classified
as a new family, the Anelloviridae, comprising
nine genera. The virions are 19-27nm in diameter,
non-enveloped, with icosahedral symmetry, and
have genomes that are circular, non-segmented,
negative-sense, single-stranded DNA, ~3,000-
4,000 nucleotides in size. The viruses or genomic
DNA have been found in commercial vaccines for
swine, enzyme preparations such as trypsin, cell
cultures and human drugs containing components
of porcine origin, so their significance as pathogens
is a matter of much ongoing research.
Tsutomu Nishizawa Hiroaki Okamoto
Nishizawa T, Okamoto H, Konishi K, Yoshizawa H, Miyakawa Y, Mayumi
M. A novel DNA virus (TTV) associated with elevated transaminase
levels in posttransfusion hepatitis of unknown etiology. Biochem Biophys
Res Commun. 1997;241:92-97.
Okamoto H, Nishizawa T, Ukita M. A novel unenveloped DNA virus
(TT virus) associated with acute and chronic non-A to G hepatitis.
Intervirology. 1999;42:196-204.
Okamoto H, Takahashi M, Nishizawa T, Tawara A, Fukai K, Muramatsu
U, Naito Y, Yoshikawa A. Genomic characterization of TT viruses (TTVs)
in pigs, cats and dogs and their relatedness with species-specific TTVs in
Torque teno virus 1 primates and tupaias. J Gen Virol. 2002;83:1291-1297.
negative contrast electron microscopy
page 486

1997 Tsutomu Nishizawa, Hiroaki Okamoto, colleagues torque teno virus, anelloviruses, new family, Anelloviridae
Some commercially
available analytes:
Adenovirus / Human
Cytomegalovirus / Human
Epstein-Barr virus EA / Human
Epstein-Barr virus NA / Human
Epstein-Barr virus VCA / Human
HBV virus Core / Human
HBV virus Envelope / Human
HBV virus Surface (Ad) / Human
HBV virus Surface (Ay) / Human
HCV virus Core / Human
HCV virus NS3 / Human
HCV virus NS4 / Human
HCV virus NS5 / Human
Hepatitis A virus / Human
Hepatitis D virus / Human
Hepatitis E virus orf2 3KD / Human
An example of microbead-based virus detection technology: Hepatitis E virus orf2 6KD / Human
Hepatitis E virus orf3 3KD / Human
James Malony, Sylvia Chong and their colleagues developed a multiplexed Herpes simplex virus 1 gD / Human
PCR assay for the detection of 20 different respiratory viruses (types/ Herpes simplex virus 1&2 / Human
Herpes simplex virus 2 gG / Human
subtypes) in a single test. The assay employed 14 virus-specific primer HIV1 gp120 / Human
pairs, followed by a multiplexed Target-Specific Primer Extension (TSPE) HIV1 gp41 / Human
reaction using 21 primers for specific respiratory virus types and subtypes. HIV1 p24 / Human
TSPE products were sorted and identified by using a fluid microsphere- HTLV-1&2 / Human
Human papillomavirus / Human
based Luminex array technology. The assay detected influenza A and B Influenza A H3N2 viruses / Human
viruses; influenza A virus subtypes H1, H3, and H5 (including subtype Influenza A viruses / Human
H5N1, Asian lineage); parainfluenza virus types 1, 2, 3, and 4; respiratory Influenza B viruses / Human
syncytial virus types A and B; adenoviruses; metapneumovirus; Measles virus / Human
Mumps virus / Human
rhinoviruses; enteroviruses; and coronaviruses OC43, 229E, and SARS Parainfluenza 1 virus / Human
coronavirus. They tested their assay using large numbers of diagnostic Parainfluenza 2 virus / Human
nasopharyngeal swab specimens, which were also tested by conventional Parainfluenza 3 virus / Human
methods, including culture they obtained very good sensitivity and Polioviruses / Human
Respiratory syncytial virus / Human
specificity. Rubella virus / Human
Mahony J, Chong S, Merante F, Yaghoubian S, Sinha T, Lisle C, Janeczko Varicella zoster virus / Human
R. Development of a respiratory virus panel test for detection of twenty
human respiratory viruses by use of multiplex PCR and a fluid microbead-
based assay. J Clin Microbiol. 2007;45:2965-2970.

Detection and identification of TSPE reaction


products captured onto beads employs proprietary
anti-tag oligonucleotides that hybridize to
complementary tag oligonucleotides. The
microbeads are sorted with a Luminex flow cell
instrument, which identifies spectrophotometrically
colored beads with one laser and a different colored
signal on the beads with a second laser.
page 487

1997> Luminex Corporation, others development of the first high throughput viral diagnostics instruments
Mechanism of RNA interference:

A. On entering the cell, long dsRNA


acts as a trigger of the RNAi
process.
B. It is first processed by the RNase
III enzyme Dicer.
C. Dicer processes long dsRNA
into 21-23 nt siRNA with 2-nt 3
overhangs.
D. The siRNAs are incorporated into
the RISC RNAi effector complex
(Argonaute protein, which cleaves
and discards the sense strand
leading to an active RISC.
E and F. The remaining antisense
strand of the siRNA duplex serves
as the guide strand and guides the
RISC to its homologous mRNA,
resulting in the endonucleolytic
cleavage of the target mRNA.

In 1998, Andrew Fire and Craig Mello discovered a mechanism that can degrade mRNA
from a specific gene; this mechanism, RNA interference (RNAi), is activated when double-
stranded RNA molecules occur in the cell. Double-stranded RNA activates the RNAi
mechanism which then degrades mRNA molecules with the same sequence as the double-
stranded RNA. When the mRNA molecules disappear, the corresponding gene is silenced
and no protein is made. RNAi is of great importance for the regulation of gene expression,
keeps jumping genes under control and participates in defense against viral infections.
RNAi is already being widely used in basic science as a method to study the function of
genes and it is hoped that it will lead to novel therapies in the future. In a series of simple
but elegant experiments, Fire and Mello deduced that RNAi is sequence-specific, that
RNAi can spread between cells and can even be inherited. It worked with tiny amounts of
Andrew Z. Fire and Craig C. Mello injected double-stranded RNA, causing Fire and Mello to proposed that RNAi is a catalytic
process. The components of the RNAi machinery were identified during the following
years. Double-stranded RNA binds to a protein complex, Dicer, which cleaves it into
fragments. Another protein complex, RISC, binds these fragments. One of the RNA strands
is eliminated but the other remains bound to the RISC complex and serves as a probe to
detect mRNA molecules. When an mRNA molecule can pair with the RNA fragment
on RISC, it is bound to the RISC complex, cleaved, degraded and silenced. In lower
organisms, such as arthropods, that have no immune system, it seems that RNAi plays a
particularly important role in defense against microorganisms and viruses.
Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic
interference by double-stranded RNA in Caenorhabditis elegans. Nature 1998;391:806-811.
page 488

1998 Andrew Fire, Craig Mello discovery of RNA interference (RNAi, RNA silencing by dsRNA)
In 1995, Larry Page and Sergey Brin met at Stanford University as graduate
students in computer science. By January of 1996, they began collaborating on
writing a program for a search engine to do back-link analysis. Fueled by rave
reviews for the search engine, they began working on an improvement, named
Google. Operating out of their dorm rooms, the pair built a server network
using cheap, used, and borrowed PCs. They tried to license their search engine
technology for what now seems a pittance, however, they failed to find a buyer
and decided to keep Google and seek financing to improve the product and take
it to the public directly. Co-founder of Sun Microsystems, Andy Bechtolsheim,
after a demo wrote a check for $100,000 and within a few weeks another $900,000
was obtained. In 1998, Google Inc. opened with a beta search engine quickly it
was answering 10,000 search queries per day. By 2010, worldwide, there are over
2 billion searches on Google per day. In addition to running the worlds most
popular search service, Google also runs a number of vertical search services
that are proving to be of great use to virologists, including: Google Book Search
(started in 2004); Google Scholar (2004); Google Maps (2004); Google Trends
(2004); Google Analytics (2007); and Google Universal Search (2007). The last
four of these have contributed to the entry of the company into public health
data analysis. For example, the Global Trends program led to Google Flu Trends,
which uses aggregated search data to estimate current influenza activity around
the world in near real-time. In a project done in collaboration with the Centers
for Disease Control and Prevention, certain search terms were found to be good
indicators of influenza activity as millions of users around the world search for
health information online. A close relationship was found between how many
people search for influenza-related topics and how many people actually have
influenza-like symptoms; results compared closely with traditional flu surveillance
systems and identified hot spots with regional and state-level specificity, all with
significantly less delay than with other systems. This project is being extended to
other infectious diseases, such as West Nile virus infection, respiratory syncytial
virus infection, and avian influenza (internationally).
Sergey Brin and Larry Page Ginsberg J, Mohebbi MH, Patel RS, Brammer L, Smolinski MS, Brilliant
L. Detecting influenza epidemics using search engine query data. Nature
2009;457:1012-1014.
Carneiro HA, Mylonakis E. Google trends: a web-based tool for real-time
surveillance of disease outbreaks. Clin Infect Dis. 2009;49:1557-1564.

the flu

Correlation between Google Flu Trends (blue)


and CDC surveillance data (red) for the U.S.
Mid-Atlantic Region from 2004 through 2008.
page 489

1998 Larry Page, Sergey Brin development of Google and web-based surveillance tools
Nancy Cox Yoshihiro Kawaoka Robert G. Webster
The molecular determinants that make certain influenza viruses highly pathogenic for mammalian species, including humans, remained poorly understood until the advent
of reverse genetic technology, wherein virulence characteristics of each viral gene from virulent and avirulent isolates could be assessed. The more that was learned, the more
complex the subject became. Influenza virus virulence in mammalian species is a polygenic trait, requiring, as first pointed out by Rudolf Rott and Christoph Scholtissek in the
1960s, a constellation of genes they also realized that viral virulence, host range and transmissibility are distinct genetic characteristics with different genetic linkages. In
1997, when highly pathogenic avian influenza (HPAI - H5N1) virus infected chickens and humans in Hong Kong with lethal effects, efforts were made in many labs, particularly
those of Yoshihiro Kawaoka, Hans-Dieter Klenk, Nancy Cox, Robert Webster and their colleagues, to determine the specific molecular determinants responsible. Determinants
responsible for: (1) receptor-binding and interspecies transmission (per hemagglutinin protein HA), (2) for host range restriction (per RNA polymerase PB2 protein); and (3)
interferon antagonistic activity (per nonstructural protein NS1) were found to correlate with virulence in complex ways. The best known virulence mechanism, discovered
earlier, is that based in the variable cleavability of the HA protein into its two active forms (HA1, HA2). The HA protein is synthesized as a precursor protein and its cleavage
is a prerequisite for fusion of viral and endosomal membranes, a key to viral infectivity in target tissues. The HAs of low pathogenicity viruses have amino acid sequences at
the cleavage site that are recognized by extracellular proteases that are secreted only by cells in the respiratory and intestinal tracts and consequently limit infections to these
organs. By contrast, highly pathogenic viruses have polybasic sequences at the cleavage site that are recognized by ubiquitous proteases that thus can trigger systemic infection.
The acquisition of a highly cleavable HA had been shown to be the key to the conversion of avirulent strains to virulent strains in several avian epidemics and this correlation
was then extended to humans (for example, all the avian viruses that killed people in Hong Kong in 1997 possessed a highly cleavable HA). Specific mutations in the PB2 and
NS1 proteins have also been specifically linked to changes in virus phenotype. More recently, mutations in several other genetic loci have been implicted in increased lethality.
Bender C, Hall H, Huang J, Klimov A, Cox N, Hay A, Gregory V, Cameron K, Lim W, Subbarao K. Characterization of the surface proteins of influenza A (H5N1) viruses
isolated from humans in 1997-1998. Virology 1999,254:115-23.
Suarez DL, Perdue ML, Cox N, Rowe T, Bender C, Huang J, Swayne DE. Comparisons of highly virulent H5N1 influenza A viruses isolated from humans and chickens from
Hong Kong. J Virol. 1998;72:6678-6688.
Hatta M, Gao P, Halfmann P, Kawaoka Y. Molecular basis for high virulence of Hong Kong H5N1 influenza A viruses. Science 2001;293:1840-1842.
Bogs J, Veits J, Gohrbandt S, Hundt J, Stech O, Breithaupt A, Teifke JP, Mettenleiter TC, Stech J. Highly pathogenic H5N1 influenza viruses carry virulence determinants
beyond the polybasic hemagglutinin cleavage site. PLoS One 2010;5:e11826. page 490

1998> Nancy Cox, Yoshihiro Kawaoka, Robert Webster, others molecular basis for influenza virus virulence
In 1998, an outbreak of acute febrile encephalitis associated with high
mortality was reported in one district of Malaysia, among pig-farmers; this was
followed by outbreaks in three other districts and in Singapore, all traceable
to the movement of pigs. A total of 265 cases of encephalitis with 105 deaths
occurred in Malaysia; this first episode was controlled through the culling of
>1 million pigs. At first Japanese encephalitis was suspected, so strongly that
alternatives were ignored. Then, Kaw Bing Chua, working in the Department
of Medical Microbiology, Faculty of Medicine, University of Malaya, isolated a
virus from serum and cerebrospinal fluid of patients. The virus rapidly caused
syncytia in infected Vero cell cultures. Chua used the infected cells as antigen
on spot slides and did indirect immunofluorescent antibody tests using patient
sera as the source of antibody (along with appropriate controls). In classical
fashion, Chua said, when I looked at the reaction, I could feel a chill going
down my spine. All the patients sera and cerebrospinal fluids had antibodies
that reacted with the infected Vero cells. The infected syncytial cells lighted
Kaw Bing Chua Kenneth Lam Sai Kit William J. Bellini up as like green fluorescence lanterns. I immediately called up the Head of
Department I told him that it was most likely a new virus and we needed
international assistance urgently to identify the virus Chua was sent to the
Centers for Disease Control and Prevention (CDC), in Fort Collins, Colorado
(not Atlanta, since the outbreak was still thought to be caused by an arbovirus).
At CDC, Nick Karabatsos ran immunofluorescence assays using the virus and
a panel of typing antibodies against known arboviruses all were negative.
Bruce Cropp examined by electron microscopy thin sections of virus-infected
Vero cells that Chua has brought from Malaysia but had not been able to
examine earlier they saw what appeared to be paramyxovirus nucleocapsids.
Everything was packed up and Chua traveled to CDC in Atlanta, where he
worked with a team led by Brian Mahy, William Bellini and Thomas Ksiazek.
The virus was rapidly identified as a novel paramyxovirus, which reacted
heterologously with antibodies to Hendra virus from Australia. This prompted
a search for the virus among fruit bats, the natural host and reservoir of
Hendra virus. Indeed, the virus was isolated from the urine and saliva of fruit
bats (Pteropus hypomelanus) in Malayia. In following years, outbreaks of
encephalitis occurred in India and Bangladesh and there have been further
Thomas G. Ksiazek Bryan Eaton Nipah, first episodes outbreaks in Malaysia.
Chua KB, Goh KJ, Wong KT, Kamarulzaman A, Tan PS, Ksiazek TG, Zaki SR,
Paul G, Lam SK, Tan CT. Fatal encephalitis due to Nipah virus among pig-
farmers in Malaysia. Lancet. 1999;354:1257-1259.
Chua KB, Bellini WJ, Rota PA, Harcourt BH, Tamin A, Lam SK, Ksiazek TG,
Rollin PE, Zaki SR, Shieh W, Goldsmith CS, Gubler DJ, Roehrig JT, Eaton
B, Gould AR, Olson J, Field H, Daniels P, Ling AE, Peters CJ, Anderson LJ,
Mahy BW. Nipah virus: a recently emergent deadly paramyxovirus. Science.
2000;288:1432-1435.
Chua KB. The discovery of Nipah virus: A personal account. Neurology Asia
page 491 Nipah virus encephalitis, human, H&E and immunohistochemistry 2004;9: 59-63.

1999 Kaw Bing Chua, Kenneth Lam, William Bellini, Thomas Ksiazek, others discovery of Nipah virus
New York
City, 1999

Marcelle Layton W. Ian Lipkin Robert S. Lanciotti


In the summer of 1999, an outbreak of encephalitis was detected in New York City, which as it progressed over the following years became more and more significant. One
way to relate the story is with a timeline. August 23-28: Deborah Asnis, chief of infectious diseases at Flushing Hospital in Queens County, New York, recognized neurological
disease in several elderly patients. She notified Marcelle Layton of the New York City Department of Health and sent samples of blood and spinal fluid to the NY State
Department of Health and to the Centers for Disease Control and Prevention (CDC). August 31: CDC sent epidemiologists to New York City to investigate what was then
being called encephalitis of unknown etiology. September 3: CDC completed tests on specimens from the patients; all were serologically positive for St. Louis encephalitis
virus, and CDC announced this as the cause of the outbreak. New York Citys mayor announced a $6 million campaign to wipe out the citys mosquitoes and aerial spraying
started. September 21: based on partial genomic sequencing of a virus isolated from a patient, W. Ian Lipkin and his colleagues at the University of California, Irvine, surmised
that the virus was not St. Louis encephalitis virus but a related virus, either Kunjin or West Nile virus. September 24: Lipkins information was communicated to the New
York State and City Departments of Health and CDC. September 25: CDC scientists reported that the mosquito-borne illness might not be St. Louis encephalitis but West
Nile virus. This was a startling finding as West Nile virus had never before been found in the Western Hemisphere. Further sequencing done by Lipkin and his colleagues and
Jonathan Smith and his colleagues at the U.S. Army Medical Research Institute for Infectious Diseases (USAMRIID) (the latter using virus from bird tissues from the Bronx
Zoo submitted by Tracey McNamara) and by Robert S. Lanciotti and his colleagues at the CDC determined that the disease was caused by a particular genetic lineage of West
Nile virus, one that had likely originated in Israel. Before the first frost and the end of the mosquito transmission season, there had been ~8,200 people infected (~2.6% of the
risk population), with ~1,700 cases of febrile illness, 62 cases of encephalitis, and 7 deaths. There were 25 cases of encephalitis in horses, with ~10 deaths. Concurrently, large
numbers of crows died in the greater New York metropolitan area. In succeeding years, the virus marched across the U.S., ultimately infecting humans, horses and wild birds
in 48 states (not Hawaii or Alaska), and Canada. There were an estimated ~16,000 cases of neuroinvasive disease by 2010. The virus also moved south, spreading across Mexico
and Central America.
Asnis DS, Conetta R, Teixeira AA, Walman G, Sampson BA. The West Nile virus outbreak of 1999 in New York: Flushing Hospital experience. Clin Infect Dis. 2000;30:413-418.
Nash D, Mostashari F, Fine A, Miller J, OLeary D, Murray K, Huang A, Rosenberg A, Greenberg A, Sherman M, Wong S, Layton M. 1999 West Nile Outbreak response
working group. The outbreak of West Nile virus infection in the New York City area in 1999. N Engl J Med. 2001;344:1807-1814.
Briese T, Jia XY, Huang C, Grady LJ, Lipkin WI. Identification of a Kunjin/West Nile-like flavivirus in brains of patients with New York encephalitis. Lancet 1999;354:1261-
1262.
Lanciotti RS, Roehrig JT, Deubel V, Smith J, Parker M, Steele K, Crise B, Volpe KE, Crabtree MB, Scherret JH, Hall RA, MacKenzie JS, Cropp CB, Panigrahy B, Ostlund
E, Schmitt B, Malkinson M, Banet C, Weissman J, Komar N, Savage HM, Stone W, McNamara T, Gubler DJ. Origin of the West Nile virus responsible for an outbreak of
encephalitis in the northeastern United States. Science 1999;286:2333-2337. page 492

1999 Deborah Asnis, Marcelle Layton, W. Ian Lipkin, Robert Lanciotti, others West Nile virus in the United States
In 2001, Albert Osterhaus, Bernadette van den Hoogen and their
colleagues in the Department of Virology of the Erasmus Medical
Center, Rotterdam, reported the discovery of a novel virus from
children with respiratory tract illness in The Netherlands. The virus
was detected in respiratory secretions of 28 children, but its genetic
characterization remained a mystery until use of a technique known
as randomly primed PCR (RAP-PCR), which allows identification of
unknown viruses grown in cultured cells. Based on sequence data,
the virus appeared to be closely related to avian pneumovirus and
so was named human metapneumovirus (hMPV). Several lines of
evidence suggested that hMPV was a common human respiratory
pathogen, causing upper respiratory tract infections, bronchiolitis,
and pneumonia; by the age of 5 nearly all individuals had evidence
of hMPV infection. It accounts for approximately 10% of respiratory
tract infections in children and may be the second most common
cause (after respiratory syncytial virus) of lower respiratory
infection in young children. The virus is distributed worldwide and
has a seasonal distribution comparable to that of influenza viruses
during winter. Reinfections with hMPV occur throughout adult life;
infections are generally subclinical or mild in adults but may be more
serious in the elderly, adults with underlying pulmonary disease, and
those who are immunocompromised. Phylogenetic analysis of strains
of hMPV has revealed that the epidemiology of hMPV is complex and
dynamic hundreds of variants have been identified. Unlike influenza
virus, where two or three strains spread around the world each year,
outbreaks of hMPV appear to be a local phenomenon. Strains of
hMPV differ from community to community, and strains identified
in one location may be quite similar to strains identified in other
Albert D.M.E. Osterhaus locations in different years.
Human metapneumovirus
negative contrast electron microscopy
(inset) ribonucleoprotein
van den Hoogen BG, de Jong JC, Groen J,
Kuiken T, de Groot R, Fouchier RA, Osterhaus
AD. A newly discovered human pneumovirus
isolated from young children with respiratory
tract disease. Nat Med. 2001;7:719-724.
Kuiken T, van den Hoogen BG, van Riel DA,
Immunohistochemistry of experimental Laman JD, van Amerongen G, Sprong L,
human metapneumovirus (hMPV) Fouchier RA, Osterhaus AD. Experimental
infection in the cynomolgus macaque. human metapneumovirus infection of
Immunoperoxidase staining of hMPV cynomolgus macaques (Macaca fascicularis)
antigens in bronchial epithelium, results in virus replication in ciliated epithelial
especially in the cilia and apical areas of cells and pneumocytes with associated lesions
ciliated epithelial cells. throughout the respiratory tract. Am J Pathol.
2004;164:1893-1900.
page 493

2001 Albert Osterhaus, Bernadette van den Hoogen, colleagues discovery of human metapneumovirus
The 2001 anthrax attacks in the United States occurred in two waves over the course of several weeks, beginning one week after the September 11th attacks. Letters containing
anthrax spores were mailed from Princeton, New Jersey, to several news media offices and to two U.S. Senators. There were eleven cases of inhalational anthrax, with five
deaths, and eleven cases of cutaneous anthrax, with no deaths. Seven letters are believed to have been mailed (three of these were inferred from the location of cases),
containing powders of two different qualities, one a crude brownish powder, the other a highly refined powder consisting of nearly pure Bacillus anthracis spores. The ensuing
investigation became one of the largest and most complex in history over the succeeding years FBI investigators interviewed 9,000 people, conducted 67 searches and issued
over 6,000 subpoenas. It is estimated that the cleanup and investigation cost >$1 billion. The analysis of the bacterium isolated from several of the envelopes indicated that
it was the Ames strain, a strain that had been distributed to sixteen labs within the U.S. (and to labs in Canada, Sweden and the United Kingdom). This strain was used most
prominently by the United States Army Medical Research Institute for Infectious Diseases (USAMRIID), Fort Detrick, Maryland. In 2008, the Department of Justice and the
FBI announced that charges were about to be brought against one individual, an employee of USAMRIID, but that he had taken his own life as he was about to be arrested.
In 2010, the government formally concluded the investigation and issued a final report, but there was so much criticism that the National Academy of Sciences was asked to
review the FBIs science and conclusions. In 2011, a report was issued that stated that there was insufficient evidence to support a definitive conclusion about the origin of the
letters mailed in 2011. There has been little progress reported since then. The anthrax attacks, in concert with the September 11th attacks, spurred significant increases in U.S.
government funding for biodefense research. For example, biodefense-related funding at the National Institute of Allergy and Infectious Diseases (NIAID) increased by $1.5
billion in 2003, and in 2004 Project Bioshield was funded, providing $5.6 billion over ten years for stockpiling vaccines and drugs against infectious agents considered most
likely to be used by terrorists. Other civilian federal agencies added considerably to the total expenditure, which reached ~$5 billion per year throughout the decade.
U.S. Department of Justice. Amerithrax investigative summary. 2010. Available from: http://www.justice.gov/amerithrax/docs/amx-investigative-summary.pdf.
Committee on review of the scientific approaches used during the FBIs investigation of the 2001 Bacillus Anthracis mailings. Review of the scientific approaches used during
the FBIs investigation of the 2001 anthrax letters. Washington DC: National Academies Press; 2011. page 494

2001> U.S. Government anthrax bioterrorism events, start of large biodefense research and response programs
Foot-and-mouth disease epidemic, United Kingdom, 2001
The 2001 epidemic of foot-and-mouth disease (FMD) in the United Kingdom was the most extensive ever recorded Impact of foot-and-mouth
in a developed country and instructive of how the virus can cause incredible economic loss. The 2001 epidemic disease, United Kingdom, 2001:
started silently on a pig farm illegally feeding unprocessed garbage (food scraps, probably from an imported meat 10 million animals killed
product); soon 90% of the herd exhibited clinical signs. The disease spread to a slaughterhouse servicing this farm, (1-in-8 of all farm animals)
and to a nearby sheep farm (the disease is difficult to recognize clinically in sheep), and from there through sheep cost 8-13 billion in 2001
markets across the country. Only after the widespread movement of infected sheep was a diagnosis made and (0.8% of GDP)(tourism 3.2
control efforts initiated; by this time, the virus had spread throughout England, Scotland and Wales. Although a billion, agriculture 2.4
strategic stockpile of vaccine was available, the control campaign relied upon the traditional policy of stamping billion, plus 1.5 billion in
out, that is slaughtering all animals on farms where infected animals were identified, and also all animals on farms 2002 and substantial
continuing costs)
where there was suspicion of exposure (called dangerous contact farms), and also on farms that were considered contiguous premises.
10,157 farms, 25% of all farms
In one area, this extended to all animals on premises within a 3 km zone around an infected premises. Stop movement orders also led to adversely affected
the slaughter of many animals on welfare grounds when feedstuff ran short. The economic consequences of infection within the United 7,800 farmers and farm
Kingdom were severe, partly grounded in the dilemma between the government wish (a) to achieve disease-free status as quickly as workers lost their jobs
possible, thereby allowing exports to resume, and (b) to achieve disease control with the minimum impact on the livestock industries and Estimates at the national level
communities. Clearly, these two ideals came into conflict, with great impact across the country. Since 2001, the British government and conceal the extent of the
the European Commission have issued new plans to deal with future epidemics, but they are remarkably similar to those in place for many, impact in the hardest-hit
many years: they are based on stamping-out of infected and in-contact herds, and on regional restrictions on the movement of susceptible rural areas
animals and their products. Provisions are made for the use of emergency vaccination. [abstracted from Gibbs P. The foot-and-mouth [in some instances official data
page 495 disease epidemic of 2001 in the UK: implications for the USA and the war on terror. J Vet Med Educ. 2003;30:121-132.] are are at odds with later studies]

2001 Foot-and-mouth disease epidemic in United Kingdom, destruction of very large numbers of livestock
Genomic structure of poliovirus 1 (PV1,
strain Mahoney) and strategy for the synthesis
of its full-length cDNA. (A) The positive-
stranded RNA of poliovirus is shown with
VPg at the 5 end of the non-translated
region (NTR). In the cDNA, VPg is replaced
by the T7 RNA polymerase promoter. The
polyprotein contains one structural (P1) and
two nonstructural (P2 and P3) domains. The 3
NTR contains a heteropolymeric region and is
polyadenylated (shown as AAAn). (B) PV1
cDNA carrying a T7 RNA polymerase
promoter at the 5 NTR end was subdivided
into three large fragments for the synthesis of
full-length cDNA. The sizes of the fragments
are shown. The genome sequence encoded by
each fragment was described separately. (C)
The three DNA fragments were synthesized
and assembled stepwise via common unique
restriction endonuclease cleavage sites to
yield full-length cDNA (F1-2-3 pBR322). The
sequence of the cDNA was confirmed.
Cello J, Paul AV, Wimmer E. Chemical
synthesis of poliovirus cDNA: generation
of infectious virus in the absence of natural
template. Science 2002;297:1016-1018.

Eckard Wimmer
In 2001, Eckard Wimmer and his colleagues, at the State University of New York at Stony Brook, stirred world-wide controversy when they announced that they had made
synthetic poliovirus in a test tube without a natural DNA or RNA template and without the use of living cells. Starting with the genomic sequence that is publicly available
from GenBank, they purchased from a commercial supplier oligonucleotides (averaging 69 nucleotides each) representing the cDNA for the entire virus genome. Given the
state of the art in 2001, the process of ligating the fragments was time consuming, but straight-forward. Once they had assembled the full-length cDNA, as shown above,
they transcribed it with RNA polymerase to make the single-stranded RNA genome of poliovirus. Next, they made infectious virus by incubating the RNA with a cell-free
extract from Hela cells. When injected into mice, the animals developed a neurological disease indistinguishable from that caused by native virus. This result for the first
time demonstrated the feasibility of biochemically synthesizing an infectious agent in the absence of a template. Two bases for controversy emerged: first, as the WHO Polio
Eradication Programme moved forward and it was presumed that one day polio vaccination would cease, the possibility that terrorists might easily synthesize a startup stock
of virus raised the question if vaccination could ever be stopped. Second, was the more theoretical question of whether other pathogens might someday also be synthesized
variola virus, et al. This, in turn, raised the question of whether genomic sequences of pathogens should be published or entered into public databases. The controversy
heated up further when the poliovirus synthesis which took two years of work was followed by Craig Venters announcement that the synthesis of the slightly smaller X174
bacteriophage from its genomic sequence had taken only a matter of weeks. Finally, the NIH, through its Recombinant DNA Advisory Committee (RAC), was forced to add a
new mandate for Institutional Biosafey Committees, that is a manditory review of biosafety considerations before approval of any recombinant DNA experiments.
page 496

2002 Eckard Wimmer, colleagues synthesis of poliovirus complementary DNA by assembling oligonucleotides
Until 2002, most molecular methods useful for virus discovery
were dependent upon precise complementarity between the
probes used and their viral targets such methods were not
easily extendable to the discovery of viruses that were not
represented in databases and probe or primer sets, although
some success was had using degenerate primers, etc. From 2002
onward, in the laboratories of Joseph DeRisi at the University
of California San Francisco and Ian Lipkin at Columbia
University, microarray technology employing longer probes
(e.g., >60 nt), which are more tolerant of sequence mismatches
and may detect agents that have only modest similarity to
those already known, evolved into platforms named Virochip
by DeRisi and GreeneChip by Lipkin. Although they differ in
design, both employ random amplification strategies to allow
an unbiased detection of viral and microbial targets. One key
to the success of these arrays was methodology for dealing
with host DNA and RNA sequences which are amplified along
with the viral or microbial sequences in the sample. Still, these
arrays have been most successful with acellular specimens,
such as virus cell culture supernatants, serum, plasma,
cerebrospinal fluid, urine, etc. Usually, hybridization of viral
or microbial sequences in specimens to probes representing
Joseph L. DeRisi W. Ian Lipkin pathogen targets is detected by binding of a fluorescent label;
(Virochip, 2002) (GreeneChip, 2005)
other readout methods are in development. Virochip and
GreeneChip arrays have had important successes: in a Marburg
hemorrhagic fever outbreak in Africa; in an Ebola virus (variant
Reston) outbreak in pigs in the Philippines; and in the discovery
that the etiologic agent of severe acute respiratory syndrome
(SARS) is a new coronavirus. The DeRisi and Lipkin labs have
established collaborative service systems to help others with
virus and microbial discovery.
Wang, D., L. Coscoy, M. Zylberberg, P. C. Avila, H. A. Boushey,
D. Ganem, and J. L. DeRisi. Microarray-based detection
and genotyping of viral pathogens. Proc Natl Acad Sci USA.
2002;99:15687-15692.
Palacios, G., P. L. Quan, O. J. Jabado, S. Conlan, D. L.
Hirschberg, Y. Liu, J. Zhai, N. Renwick, J. Hui, H. Hegyi, A.
Grolla, J. E. Strong, J. S. Towner, T. W. Geisbert, P. B. Jahrling,
C. Buchen-Osmond, H. Ellerbrok, M. P. Sanchez-Seco, Y.
Lussier, P. Formenty, M. S. Nichol, H. Feldmann, T. Briese, and
W. I. Lipkin. Panmicrobial oligonucleotide array for diagnosis
of infectious diseases. Emerg Infect Dis. 2007;13:73-81.
Lipkin WI. Microbe hunting. Microbiol Mol Biol Rev.
page 497
2010;74:363-377.

2002> Joseph DeRisi, W. Ian Lipkin, others application of microarray technology for identification of viruses
More than 99% of cervical cancers contain one or more of the
approximately 15 human papillomavirus (HPV) genotypes that
have been associated with the development of cervical cancer.
Approximately 50-60% of these cancers contain HPV16, and
another 10-20% contain HPV18 DNA; hence, these two HPV
genotypes were the focus of vaccine development efforts. For
many years, it was difficult to develop practical papillomavirus
vaccines because these viruses do not grow efficiently in
cultured cells. Even if they did, live attenuated vaccines would
contain viral oncogenes, which would prohibit their use. Ian
Frazer and Jian Zhou started work on a human papillomavirus
vaccine in 1990, using molecular methods to synthesize
virus-like particles (VLPs) that could mimic the virus as an
immunogen. In 1991 Zhous wife, Xiao-Yi Sun, combined two
expressed viral proteins into the first VLPs. Frazer and Zhou
filed for a patent in 1991 and sold partial rights to Merck,
where further development went forward. GlaxoSmithKline
independently used the same VLP-approach to develop a
similar vaccine under licensing of Frazers intellectual property.
It was found that a vaccine based solely on the L1 major capsid
protein was feasible the L1 protein alone folded correctly
and self-assembled into virus-like particles (VLPs) when
expressed in eukaryotic cells. These VLPs not only closely
resemble native virions morphologically, but also induce high
titers of protective antibodies. Since L1 is the only viral protein
required for the self-assembly of VLPs, in 2002 they began
producing it in recombinant yeast and baculovirus systems.
Ian Frazer vaccinating a young woman with HPV vaccine In 2008-2009, the FDA issued licenses to Merck and Glaxo
SmithKline and a drive to vaccinate young women began.
Human papillomavirus virus-like particles Zhou J, Sun XY, Stenzel DJ, Frazer IH. Expression of vaccinia
(VLPs), the basis for HPV vaccine recombinant HPV 16 L1 and L2 ORF proteins in epithelial cells
is sufficient for assembly of HPV virion-like particles. Virology
1991;185:251-257.
Kirnbauer R, Booy F, Cheng N, Lowy DR, Schiller JT.
Papillomavirus L1 major capsid protein self-assembles into
virus-like particles that are highly immunogenic. Proc Natl
Acad Sci USA. 1992;89:12180-12184.

Koutsky LA, Ault KA, Wheeler CM, Brown DR, Barr E,


Alvarez FB, Chiacchierini LM, Jansen KU. A controlled trial
of a human papillomavirus type 16 vaccine. N Engl J Med.
2002;347:1645-1651.
Schiller JT, Davies P. Delivering on the promise: HPV vaccines
and cervical cancer. Nat Rev Microbiol. 2004;2:343-347.
page 498

2002 Ian Frazer, Sanofi Pasteur, Merck Sharp & Dohme, GlaxoSmithKline development of papillomavirus vaccine
Thomas Weller, Frederick
Max Theiler, 1951 Robbins, John Enders, 1954

Erling Carl Jacob Norrby


Norrby E. A century of Nobel Prizes. Proc Am Philos
Soc. 2002;146, No. 4, 2002.
Norrby E. Yellow fever and Max Theiler: the only Nobel Peter Doherty, Rolf Zinkernagel, 1996 Stanley Prusiner, 1997
Prize for a virus vaccine. J Exp Med. 2007;204:2779-2784.
Since 2002, Erling Norrby has published a series of papers, and then a book, on the Nobel Laureates whose
Norrby E, Prusiner SB. Polio and Nobel prizes: looking discoveries in the field of virology have been seminal in the advance of the science. He has used his experiences
back 50 years. Ann Neurol. 2007;61:385-395. as Permanent Secretary of the Royal Swedish Academy of Sciences from 1997 to 2003, as well as those as a
Norrby E. Nobel Prizes and the emerging virus concept. distinguished virologist and professor and chair of the Department of Virology of the Karolinska Institute, as
Arch Virol. 2008;153:1109-1123. sources of information and perspective. The proceedings leading to the award of the Nobel Prizes are secret, but the
Norrby E. Nobel prizes and life sciences. Singapore: archives are opened 50 years after each award it is from these archives that much of the content of these papers
World Scientific; 2010. and book have come. Such interesting insight into the views and decisions of those involved! As more and more
Nobel Prizes in virology and related fields reach this 50 year anniversary, it must be hoped that Norrby will continue
page 499 with this project.

2002> Erling Norrby papers and book describing the awarding of the Nobel Prizes pertinent to virology
Eric Leroy and Jean-Paul Gonzalez Pierre Formenty Robert Swanepoel Pierre Rollin
Since the 1990s Ebola virus (strains
Zaire and Cte dIvoire) has been
spreading across the habitat of the
western lowland gorilla, decimating
the highly endangered species, even
possibly threatening its extinction. For
example, in the Democratic Republic of
Congo an epidemic that started in 2002
killed from 3,500 to more than 6,000
individuals, and it continues to spread
through the largest reserves in the
region. In the same region, in the same
epidemic, the disease also killed ~83%
of chimpanzees. As these episodes
continue, preliminary plans have been
Peter D. Walsh made to test and use one or more of the
Walsh PD, Abernethy KA, Bermejo M, Beyers R, De Wachter P, Akou ME, Huijbregts B, Mambounga DI, Toham AK, Kilbourn AM, Ebola virus vaccines currently under
Lahm SA, Latour S, Maisels F, Mbina C, Mihindou Y, Obiang SN, Effa EN, Starkey MP, Telfer P, Thibault M, Tutin CE, White LJ, development for human use, but the
Wilkie DS. Catastrophic ape decline in western equatorial Africa. Nature 2003;422:611-614. feasibility (vaccination via darts or
oral baiting with heat-stable vaccine),
Leroy EM, Rouquet P, Formenty P, Souquire S, Kilbourne A, Froment JM, Bermejo M, Smit S, Karesh W, Swanepoel R, Zaki SR, approvals and funding for this pose
Rollin PE. Multiple Ebola virus transmission events and rapid decline of central African wildlife. Science 2004;303:387-390. substantial challenges.
Rouquet P, Froment JM, Bermejo M, Yaba P, Dlicat A, Rollin PE, Leroy EM. Wild animal mortality monitoring and human Ebola
outbreaks, Gabon and Republic of Congo, 2001-2003. Emerg Infect Dis. 2005;11:283-290.
Bermejo M, Rodrguez-Teijeiro JD, Illera G, Barroso A, Vil C, Walsh PD. Ebola outbreak killed 5000 gorillas. Science. 2006;314:1564. page 500

2003 Peter Walsh, Eric Leroy, others epidemics of Ebola disease in endangered great apes threaten species survival
Scenes from sequencing centers of the Human Genome Project

The Human Genome Project (HGP) was the largest international collaboration ever undertaken in
biology. The HGP consortium included thousands of scientists worldwide who undertook the immense
task of sequencing the 3 billion bases of the euchromatic human genome, arrayed in its 24 chromosomes.
There were 20 sequencing centers in the United States, Great Britain, France, Germany, Japan, and
China. The five institutions that generated the most sequence were Baylor College of Medicine, Houston;
Washington University School of Medicine, St. Louis; Whitehead Institute/MIT Center for Genome
Research, Cambridge; Department of Energys Joint Genome Institute, Walnut Creek; and The Wellcome
Trust Sanger Institute, Cambridge, England. Between 2000, when the draft sequence was reported,
and 2003, the finished sequence was completed (with an accuracy of 99.99%), with highly contiguous
long runs (with the only remaining gaps corresponding to regions whose sequence cannot be reliably
resolved with current technology centromere, telomere regions, etc.). The estimated number of human
genes dropped below 25,000, according to the analysis of the finished human genome sequence. This
is a third fewer genes than was reported for the draft genome sequence, mostly because of improved
discrimination between genes and pseudogenes. The workhorse of the HGP was the automated capillary Improvements in the speed of DNA sequencing over the past
sequencer, which at the time was the state-of-the-art for throughput and accuracy. Even so, it took 30 years and into the future
over a year to read a DNA fragment one gigabase (one billion bases, or a third of the human genome) in size at a cost of ~$0.10 per 1000 bases. [By 2008, the worlds largest
sequencing center, the Wellcome Trust Sanger Institute, sequenced the equivalent of 300 human genomes in 6 months, and since then throughput has increased again using
machines that have reduced the cost per base down to ~$0.001 per 1000 bases.] In the HGP, the genome was first broken into conveniently sized chunks, fragments of about
150 kilobases. Each fragment was inserted into a bacterial artificial chromosome (BAC) cloning vector and the DNA fragments were produced in large amounts. The BACs
were then mapped, making re-assembly of the sequenced fragments easier and more accurate. Each of the large clones was then shotgunned broken into pieces of ~1,500
base pairs, either enzymatically or by physical shearing and the fragments were sequenced separately. Computers then analyzed the sequences of the small fragments and
assembled the original sequence of the clone. Finally, the whole genome was assembled and published in open databases.
International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 2001;409:860-921.
International Human Genome Sequencing Consortium. Finishing the euchromatic sequence of the human genome. Nature. 2004;431:931-945.
page 501

2003 International Human Genome Project completion of consensus sequence of the euchromatic human genome
Carlo Urbani (1956-2003) Leo Poon Thomas Ksiazek Albert Osterhaus Malik Peiris
Severe acute respiratory syndrome (SARS) emerged in 2003 in Guangdong, China,
and quickly spread to Hong Kong, and from there to 37 countries. By the time the
epidemic ended there had been 8,422 cases and 916 deaths (a case-fatality rate of 10.9%).
Carlo Urbani, the WHO staffer in Hong Kong, became infected in the course of his
epidemiologic investigation and died. Malik Peiris and his colleagues at the University
of Hong Kong discovered a new coronavirus as the cause of the disease, first by growing
it in cell culture, then by electron microscopy. Albert Osterhaus and his colleagues in
Rotterdam proved the etiologic association of the virus and disease using a cynomolgus
macaque (Macaca fascicularis) model. Tissues of palm civets (Paguma spp.) sold as food
in local markets were found to be silently infected with the virus, but later the reservoir
host was found to be bats (Rhinolophus spp.), which also were infected silently.
Ksiazek TG, Erdman D, Goldsmith CS, Zaki SR, Peret T, Emery S, Tong S, Urbani C,
Comer JA, Lim W, Rollin PE, Dowell SF, Ling AE, Humphrey CD, Shieh WJ, Guarner J,
Paddock CD, Rota P, Fields B, DeRisi J, Yang JY, Cox
N, Hughes JM, LeDuc JW, Bellini WJ, Anderson LJ.
A novel coronavirus associated with severe acute
respiratory syndrome. N Engl J Med. 2003;348:1953-
1966.
Drosten C, Gnther S, Preiser W, van der Werf
S, Brodt HR, Becker S, Rabenau H, Panning M,
Kolesnikova L, Fouchier RA, Berger A, Burguire
AM, Cinatl J, Eickmann M, Escriou N, Grywna
K, Kramme S, Manuguerra JC, Mller S, Rickerts
V, Strmer M, Vieth S, Klenk HD, Osterhaus AD,
Schmitz H, Doerr HW. Identification of a novel
coronavirus in patients with severe acute respiratory
syndrome. N Engl J Med. 2003;348:1967-1976.
page 502

2003 Carlo Urbani, Malik Peiris, Leo Poon, others discovery of SARS coronavirus and the global epidemic
During routine tests for Legionella bacteria in water from
a cooling tower in Bradford, UK in 1992, a strange new
type of virus was unknowingly isolated by a team led
by Thomas Rowbotham. At first, due to its size, it was
thought to be just another bacterium and was put in the
freezer. Subsequent investigations led to the discovery
of the remarkable Acanthamoeba polyphaga mimivirus
(mimivirus; mimi from mimicking microbe), initially
described by Bernard La Scola, Didier Raoult, Jean-
Michel Claverie and their colleagues at the Universit
de la Mditerrane, Facult de Mdecine, in 2003. The
virus was shown to infect the protozoan Acanthamoeba
polyphaga. At the time, mimivirus had the largest virion
size and largest genome of any virus known larger
than some bacteria. The genome is 1.18 Mbp in size,
and encodes ~1,262 genes, only ~10% of which encode
proteins of known function. The mimivirus icosahedral
capsid is about 500nm in size. From the capsid, there is
a dense fringe of 125nm long fibers, so the virion has a
total diameter of ~750nm (0.75 m; when stained the
virus is visible in the light microscope). The interior of the
Bernard B. La Scola Didier Raoult virion appears quite complex, with two lipid membranes
and a double-teardrop-shaped core. Recently, La Scola
and his colleagues discovered of an even larger relative
of mimivirus in a cooling tower in Paris; they named it
mamavirus. This virus was shown to infect the protozoan
Acanthamoeba castellanii. Mimivirus was shown to be
related to viruses infecting certain phytoplankton found
in marine environments the first of these organisms
shown to harbor a virus has been a marine microflagellate
called Cafeteria roenbergensis and the virus cafeteria
roenbergensis virus; it was discovered by Curtis Suttle and
his colleagues. The size and complexity of the mimivirus
genome challenge the established dividing line between
viruses and parasitic cellular organisms, suggesting that
ancient DNA viruses could have given rise to eukaryotic
cells as found in the plant and animal kingdoms. Mimivirus
has also been shown to be a human pathogen, associated
with community and hospital-acquired pneumonia.
La Scola B, Audic S, Robert C, Jungang L, de Lamballerie
X, Drancourt M, Binges R, Claverie JM, Raoult D. A giant
virus in amoebae. Science 2003;299:2033.
Acanthamoeba polyphaga mimivirus La Scola B, Marrie TJ, Auffray JP, Raoult D. Mimivirus in
thin section electron microscopy pneumonia patients. Emerg Infect Dis. 2005;11:449-452.
page 503

2003 Bernard La Scola, Didier Raoult, others discovery of mimivirus, at the time the largest virus known
APDS (Autonomous Pathogen Detection System)
Instrument next to lamppost in Times Square, New York City
Even before the 9/11 and anthrax terrorist attacks in 2001, a team at Lawrence Livermore National Laboratory (LLNL) had been developing what became the Autonomous
Pathogen Detection System (APDS) to provide early warning of aerosol-delivered biothreats. After 2001, its ownership fell to the U.S. Department of Homeland Securitys
BioWatch Program and was linked to the Centers for Disease Control and Preventions Laboratory Response Network. [The manual instrumentation developed before the
automted, autonomous system was called BASIS (Biological Aerosol and Sentry Information System)]. The analytical core of the series of instruments that were built over
the decade was a Luminex assay platform and flow cytometer, eventually to allow detection of many pathogen signatures at once. The program included instrumentation for
continuous air sampling and collection of micro-particulates, biological reagent (signature) development, technology for sample preparation, a system for result analysis, proof
testing, field packaging, communications, maintenance support, and remote monitoring. A requirement was that the system needed to continuously sample the air at a location
of interest over many months and produce regular timely reports on whether a biological threat agent was present. Other operational systems with similar missions were the
U.S. Postal Services Biohazard Detection System and the Department of Defenses Joint Biological Point Detection System (JBPDS). By 2002, units were developed that ran
multiplexed assays unattended for at least a week at a time, with results transmitted via a radio link. By 2004, the APDS was deployed to dozens of classified locations around
the country, and to two acknowledged locations: a Washington, DC metro station and Times Square in New York City. By 2006, in addition to multiplexed immunoassays
the APDS incorporated nucleic acid detection (using a flow-through PCR module and multiple primers developed for each pathogen signature of interest, running through a
Luminex platform). Criticisms have been that only few outdoor and indoor sites are covered, and that by focusing on air sampling other threats are left uncovered (e.g., water,
food, agricultural threat sites). The worst criticisms have concerned the endless jurisdictional turf wars between federal and other government agencies involved.
page 504

2003> Lawrence Livermore National Laboratory Autonomous Pathogen Detection System for biothreat agents
Top: Pierre Formenty, Tai
Forest, Gabon, 1997
Middle: Egyptian
fruit bats, Rousettus
aegyptiacus.
Bottom: Marburg
virus reservoir host
investigation, Kitaka mine
and cave, 2007.
Right: Robert Swanepoel,
Rousettus aegyptiacus.

From the time of the first known outbreak of Ebola hemorrhagic fever, in Sudan in 1976, bats were
suspected of playing a role in the natural history of the filoviruses. Bats were found under the roof of
the Nzara Cotton Factory where the index case and two other primary cases worked. The index case
of a 1979 outbreak also worked at the Nzara Cotton Factory. In the years following, some effort was
made to study the role of bats as reservoir hosts, usually with negative results, but in 1996 Robert
Swanepoel and his colleagues succeeded in finding evidence of infection in bats after inoculation of
Ebola virus. Then, between 2001 and 2005 Ebola virus was detected by Eric Leroy and his colleagues
at the Centre International de Recherches Mdicales de Franceville, Gabon, in three different bat
species (Hypsignathus monstrosus, Epomops franqueti and Myonycteris torquata) in Gabon and the
Republic of the Congo of 679 bats studied, specific Ebola antibody was found in 16 and viral RNA in
13. In the same region, in the wake of a large outbreak of Ebola hemorrhagic fever, massive migrations
of fruit bats were observed, with native people hunting them as bushmeat, and with index cases found
to have purchased freshly killed bats. In 2007, Marburg virus was detected in fruit bats in Gabon
and Republic of Congo in a study in which liver and spleen samples of 1,138 bats were analyzed
using a Marburg virus-specific real-time RT-PCR assay, four bats, all the Egyptian fruit bat Rousettus
aegyptiacus, were positive.
Leroy EM, Kumulungui B, Pourrut X, Rouquet P, Hassanin A, Yaba P, Dlicat A, Paweska JT, Gonzalez
JP, Swanepoel R. Fruit bats as reservoirs of Ebola virus. Nature 2005;438:575-576.
Towner JS, Pourrut X, Albario CG, Nkogue CN, Bird BH, Grard G, Ksiazek TG, Gonzalez JP, Nichol
ST, Leroy EM. Marburg virus infection detected in a common African bat. PLoS One. 2007;2:e764.
Leroy EM, Epelboin A, Mondonge V, Pourrut X, Gonzalez JP, Muyembe-Tamfum JJ, Formenty P.
Human Ebola outbreak resulting from direct exposure to fruit bats in Luebo, Democratic Republic of
Congo, 2007. Vector Borne Zoonotic Dis. 2009;9:723-728.
Pourrut X, Souris M, Towner JS, Rollin PE, Nichol ST, Gonzalez JP, Leroy E. Large serological
survey showing cocirculation of Ebola and Marburg viruses in Gabonese bat populations, and a high
page 505
seroprevalence of both viruses in Rousettus aegyptiacus. BMC Infect Dis. 2009;9:159.

2005> Eric Leroy, Jonathan Towner, others finding that reservoir hosts of Ebola and Marburg viruses are fruit bats
Beginning in 1995, Jeffery Taubenberger and his colleagues
at the Armed Forces Institute of Pathology obtained gene
sequences of the 1918 influenza virus from formalin-fixed
lung autopsy materials and from frozen lung tissues from
an Alaskan influenza victim buried in permafrost in 1918.
Painstakingly, over ten years the eight viral RNA genome
segments were reconstructed by plasmid-based reverse
genetics; that is, viral RNA was isolated (from fragments
under 200 nucleotides in length), then each fragment was
reverse transcribed into cDNA and amplified by RT-PCR.
The cDNAs were inserted into plasmids and amplified in
bacteria and then sequenced and assembled for infection
of cells in culture. When the reconstructed virus obtained
from the cell cultures was analyzed, it was found that all
eight genome segments were of avian derivation and all
differed in important ways from all other human viruses.
When the reconstructed virus was used to infect mice it was
extremely virulent it replicated to very high titer and was
much more lethal than contemporary viruses. Reassortant
Jeffery K. Taubenberger Terrence M. Tumpey Peter Palese viruses containing mixtures of genes from the 1918 virus
and other virus strains indicated that virulence was largely
based in the hemagglutinin gene, but was polygenic. Some
researchers had predicted that the 1918 virus would not be
more pathogenic than other human influenza viruses, that
its massive human toll in 1918 was the result of secondary
bacterial pneumonias and the absence of antibiotics, perhaps
along with the stress from World War I. The death of mice by
the reconstructed 1918 virus makes that scenario less likely.
Taubenberger JK, Reid AH, Krafft AE, Bijwaard KE, Fanning
TG. Initial genetic characterization of the 1918 Spanish
influenza virus. Science 1997;275:1793-1796.
Reid AH, Taubenberger JK, Fanning TG. Evidence of an
absence: the genetic origins of the 1918 pandemic influenza
virus. Nat Rev Microbiol. 2004;2:909-914.
The nucleotide sequence of Tumpey TM, Garca-Sastre A, Taubenberger JK, Palese P,
the eight RNA segments was Swayne DE, Basler CF. Pathogenicity and immunogenicity of
obtained using random primers, influenza viruses with genes from the 1918 pandemic virus.
reverse transcriptase and PCR.
Adolfo Garcia-Sastre cDNAs were constructed from Proc Natl Acad Sci USA. 2004;101:3166-3171.
synthetic oligonucleotides and Tumpey TM, Basler CF, Aguilar PV, Zeng H, Solrzano A,
the cDNAs cloned in plasmids. Swayne DE, Cox NJ, Katz JM, Taubenberger JK, Palese P,
Cells were transfected with
Reconstructed 1918 influenza the plasmids plus four helper Garca-Sastre A. Characterization of the reconstructed 1918
virus, negative contrast plasmids and influenza virus was Spanish influenza pandemic virus. Science 2005;310:77-80.
electron microscopy rescued and used to infect mice.
Cynthia Goldsmith, CDC page 506

2005 Jeffery Taubenberger, Terrence Tumpey, others 1918 influenza virus sequenced and virus reconstructed
Phylogenetic
relationships of
HTLV1/STLV1,
HTLV2/STLV2,
STLV3/HTLV-3
per 6812 bp gag-
Renaud Mahieux Antoine Gessain pol-env-tax of
HTLV3
(PTLV=primate
T-lymphotropic virus)
The discovery of close homologies between human T-lymphotropic virus 1 (HTLV1) and simian T-lymphotropic virus 1 (STLV1) subtypes (in a quite complex phylogeny),
led to the demonstration that most HTLV1 subtypes arose from interspecies transmission between monkeys and humans. Since a great number of STLV viruses had been
discovered in many species of monkeys in several countries in East, Central, and West Africa, in very diverse habitats, that it seemed likely that at least some of these viruses
would also have been transmitted to humans. Antoine Gessain, Renaud Mahieux and their colleagues of the Institut Pasteur in Paris investigated whether there might be
other human T-lymphotropic retroviruses and whether they might be pathogenic. This was technically difficult, since there could be no specific molecular probes, primers or
immune-reagents to search for unknown viruses. In 2005, they detected two new viruses in clinically normal people from Cameroon and named them HTLV3 and 4. [These
names are confusing: HTLV-III had been used for HIV early on, and HTLV-IV had been used to describe HIV2.] Sequencing showed that HTLV3 is a close homolog of STLV3,
but HTLV4 is not related to any known STLV. As part of their approach, they used a series of PCR assays that were designed to amplify all known HTLVs and STLVs. They also
used HTLV1 and 2 serologic tests (ELISA and western blot) results suggested that the new viruses are widespread but their prevalence is low. There was a substantial number
of indeterminate western blot test results, suggesting that there are more HTLVs yet to be discovered, of course with undetermined human pathogenicity.
Mahieux R, Gessain A. Les rtrovirus humains HTLV-3 et HTLV-4: nouveaux membres de la famille des HTLV [New human retroviruses: HTLV-3 and HTLV-4]. Med Trop
(Mars). 2005;65:525-528.
Calattini S, Chevalier SA, Duprez R, Bassot S, Froment A, Mahieux R, Gessain A. Discovery of a new human T-cell lymphotropic virus (HTLV-3) in Central Africa.
Retrovirology 2005;2:30.
Mahieux R, Gessain A. The human HTLV-3 and HTLV-4 retroviruses: new members of the HTLV family. Pathol Biol (Paris). 2009;57:161-166.
page 507

2005 Renaud Mahieux, Antoine Gessain discovery of human T lymphotropic viruses 3 and 4
Hepatitis C virus infection of a hepatoma cell line; immunofluorescence
After the discovery of hepatitis C virus (HCV) in 1988, and the rapid development of
nucleic acid-based diagnostic tests which allowed exclusion of the virus from the blood
Takaji Wakita Charles M. Rice supply, further progress was greatly inhibited by the inability to grow the virus in cell
culture. Then, in 2005 there were great breakthroughs in the labs of Frank Chisari at the
Scripps Research Institute and Charles Rice at Rockefeller University. The breakthroughs
stemmed from the use of an unusual isolate of HCV that was discovered in 2001 by
Takaji Wakita and his colleagues at the Tokyo Metropolitan Institute for Neuroscience
a replicon made from the nonstructural regions of this virus, when transfected into
hepatoma cells, was found to support virus replication; higher titers were obtained by
further optimizing hepatoma cell lines to be maximally compatible with the replicon. Full
length infectious clones were produced and drug targets identified, but since all this was
done with only one of the six major HCV genotypes, chimeras had to be produced to
capture the specificities of the other genotypes. Based on these advances, an HCV vaccine
now for the first time seems feasible.
Wakita T, Pietschmann T, Kato T, Date T, Miyamoto M, Zhao Z, Murthy K, Habermann
A, Krusslich H-G, Mizokami M, Bartenschlager R, Liang TJ. Production of infectious
hepatitis C virus in tissue culture from a cloned viral genome. Nat Med. 2005;11:791-796.
Lindenbach BD, Evans MJ, Syder AJ, Wlk B, Tellinghuisen TL, Liu CC, Maruyama T,
Hynes RO, Burton DR, McKeating JA, Rice CM. Complete replication of hepatitis C virus
in cell culture. Science 2005;309:623-626.
Zhong J, Gastaminza P, Cheng G, Kapadia S, Kato T, Burton DR, Wieland SF, Uprichard S,
Wakita T, Chisari FV. Robust hepatitis C virus infection in vitro. Proc Natl Acad Sci USA.
2005;102:9294-9299.
Moradpour D, Penin F, Rice CM. Replication of hepatitis C virus. Nat Rev Microbiol.
2007;5:453-463.
page 508

2005 Takaji Wakita, Charles Rice, others in vitro replication of hepatitis C virus by manipulation of virus and cells
Sample Input and Fragmentation
A variety of starting materials including
genomic DNA, PCR products, BACs and cDNA
may be used. Large DNAs are fractionated into
small, 300- to 800-basepair fragments.
Library Preparation
Short adaptors specific for both the 3 and 5
ends are added to each fragment. The adaptors
are used for purification, amplification, and
sequencing steps. Single-stranded fragments
with A and B adaptors compose the sample
library.
Bead Technology
The single-stranded DNA library is
immobilized onto DNA capture beads. Each
Roche/454 workstation the bright bead carries a unique single-stranded DNA
dots on the screen are individual
sequencing reactions occurring in the library fragment. The beads are emulsified (the
wells in a PicoTiter plate. If the entire water phase contains PCR reaction ingredients)
plate were full, there would be about resulting in isolation of each reaction bead in its
1.2 million bright dots.
own lipid micelle, thereby excluding competing
or contaminating sequences.
Emulsion PCR Amplification
Margulies M, Egholm M, Altman WE, Each DNA fragment is amplified within its own
Attiya S, Bader JS, Bemben LA, Berka
lipid micelle bead microreactor resulting in a
J, Braverman MS, Chen YJ, Chen Z,
copy number of several million per bead. This is
Dewell SB, Du L, Fierro JM, Gomes
XV, Godwin BC, He W, Helgesen S,
done in a massively parallel instrument. Then,
Ho CH, Irzyk GP, Jando SC, Alenquer the emulsion is removed and the amplified
fragments remain bound to their specific beads.
Jonathan M. Rothberg ML, Jarvie TP, Jirage KB, Kim JB,
Sequencing
Knight JR, Lanza JR, Leamon JH,
Jonathan Rothberg is the inventor of massively parallel Lefkowitz SM, Lei M, Li J, Lohman The clonally amplified DNA fragments are
DNA sequencing; he founded 454 Life Sciences KL, Lu H, Makhijani VB, McDade KE, enriched and loaded into a liquid-handling
Corporation, later acquired by Roche Diagnostics, and McKenna MP, Myers EW, Nickerson workstation (e.g., PicoTiterPlate, which
other companies, that have commercialized technologies E, Nobile JR, Plant R, Puc BP, Ronan has wells of a diameter that allow only one
for DNA sequencing that have significantly reduced MT, Roth GT, Sarkis GJ, Simons JF, bead per well). The sequencing reactions
the cost and increased throughput. The 454 technology Simpson JW, Srinivasan M, Tartaro (pyrosequencing by primer extension) are read
has been considered one of the great breakthroughs in KR, Tomasz A, Vogt KA, Volkmer GA, in the hundreds of thousands of wells using
molecular biology. He has also invented other futuristic Wang SH, Wang Y, Weiner MP, Yu chemiluminescent signals recorded by a CCD
technologies: an instrument for sequencing on a chip with P, Begley RF, Rothberg JM. Genome camera-microscope.
genomic information fed directly to digital information sequencing in microfabricated high- Data Analysis
analysis software; the first non-bacterial cloning system; density picolitre reactors. Nature. The positional information generated across
etc. 2005;437:376-380. the plate device allows software to determine
Rothberg JM, Leamon JH. The the sequence of ~1,200,000 individual reads
development and impact of 454 every 10-hour run. Sequencing data analysis
sequencing. Nat Biotechnol. and other bioinformatics tools support de novo
2008;26:1117-1124. assembly of up to 400 megabases.
page 509

2005 Jonathan Rothberg, 454 Life Sciences (Roche) development of 454 pyrosequencing
Tomteboda Home of European Centre for Disease Prevention and Control
(ECDC), Stockholm, Sweden

The European Centre for Disease Prevention and Control (ECDC) was established in 2005. It is an EU agency aimed at strengthening Europes defenses against infectious
diseases. It is seated in Stockholm, Sweden. Its mission is to identify, assess and communicate current and emerging threats to human health posed by infectious diseases.
ECDC works in partnership with national health protection bodies across Europe to strengthen and develop continent-wide disease surveillance and early warning systems. By
working with experts throughout Europe, ECDC pools Europes health knowledge to develop authoritative scientific opinions about the risks posed by current and emerging
infectious diseases. The Centre collects, collates, evaluates and disseminates relevant scientific and technical data; provides scientific opinions and scientific and technical
assistance including training; provides timely information to the European Commission, the Member States, Community agencies and international organizations active
within the field of public health; coordinates European networking of organizations operating in the public health; and exchanges information, expertise and best practices, and
facilitates the development and implementation of joint actions. What is does not do directly is laboratory work and research; instead it relies on each member country to carry
out these activities. [This is quite different from the work of the U.S. Centers for Disease Control and Prevention, where research is an integral part of all activities.]
page 510

2005 Founding of the European Centre for Disease Prevention and Control (ECDC)
Human bocavirus (HBoV) was discovered in 2005 by
Tobias Allander and his colleagues at the Karolinska
University Hospital in Stockholm. Since the first report,
the virus has been shown to occur worldwide. The
virus was the first to be identified by a procedure based
on DNase treatment of clinical specimens, random
PCR amplification and cloning, followed by large scale
sequencing and bioinformatic analyses the virus has
not been grown in cell culture or experimental animals
it is only known from its DNA sequence. By its genomic
organization and sequence homology, the virus has
been classified in the family Parvoviridae, subfamily
Parvovirinae, the first member of the genus Bocavirus.
There are now four genotypes of the virus, types 1 to 4.
The fact that HBoV was first detected in nasopharyngeal
aspirates of children with respiratory tract infections
suggested that the virus may be etiologically associated
with such infections. In the first study this hypothesis
was supported, but proving it has been difficult. Most
evidence indicates that the virus is associated with
clinical signs of acute respiratory tract infection, but
because the virus is only detected by PCR and because
the virus persists after primary infection for a much
longer period than other respiratory viruses (in some
cases for months), it cannot be certain from prevalence
data alone just when the virus is the cause of the acute
infection of interest and when it is just a confounding
finding. This is also making it difficult to prove an
etiologic association of the virus with gastroenteritis
and lower respiratory infections. This problem is being
Tobias Allander resolved with serologic studies using as antigen virus-
like particles (VLPs) derived from expressed major virus
capsid protein cloned into a baculovirus vector and
transfected into insect cells.
Allander T, Tammi MT, Eriksson M, Bjerkner A,
Tiveljung-lindell A, Andersson B. Cloning of a human
parvovirus by molecular screening of respiratory tract
samples. Proc Natl Acad Sci USA. 2005;102:12891-12896.
Human bocavirus: Allander T, Jartti T, Gupta S, Niesters HG, Lehtinen
(top) negative contrast electron P, Osterback R, Vuorinen T, Waris M, Bjerkner A,
microscopy, colorized; Tiveljung-Lindell A, van den Hoogen BG, Hyypi T,
(bottom) model reconstruction Ruuskanen O. Human bocavirus and acute wheezing in
of virion from cryo-electron children. Clin Infect Dis. 2007;44:904-910.
microscopic images
page 511
Allander T. Human bocavirus. J Clin Virol. 2008;41:29-33.

2005 Tobias Allander, colleagues discovery of human bocavirus (parvovirus)


Laura H. Kahn Bruce Kaplan Thomas P. Monath

The One Health Initiative is a movement to forge collaborations between physicians, veterinarians and other professionals, all of whom recognize the merit of collaboration
at the interface of medicine, veterinary medicine, public health and related fields, all with the purpose of advancing the understanding, prevention and control of disease. The
concept, first called One Medicine, originated in the writings of Rudolf Virchow and William Osler in the mid-1800s, and was resurrected by Calvin Schwabe in the 1960s,
only to lose visibility by the 1990s. The concept, now called One Health, was again resurrected, this time in 2006 by Laura Kahn, Bruce Kaplan, Thomas Monath and Jack
Woodall. The Initiative now has a website, newsletter, publications, educational activities, and the endorsement of many organizations, institutions and individuals.
Schwabe CW. Veterinary medicine and human health. Baltimore: Williams & Wilkins; 1964.
Kahn LH, Kaplan B, Steele JH. Confronting zoonoses through closer collaboration between medicine and veterinary medicine (as one medicine). Vet Ital. 2007;43:5-19.
Kahn LH, Kaplan B, Monath TP, Steele JH. Teaching one medicine, one health. Am J Med. 2008;121:169-170.
page 512

2006 Laura Kahn, Bruce Kaplan, Thomas Monath, others development of the One Health / One Medicine Initiative
Attendees at an Extended Evolutionary Synthesis Workshop, 2008: From left to right: Sergey Gavrilets, Stuart Newman, David Sloan
Wilson, John Beatty, John Odling-Smee, Michael Purugganan, Greg Wray, David Jablonski, Marc Kirschner, Ers Szathmary, Gnter
Wagner, Werner Callebaut, Eva Jablonka, Gerd Mller, Massimo Pigliucci, Alan Love.
The modern evolutionary synthesis (yellow oval) was a union of ideas from several biological fields which provided a widely accepted account of evolution; the concept
evolved throughout the 1930s-1940s, and added to the concepts originated by Charles Darwin (e.g., common descent, natural selection cyan oval). In this era before the
dawn of molecular biology, it added ideas from population genetics, experimental Mendelian genetics, paleontology, botany and ecology the term was coined by Julian
Huxley in his book, Evolution: The Modern Synthesis (1942). Other major figures in the modern synthesis included Theodosius Dobzhansky, R. A. Fisher, J.B.S. Haldane,
Sewall Wright, E.B. Ford, Ernst Mayr, Bernhard Rensch, Sergei Chetverikov, George Gaylord Simpson, and G. Ledyard Stebbins. The extended evolutionary synthesis
(magenta oval) was elaborated in ~2007 to reflect more recent research, especially derived from the molecular biology revolution (genomics and other -omics), genetic study
of natural populations, phylogenetics, species-level stasis and punctuated evolution, and developmental biology (evo-devo evolution via genetic changes in developmental
processes). It is largely additive, not undoing any of Darwins central concepts or any concepts from the earlier modern
evolutionary synthesis. The new concepts include evolvability, developmental plasticity, phenotypic and
genetic accommodation, punctuated equilibrium, phenotypic innovation, facilitated variation, epigenetic
inheritance, and multi-level selection. The new concept has paid little attention to the viruses, but
phenomena studied widely in virology are part of its substance: recombination, horizontal gene
transfer, genetic drift and shift, etc. Horizontal gene transfer (the non-genealogical transmission
of genetic material from one organism to another) is now known to be an important force
driving the evolution of bacteria and archaea, as well as that of unicellular eukaryotes again,
many evolutionary scientists seem to ignore the viruses and their great role in horizontal
gene transfer (not so much their central role as vectors of gene transfer, but targets of gene
transfer). Overall, the extended evolutionary synthesis has been quite controversial, especially
in trying to integrate its elements into the Darwinian foundation for example, how important
is horizontal gene transfer or recombination in the evolution of vertebrates? Does horizontal
gene transfer challenge the construction of phylogenetic trees of higher organisms? How
does horizontal gene transfer challenge the traditional view of evolution as a gradual process of
variation?

Pigliucci M, Mller GB, editors. Evolutionthe extended synthesis. 2nd ed. Cambridge: MIT Press; 2010.
page 513

2007 Massimo Pigliucci, Gerd Mller, others development of the Extended Evolutionary Synthesis
Two human polyomaviruses, BK virus and JC virus, were discovered
and associated with serious diseases, including polyomavirus
nephropathy and progressive multifocal leukoencephalopathy, more
than four decades ago. Only recently has the list been expanded by the
identification of three new viruses (and a fourth, not yet associated
with disease). Viral genomes cloned from DNA sequences have been
designated KI polyomavirus in work from the lab of Tobias Allander
at the Karolinska University Hospital in Stockholm; WU polyomavirus
in work from the lab of David Wang at Washington University, St.
Louis; and Merkel cell (MC) polyomavirus in work from the lab of
Patrick Moore and Yuan Chang at the University of Pittsburgh School
of Medicine. KI and WU viruses were identified from large-scale
high-throughput screens of respiratory secretions from patients with
respiratory tract infections and MC virus was identified in Merkel cell
carcinomas (MCC) using digital transcriptome subtraction. MC virus
has been implicated in the etiology of Merkel cell carcinoma, a rare
but aggressive form of skin cancer that is increasing in incidence.
Allander T, Andreasson K, Gupta S, Bjerkner A, Bogdanovic G,
Persson MA, Dalianis T, Ramqvist T, Andersson B. Identification of a
third human polyomavirus [KI polyomavirus]. J Virol. 2007;81:4130-
4136.
Gaynor AM, Nissen MD, Whiley DM, Mackay IM, Lambert SB, Wu G,
Brennan DC, Storch GA, Sloots TP, Wang D. Identification of a novel
polyomavirus from patients with acute respiratory tract infections
Tobias Allander David Wang [WU polyomavirus]. PLoS Pathog. 2007;3:e64.
Feng H, Shuda M, Chang Y, Moore PS. Clonal integration of a
polyomavirus in human Merkel cell carcinoma [MC polyomavirus].
Science 2008;319:1096-1100.

Merkel cell carcinoma infiltrating skin tissue,


stained for MC polyomavirus large T protein
Masahiro Shuda, Huichen Feng, Yuan Chang, Patrick Moore page 514

2007 Tobias Allander, David Wang, Yuan Chang, others human polyomaviruses KI, WU, MC (Merkel carcinoma)
Bernard La Scola Didier Raoult
After the discovery of the giant mimivirus in 2007, Bernard La Scola, Didier Raoult and their
colleagues, at the Universit Aix-Marseille, in 2008, discovered an even larger virus infecting the
amoeba Acanthamoeba castellanii, and named it mamavirus (Acanthamoeba polyphaga mimivirus,
APMV). Most remarkably, mamavirus was found to be infected by a smaller virus, which they
named Sputnik by analogy to the term bacteriophage, they coined the term virophage for viruses
infecting viruses. Sputnik virophage virions are icosahedral, 50nm in diameter, and have a circular,
double-stranded DNA genome, 18.3 kb in size, which encodes 21 ORFs, some of which are
derived from the mamavirus, some from the amoeba and many of unknown origin and function.
Sputnik virophage replication occurs only in amoeba co-infected with mamavirus its presence
is detrimental to the replication of mamavirus, causing abortive or abnormal capsid assembly, and
greatly lowered yield of infectious virions. Sputnik has no known homologues with other viruses
and will be the founding member of a new family. In 2011, two more virophages were discovered:
one called mavirus (short for maverick virus), which infects the giant cafeteria roenbergensis virus
of amoeba; the other called Organic Lake virophage, which was found infecting a phycodnavirus in a
phototrophic algae in Organic Lake, a hypersaline lake in Antarctica.
La Scola B, Desnues C, Pagnier I, Robert C, Barrassi L, Fournous G, Merchat M, Suzan-Monti M,
Forterre P, Koonin E, Raoult D. The virophage as a unique parasite of the giant mimivirus [Sputnik]. Top: Sputnik virophage virions and two mamavirus virions at the
Nature. 2008;455:100-104. edge of a cytoplasmic factory where replication/assembly occur.
Fischer MG, Suttle CA. A virophage at the origin of large DNA transposons [mavirus]. Science. Bottom: Sputnik virophage virions within a mamavirus virion.
2011;332:231-234. Thin section electron microscopy. Cryo-electron microscopy
page 515 reconstruction of Sputnik virophage.

2008 Bernard La Scola, Didier Raoult, others discovery of the first virophage Sputnik, infecting mamavirus
Rinderpest, a disease that had devastated cattle herds and their
human keepers across Eurasia and Africa for millennia, was
eradicated globally by a vaccination campaign organized principally
by the U.N. Food and Agriculture Organization (FAO) it joins
smallpox as the only infectious diseases to have been eradicated.
Following World War II, precarious food supplies were threatened
by rinderpest epidemics, leading the newly created FAO to launch a
regional vaccination campaign in Africa. One key to the feasibility
of this was the development in the 1960s by Walter Plowright of a
safe, efficacious attenuated live-virus vaccine. Campaign followed
campaign, each failing as disease incidence declined locally, only
to be followed by a resurgence of disease as interest waned. One
campaign initiated in 1962 came close to freeing Africa of the virus
but, again, as incidence declined countries terminated vaccination
and surveillance programs and by the late 1970s the disease fanned
out from two lingering foci of infection in Mali and Sudan; the same
thing happened in Asia, from Turkey to Bangladesh. One estimate
was that 100 million cattle died during this resurgence. By the
1980s, it was realized that reintroduction of the virus from Asia into
Africa via the live cattle trade was such that only a global campaign
could succeed from this the FAO Global Rinderpest Eradication
Programme (GREP) was initiated in 1993. In 1990, Jeffrey Mariner,
at Tufts University School of Veterinary Medicine, developed a
heat-stable variation of the Plowright vaccine that allowed vaccine to
be backpacked into the remote areas where the virus lingered. The
program met its goal of eradicating the disease by 2004 and from
then until 2010 intensive surveillance was done to assure that there
were no lingering pockets of contagion. The virus was last detected
in 2001 in wild buffalo in Meru National Park in Kenya. Many people
worked in Africa and Asia for all the years of the GREP campaign,
Yoshihiro Ozawa Peter Roeder particularly an international group of veterinarians (e.g., Peter
Roeder, William Taylor, Yoshihiro Ozawa, many others) and as time
went along many African and Asian veterinarians and technicians
played central roles. Over time, surveillance and epidemiological
investigations were augmented by new molecular and serological
tools unavailable in previous campaigns. Molecular analyses
allowed the tracking of outbreak viruses to their source reservoirs.
Sensitive tests for antibodies helped monitor the effectiveness of
vaccination and were crucial for checking for pockets of infection
once vaccination was stopped. The FAO announced in 2010 that
the global eradication effort had succeeded and that all follow-up
fieldwork had ended. Curiously, this grand achievement has received
far less public notice than might be expected; this, in turn, may
dampen chances of further global eradication campaigns against
William Taylor Ricky Ireri animal or human viral diseases.
page 516

2010 FAO Global Rinderpest Eradication Programme global eradication of rinderpest


Wild-type Polio Cases, Globally, 2011
650 cases

The WHO global Polio Eradication Initiative, begun in 1988 and led by the World Health Organization, UNICEF
and The Rotary Foundation, has reduced the number of cases from the hundreds of thousands per year to <1,000
(650 cases in 2011). Despite great difficulties in the end-game, it is hoped that the disease will be eradicated by
2015 it will be the third disease globally eradicated, after smallpox and rinderpest. The most important step
in polio eradication is interruption of endemic transmission by universal infant vaccination using oral vaccine
(OPV; often via national immunization days), supplementary vaccination campaigns where needed, intensive
surveillance of cases of flaccid paralysis, and in some places detection of virus in sewage. The poorest countries
of the world are anxious to complete the eradication program and then eventually to stop vaccination, thereby
avoiding continuing costs this will prove to be a very difficult decision with global political ramifications.
page 517

~2015 WHO Global Polio Eradication Initiative planned global eradication of poliomyelitis
Further Reading Waterson AP, Wilkinson L. An introduction to the history of virology. Cambridge, United Kingdom:
Cambridge University Press; 1978.
Andrewes C. The growth of virus research 1928-1978. Postgrad Med J. 1979;55:73-77.
In a volume on history, it is obvious that a Further Reading list will include some
papers and books that are out-of-date. Such is true here; the following list includes Wilkinson L. The development of the virus concept as reflected in corpora of studies on individual
pathogens. 5. Smallpox and the evolution of ideas on acute (viral) infections. Med Hist. 1979;23:1-28.
contemporary works along with timeless classics that were highly influential in their Witkowski JA. Alexis Carrel and the mysticism of tissue culture. Med Hist. 1979;23:279-296.
day. The latter may create a sense of the context of the time, a sense of what it was like
Enders JF, Robbins FC, Weller TH. Classics in infectious diseases. The cultivation of the poliomyelitis
to be around when great discoveries were made by the scientists who populate this viruses in tissue culture. Rev Infect Dis. 1980;2:493-504.
book: Wilkinson L. Rinderpest and mainstream infectious disease concepts in the eighteenth century. Med Hist.
Reed W, Carroll J, Agramonte A, Lazear JW. The etiology of yellow fever - a preliminary note. Public Health 1984;28:129-150.
Pap Rep. 1900;26:37-53. Murphy FA. The epidemiology of infectious diseases of livestock. Onderstepoort J Vet Res. 1985;52:195-
Rivers TM. Filterable viruses: A critical review. J Bacteriol. 1927;14:217-258. 200.
Rivers TM, editor. Filterable viruses. Baltimore: Williams and Wilkins; 1928. Fenner F, Gibbs A. Portraits of viruses: a history of virology. Basel, Switzerland: Karger; 1988.
Rivers TM. Some general aspects of pathological conditions caused by filterable viruses. Am J Pathol. Hsiung GD. The impact of cell culture sensitivity on rapid viral diagnosis: a historical perspective. Yale J Biol
1928;4:91-124. Med. 1989;62:79-88.
Woodruff AM, Goodpasture EW. The susceptibility of the chorio-allantoic membrane of chick embryos to Grafe A. A history of experimental virology. Vienna, Austria: Springer-Verlag; 1991.
infection with the fowl-pox virus. Am J Pathol. 1931;7:209-222. Monath TP. Yellow fever: Victor, Victoria? Conqueror, conquest? Epidemics and research in the last forty
McKinley EB. A concept of the ultramicroscopic virus diseases and a classification. Science 1932;76:449- years and prospects for the future. Am J Trop Med Hyg. 1991;45:1-43.
454. Kevles DJ. Renato Dulbecco and the new animal virology: medicine, methods, and molecules. J Hist Biol.
Theiler M, Smith HH. The use of yellow fever virus modified by in vitro cultivation for human 1993;26:409-442.
immunization. J Exp Med. 1937;65:787-800. Koprowski H. Yesterday, today and tomorrow in virology. Intervirology 1993;35:9-15.
Ellis EL, Delbrck M. The growth of bacteriophage. J Gen Physiol. 1939;22:365-384. Mahy BWJ, Lvov D, editors. Concepts in virologyFrom Ivanovsky to the present. Chur, Switzerland:
Williams G. Virus Hunters. New York: Knopf; several editions, 1941-1959. Harwood Academic Publishers; 1993.
Fenner F. The pathogenesis of the acute exanthems; an interpretation based on experimental investigations Summers WC. How bacteriophage came to be used by the Phage Group. J Hist Biol. 1993;26:255-267.
with mousepox (infectious ectromelia of mice). Lancet 1948;2:915-920. Johnson RT. The Soriano Award Lecture. Emerging infections of the nervous system. J Neurol Sci.
Watson JD, Crick FH. Molecular structure of nucleic acids; a structure for deoxyribose nucleic acid. Nature 1994;124:3-14.
1953;171:737-738. Shope RE. The discovery of arbovirus diseases. Ann N Y Acad Sci. 1994;740:138-145.
Watson JD, Crick FH. Genetical implications of the structure of deoxyribonucleic acid. Nature Mackenzie JS, Broom AK. Australian X disease, Murray Valley encephalitis and the French connection. Vet
1953;171:964-967. Microbiol. 1995;46:79-90.
Lumsden LL. St. Louis encephalitis in 1933; observations on epidemiological features. Public Health Rep. Fredericks DN, Relman DA. Sequence-based identification of microbial pathogens: a reconsideration of
1958;73:340-353. Kochs postulates. Clin Microbiol Rev. 1996;9:18-33.
Meselson M, Stahl FW. The replication of DNA in Escherichia coli. Proc Natl Acad Sci USA. Kapikian AZ. Overview of viral gastroenteritis. Arch Virol Suppl. 1996;12:7-19.
1958;44:671-682.
Koprowski H, Oldstone MBA. Microbe hunters then and now. Lansing, Michigan: Medi-Ed Press; 1996.
Casals J. Procedures for identification of arthropod-borne viruses. Bull World Health Organ.
1961;24:723-734. Kristensson K, Dastur DK, Manghani DK, Tsiang H, Bentivoglio M. Rabies: interactions between
neurons and viruses. A review of the history of Negri inclusion bodies. Neuropathol Appl Neurobiol.
Coons AH. The beginnings of immunofluorescence. J Immunol. 1961;87:499-503. 1996;22:179-187.
Hirst GK. Development of virology as an independent science. Br Med J. 1962;1:1431-1437. Lambert L, Mulvey T. Ernst Ruska (1906-1988), designer extraordinaire of the electron microscope: a
Kissling RE, Murphy FA, Henderson BE. Marburg virus. Ann N Y Acad Sci. 1970;174:932-945. memoir. Adv Imag Electron Phys. 1996; 95:3-62.
Kalter SS, Heberling RL. Comparative virology of primates. Bacteriol Rev. 1971;35:310-364. Biggs PM. The Leeuwenhoek Lecture, 1997. Mareks disease herpesvirus: oncogenesis and prevention.
Philos Trans R Soc Lond B Biol Sci. 1997;352:1951-1962.
Andrewes C. Fifty years with viruses (Christopher H. Andrewes). Annu Rev Microbiol. 1973;27:1-11.
Nathanson N, Wilesmith J, Griot C. Bovine spongiform encephalopathy (BSE): causes and consequences of
Wilkinson L. The development of the virus concept as reflected in corpora of studies on individual a common source epidemic. Am J Epidemiol. 1997;145:959-969.
pathogens. 1. Beginnings at the turn of the century. Med Hist. 1974;18:211-221.
del Regato JA. James Carroll: a biography. Ann Diagn Pathol. 1998;2:335-349.
Stuart-Harris C. The contribution of virology to contemporary medicine. Br J Prev Med. 1975;29:1-17
Murphy FA. Emerging zoonoses. Emerg Infect Dis. 1998;4:429-435.
Murphy FA. Arenavirus taxonomy: a review. Bull World Health Organ. 1975;52:389-391.
Domingo E. Quasispecies and the implications for virus persistence and escape. Clin Diagn Virol.
Wilkinson L, Waterson AP. The development of the virus concept as reflected in corpora of studies on 1998;10:97-101.
individual pathogens. 2. The agent of fowl plague a model virus. Med Hist. 1975;19:52-72.
Barrett T, Rossiter PB. Rinderpest: the disease and its impact on humans and animals. Adv Virus Res.
Wilkinson L. The development of the virus concept as reflected in corpora of studies on individual 1999;53:89-110.
pathogens lessons of the plant viruses. 3. Tobacco mosaic virus. Med Hist. 1976;20:111-134.
Biel SS, Gelderblom HR. Diagnostic electron microscopy is still a timely and rewarding method. J Clin
Johnson KM, Lange JV, Webb PA, Murphy FA. Isolation and partial characterisation of a new virus causing Virol. 1999;13:105-119.
acute haemorrhagic fever in Zaire [Ebola virus]. Lancet 1977;1:569-571.
Brock T. Milestones in microbiology. Classic papers translated and edited by Thomas Brock. Chapter:
Wilkinson L. The development of the virus concept as reflected in corpora of studies on individual Report of the commission for research on the foot-and-mouth disease, 1898, by Friedrich Loeffler and P.
pathogens. 4. Rabies two millennia of ideas and conjecture on the aetiology of a virus disease. Med Frosch. Washington DC: ASM Press; 1999.
Hist. 1977;21:15-31.
Calisher CH, Horzinek MC, editors. Virology: the first 100 years. Vienna, Austria: Springer-Verlag; 1999.
Diener TO. Viroids and the nature of viroid diseases. Arch Virol Suppl. 1999;15:203-220. Joklik WK. Adventures of a biochemist in virology. J Biol Chem. 2005;280:40385-40397.
Ginsberg HS. The life and times of adenoviruses. Adv Virus Res. 1999;54:1-13. Pourrut X, Kumulungui B, Wittmann T, Moussavou G, Dlicat A, Yaba P, Nkoghe D, Gonzalez JP, Leroy
EM. The natural history of Ebola virus in Africa. Microbes Infect. 2005;7:1005-1014.
Gottesman M. Bacteriophage lambda: the untold story. J Mol Biol. 1999;293:177-180.
Joklik WK, Grossberg SE. How the American Society for Virology was founded. Virology 2006;344:250-257.
Gubler DJ, Meltzer M. Impact of dengue/dengue hemorrhagic fever on the developing world. Adv Virus
Res. 1999;53:35-70. Koonin EV, Senkevich TG, Dolja VV. The ancient virus world and evolution of cells. Biol Direct. 2006;1:29.
Wagner RR. The Journal of Virology: a personal retrospective. J Virol. 1999;73:3515-3519. Levine A, Enquist L. History of virology. In: Knipe DM, Howley PM, editors. Fields Virology, 5th edition.
Philadelphia: Lippincott Williams & Wilkins; 2006.
Murphy FA. The evolution of viruses, the emergence of viral diseases: a synthesis that Martinus Beijerinck
might enjoy. Arch Virol Suppl. 1999;15:73-85. Molineux IJ. Fifty-three years since Hershey and Chase; much ado about pressure but which pressure is it?
Virology 2006;344:221-229.
Fenner F. Adventures with poxviruses of vertebrates. FEMS Microbiol Rev. 2000;24:123-133.
Peters CJ. Emerging infections: lessons from the viral hemorrhagic fevers. Trans Am Clin Climatol Assoc.
Gaudillire J. Rockefeller strategies for scientific medicine: molecular machines, viruses and vaccines. Stud 2006;117:189-196.
Hist Phil Sci Part C 2000;31:491-509.
Weiss RA. The discovery of endogenous retroviruses. Retrovirology 2006;3:67.
Hilleman MR. Vaccines in historic evolution and perspective: a narrative of vaccine discoveries. Vaccine
2000;18:1436-1447. Chany C. Early days in interferon research. Biochimie 2007;89:721-722.
Kapikian AZ. The discovery of the 27-nm Norwalk virus: an historic perspective. J Infect Dis. 2000;181 Childs JE, Richt JA, Mackenzie JS. Introduction: conceptualizing and partitioning the emergence process of
Suppl 2:S295-302. zoonotic viruses from wildlife to humans. Curr Top Microbiol Immunol. 2007;315:1-31.
Kruger DH, Schneck P, Gelderblom HR. Helmut Ruska and the visualisation of viruses. Lancet Greene WC. A history of AIDS: looking back to see ahead. Eur J Immunol. 2007;37 Suppl 1:S94-102.
2000;355:1713-1717.
Klein SL, Calisher CH. Emergence and persistence of hantaviruses. Curr Top Microbiol Immunol.
Oldstone MBA, Levine A. Virology in the next millennium. Cell 2000;100:139-142. 2007;315:217-252.
Epstein MA. Historical background [Epstein-Barr virus]. Philos Trans R Soc Lond B Biol Sci. Nel LH, Rupprecht CE. Emergence of lyssaviruses in the Old World: the case of Africa. Curr Top Microbiol
2001;356:413-420. Immunol. 2007;315:161-193.
Field H, Young P, Yob JM, Mills J, Hall L, Mackenzie J. The natural history of Hendra and Nipah viruses. Norrby E, Prusiner SB. Polio and Nobel prizes: looking back 50 years. Ann Neurol. 2007;61:385-95.
Microbes Infect. 2001;3:307-314.
Norrby E. Yellow fever and Max Theiler: the only Nobel Prize for a virus vaccine. J Exp Med. 2007;204:2779-
Gust ID, Hampson AW, Lavanchy D. Planning for the next pandemic of influenza. Rev Med Virol. 2784.
2001;11:59-70.
Ho M. The history of cytomegalovirus and its diseases. Med Microbiol Immunol. 2008;197:65-73.
Simmonds P. The origin and evolution of hepatitis viruses in humans. J Gen Virol. 2001;82:693-712.
Javier RT, Butel JS. The history of tumor virology. Cancer Res. 2008;68:7693-706.
Taubenberger JK, Reid AH, Janczewski TA, Fanning TG. Integrating historical, clinical and molecular
genetic data in order to explain the origin and virulence of the 1918 Spanish influenza virus. Philos Kaufmann SH. Immunologys foundation: the 100-year anniversary of the Nobel Prize to Paul Ehrlich and
Trans R Soc Lond B Biol Sci. 2001;356:1829-1839. Elie Metchnikoff. Nat Immunol. 2008;9:705-712.

Tennant BC, Gerin JL. The woodchuck model of hepatitis B virus infection. ILAR J. 2001;42:89-102. Staples JE, Monath TP. Yellow fever: 100 years of discovery. JAMA. 2008;300:960-962.

Bausch DG, Ksiazek TG. Viral hemorrhagic fevers including hantavirus pulmonary syndrome in the Thorley-Lawson DA, Allday MJ. The curious case of the tumour virus: 50 years of Burkitts lymphoma. Nat
Americas. Clin Lab Med. 2002;22:981-1020. Rev Microbiol. 2008;6:913-924.

Kitching RP. Future research on foot-and-mouth disease. Rev Sci Tech. 2002;21:885-889. Trel zkul R, Tamay Basaga G. The colloboration of Maurice Nicolle et Adil Mustafa. The discovery of
rinderpest agent. Revue Md Vt. 2008;159:243-246.
Patterson KB, Runge T. Smallpox and the Native American. Am J Med Sci. 2002;323:216-222.
Vasilakis N, Weaver SC. The history and evolution of human dengue emergence. Adv Virus Res.
Sharp PM. Origins of human virus diversity. Cell 2002;108:305-312. 2008;72:1-76.
Brown F. The history of research in foot-and-mouth disease. Virus Res. 2003;91:3-7. Norrby E. Nobel Prizes and the emerging virus concept. Arch Virol. 2008;153:1109-1123.
Campbell A. The future of bacteriophage biology. Nat Rev Genet. 2003;4:471-477. Murphy FA. Emerging zoonoses: the challenge for public health and biodefense. Prev Vet Med.
2008;86:216-223.
Morgan GJ. Historical review: viruses, crystals and geodesic domes. Trends Biochem Sci. 2003;28:86-90.
Bishop R. Discovery of rotavirus: Implications for child health. J Gastroenterol Hepatol.
Novella IS. Contributions of vesicular stomatitis virus to the understanding of RNA virus evolution. Curr 2009;24 Suppl 3:S81-5.
Opin Microbiol. 2003;6:399-405.
Chapman LE. Xenotransplantation, xenogeneic infections, biotechnology, and public health. Mt Sinai J
Reid AH, Taubenberger JK. The origin of the 1918 pandemic influenza virus: a continuing enigma. J Gen Med. 2009;76:435-441.
Virol. 2003;84:2285-2292.
Houghton M. The long and winding road leading to the identification of the hepatitis C virus. J Hepatol.
Barry JM. The site of origin of the 1918 influenza pandemic and its public health implications. J Transl Med. 2009;51:939-948.
2004;2:3.
Katz M. A short history of the long and productive search for the cause of subacute sclerosing
Chua KB. The discovery of Nipah virus: A personal account. Neurology Asia 2004;9:59-63. panencephalitis. Folia Neuropathol. 2009;47:145-148.
Johnson KM. The discovery of hantaan virus: comparative biology and serendipity in a world at war. J Infect Okamoto H. History of discoveries and pathogenicity of TT viruses. Curr Top Microbiol Immunol.
Dis. 2004;190:1708-1710. 2009;331:1-20.
Ligon BL. Monkeypox: a review of the history and emergence in the Western hemisphere. Semin Pediatr Biggs PM. Walter Plowright. 20 July 1923 - 20 February 2010. Biogr Mems Fell R Soc. 2010;56:341-358.
Infect Dis. 2004;15:280-287.
Montagnier L. 25 years after HIV discovery: prospects for cure and vaccine. Virology 2010;397:248-254.
Petri WA Jr. America in the world: 100 years of tropical medicine and hygiene. Am J Trop Med Hyg.
2004;71:2-16. Morens DM, Taubenberger JK. Historical thoughts on influenza viral ecosystems, or behold a pale horse,
dead dogs, failing fowl, and sick swine. Influenza Other Resp Viruses 2010;4:327-337.
Tomori O. Yellow fever: the recurring plague. Crit Rev Clin Lab Sci. 2004;41:391-427.
Sencer DJ. Perspective: swine-origin influenza: 1976 and 2009. Clin Infect Dis. 2011;52 Suppl 1:S4-7.
Chia R, Achilli F, Festing MF, Fisher EM. The origins and uses of mouse outbred stocks. Nat Genet.
2005;37:1181-1186. Murphy FA. In memoriam: Frank John Fenner (1914-2010). Arch Virol. 2011;156:363-367.
Some Major Image Libraries
Of course, an eBook like this would have been impossible to write and illustrate without Google Images (http://www.google.com/imghp) and the main Google search engine
(http://www.google.com/). The work has also been helped very much by the resources of PubMED (http://www.ncbi.nlm.nih.gov/pubmed/), Wikipedia (http://www.wikipedia.
org/) and especially by the staff of the University of Texas Medical Branch Moody Medical Library. Since this is primarily a picture book, public and private image libraries,
archives and databases have been explored exhausively, not only to find images with suitable content, but to find images of the highest quality available without resorting to
expensive stock photo agencies.
1. U.S National Library of Medicine (NLM). History of Medicine Division (HMD). Images from the History of Medicine (IHM) (access to 70,000 images).
http://www.nlm.nih.gov/hmd/ihm/.

2. National Library of Medicine. Images from the History of the Public Health Service. Disease Control and Prevention.
http://www.nlm.nih.gov/exhibition/phs_history/ind.html.

3. Library of Congress (LOC). Prints & Photographs Online Catalog.


http://www.loc.gov/pictures/.

4. Smithsonian Libraries. Digital Collection (20 libraries).


http://siris-libraries.si.edu/.

5. Smithsonian Libraries. Dibner Library of the History of Science and Technology.


http://www.sil.si.edu/DigitalLibrary.cfm.

6. Wellcome Collection. Wellcome Library. History of Medicine Collection. Wellcome Images.


http://images.wellcome.ac.uk/.

7. Institut Pasteur. Scientific Image Bank.


http://www.pasteur.fr/infosci/biblio/english/ressources/imagebank/.

8. Rockefeller Archive Center. Rockefeller Foundation Archives.


http://www.rockarch.org/collections/rf/

9. University of Texas Medical Branch, Moody Medical Library, Truman G. Blocker Jr. History of Medicine Collection. Most of image collection not digitized.
http://ar.utmb.edu/ar/Library/BlockerHistoryofMedicineCollection/tabid/183/Default.aspx.

10. University of Virginia. Claude Moore Health Sciences Library. Philip S. Hench Walter Reed Yellow Fever Collection.
http://yellowfever.lib.virginia.edu/reed/.

11. U.S. National Academy of Sciences. Biographical Memoirs.


http://www.nasonline.org/site/PageServer?pagename=Memoirs_A.

12. National Institutes of Health. Office of History.


http://history.nih.gov/.

13. The Royal Society. Biographical Memoirs of the Fellows of the Royal Society.
http://rsbm.royalsocietypublishing.org/.

14. U.S. Centers for Disease Control and Prevention. Public Health Image Library (PHIL).
http://phil.cdc.gov/phil/home.asp.

15. The Nobel Foundation.


http://nobelprize.org/nobel_prizes/.

16. National Museum of Health and Medicine (formerly part of the Armed Forces Institute of Pathology, now in transition to new facilities, many digital images still available)
http://nmhm.washingtondc.museum/news/exhibits_closing.html.

17. University of Wisconsin. Bock Laboratories. Institute for Molecular Virology. VirusWorld. Jean-Yves Sgro.
http://www.virology.wisc.edu/virusworld/viruslist.php.

18. The Garry Lab. David. M. Sander, Department of Microbiology and Immunology, School of Medicine, Tulane University. The Big Picture book of Viruses and All the Virology on the WWW.
http://www.virology.net/big_virology/BVHomePage.html.
Index Argentine hemorrhagic fever vaccine, development
of 435
Binnig, Gerd 419
biodefense research and response programs 494
Carr, Henri 90, 94
Carrel, Alexis 108, 109
3 poly(A) tail on many mRNAs, discovery of 358 Arita, Isao 398 Biohazards in Biological Research, Asilomar Carroll, James 68, 69, 71
5 cap on messenger RNAs, discovery of 388 Armstrong, Charles 143, 144, 159, 172, 185, 186 Conference 374 Carruthers, Marvin 431
5-hydroxymethylcytosine in some phage DNA, arthropod transmission of disease, first proof of 60 Bishop, David H. L. 422 Carter, Henry Rose 67
discovery of 239 Arvin, Ann 320 Bishop, J. Michael 392 Casals-Ariet, Jordi 306, 350
5-iodo-2-deoxyuridine, the first antiviral drug 289 Ashburn, Percy Moreau 97 Bishop, Ruth F. 376 Caspar, Donald 311
454 pyrosequencing, development of 509 Asibi, yellow fever patient 168 Bittner, John Joseph 153 catalytic properties of RNA, discovery of ribozymes
Abbe, Ernst Karl 25 Asnis, Deborah 492 BK virus (kidney, bladder disease), discovery of 367 416
Abelson, Herbert T. 231 association of a specific pathogen with a specific Blanden, Robert 296 categorization of viruses based on replicative path
Acanthamoeba polyphaga mimivirus 503 disease 40 bluetongue viruses, discovery of 83 to mRNA 365
Achong, Bert Geoffrey 323, 369 strm, Karl-Erik 278 Blumberg, Baruch 327, 408 Cech, Thomas R. 416
achromatic lenses, invention of 25 astroviruses, discovery of 387 Boophilus annulatus 60 cell as unit of structure of all organisms 27
acquired immunodeficiency syndrome (AIDS) 417 Atchison, Wayne 329 Bordet, Jules 49 cell culture, development of 108, 110
acyclovir, antiviral drug, development of 363 atomic structure of viruses, visualization of 407 Borna disease virus, discovery of 140 cell culture, first cultivation of virus in 111
adeno-associated viruses, discovery of 329 Aujeszky, Aladr 80 Borrel, Amde 81 cell culture methodology for polio, measles viruses,
adenoviruses, discovery of human 240 Australia antigen, hepatitis B virus, discovery of Borst, Piet 377 development of 226
adenovirus structural / functional elements, 327 Boshell-Manrique, Jorge 161 cellular immune system recognition of virus-
discovery of 330 autoclave, invention of 52 Bouteille, Michel 326 infected cells 381
Adil Bey, Mustafa 82 Autonomous Pathogen Detection System 504 bovine rotavirus, discovery of 351 cellular (microscopic) pathology, father of 35
adjuvants for viral vaccines 122 Avery, Oswald Theodore 207 bovine spongiform encephalopathy (BSE), discovery Centanni, Eugenio 79
Aedes aegypti 48 avian infectious bronchitis virus, discovery of 176 in United Kingdom 441 Centers for Disease Control and Prevention 304,
Aedes aegypti, vector-borne disease control 187 avian leukemia viruses, discovery of 99 bovine torovirus, discovery of 428 340
African horse sickness viruses, discovery of 75 AZT, first anti-HIV drug, development of 447 bovine virus diarrhea virus, discovery of 271 cervical carcinoma, papillomaviruses, discovery of
African swine fever virus, discovery of 123 Babesia bigemina 60 Boyer, Herbert W. 373, 412 association 384
agarose gel electrophoresis, development of 377 bacteria, discovery of 11 Bradley, Daniel W. 458 Chaille, Stanford Emerson 46
Agramonte, Aristides 68, 72 bacteriophages, discovery of 115 Breese Hall, Caroline 224 Chalfie, Martin 318
Ahmed, Rafi 296 bacteriophage , discovery of 232 Breinl, Anton 120 Chamberland, Charles 50, 51, 52
AIDS 417 Baer, George Martin 370 Brenner, Sydney 282, 302, 374 Chang, Yuan 474, 514
Aitken, Thomas H. G. 321 Balayan, Mikhail Surenovich 429 Brent, Leslie Baruch 245 Chanock, Robert Merritt 258, 263
Aleutian disease of mink virus, discovery of 265 Baltimore, David 355, 356, 365, 418 Brin, Sergey 489 Chargaff, Erwin 228
Allander, Tobias 511, 514 Bang, Frederik B. 243 Brock, Thomas D. 457 Chargaff s Rules, A+T=C+G 228
Almeida, June 328 Bang, Oluf 99 Broder, Samuel 447 Chase, Martha Cowles 234
Alpers, Michael 337 Barin, Francis 439 Broderson, Roger 308 Chelle, Paul-Louis 171
Alter, Harvey J. 327 Barrera Oro, Julio G. 435 Brown, Patrick O. 459 chemotherapy, founding of 49
Altman, Sidney 416 Barr-Sinoussi, Francoise 430 Bryans, John T. 270 Chermann, Jean-Claude 430
American Committee on Arthropod-Borne Viruses, Barr, Yvonne 323 Buchmeier, Michael 296 Chevalier, Charles 26
founding of 290 Bauer, Johannes 137 Buckley, Sonja 350 chikungunya virus, discovery of 262
American Society for Virology, founding of 422, Bawden, Frederick Charles 170 budding, release of virus at the cell surface, childbed fever 31
423 Beard, Joseph Willis 106, 156, 173 discovery of 243 Childs, Jamie E. 472
Amici, Giovanni Battista 25 Beards, Graham 428 Buist, John Brown 54 Chisari, Frank V. 508
Anderson, Charles R. 321 Beasley, R. Palmer 408 Bunyamwera virus, discovery of 216 cholera epidemic, London 28
Anderson, John F. 104 Beaudette, Fred Robert 176 Burke, Alexander 189 Choo, Qui-Lim 458
Anderson, Norman G. 314 Beckman, Arnold O. 164, 219 Burkitt, Denis Parsons 277 Chow, Marie 407
Andrewes, Christopher Howard 148, 155 Beijerinck, Martinus Willem 63 Burkitts lymphoma, description of 277 Christofferoni, J. 139
anelloviruses, discovery of 486 Bellini, William J. 491 Burmester, Ben Roy 341 chromosome, nature and role of 103
An Introduction to the History of Virology, Benfield, David A. 464 Burnet, Frank Macfarlane 150, 218, 266 chronic wasting disease of deer and elk, discovery
publication of book 406 Benzer, Seymour 256, 374 Butel, Janet S. 362 as prion disease 415
Anslow, Ralph Owen 294 Bergh, ivind 461 B virus, discovery of 154 Chua, Kaw Bing 491
anthrax 40, 44 Berg, Paul 256, 374 Cairns, John 303 Chumakov, Mikhail Petrovich 178, 212
anthrax bioterrorism events 494, 504 Berners-Lee, Tim 465 Calisher, Charles H. 306 Churchill, Anthony 341
antibody molecules, discovery of structure 285 Bernhard, Wilhelm 246 Callaway, Edward M. 483 circoviruses, discovery of 385
antigen presentation, discovery of dendritic cells Berson, Solomon A. 287 Campbell, Carlos (Kent) 350 Ciuffo, Giuseppe 98
378 Beutler, Bruce A. 482 canine distemper virus, discovery of 94 Clark, H. Fred 353, 452
Appel, Max J. G. 404 Bhatt, Pravin N. 269 canine parvovirus, discovery and characterization classical swine fever virus, discovery of 84
Arber, Werner 348 BHK-21 cell line, development of 301 of 404 classification of organisms, system for 13
arboviruses, antigenic classification of 306 Biebricher, Christoph 454 Capecchi, Mario R. 448 classification of viruses based upon virion
Archaea, discovery of viruses of 383 Biggs, Hermann Michael 28, 58 carbolic acid, disinfection 31 characteristics 310
Archiv fr die gesamte Virusforschung (Archives of Biggs, Peter M. 341 Carey, Donald E. 368 Claverie, Jean-Michel 503
Virology), first international virology journal 184 Billingham, Rupert E. 245 Carmichael, Leland E. (Skip) 404 Cleland, John Burton 120
clonal selection as the central mechanism in dengue hemorrhagic fever and shock syndrome electronic pH meter, invention of 164 Fite, George L. 160
immunity, discovery of 266 401 electron micrographs of viruses, first 183 Flewett, Thomas 376
Cohen, Seymour Stanley 239 dengue viruses, discovery of 97 electron microscope, invention of 157, 158 Flexner, Simon 78
Cohen, Stanley Norman 373 Department of Filterable Viruses, Johns Hopkins electrophoresis, development of 89 Florio, Lloyd 217
Cohn, Zanvil A. 378 University 133 Eletr, Sam H. 431 Fodor, Stephen P. A. 459
Coimbra, Terezinha Lisieux Moraes 476 DeRisi, Joseph L. 497 Elford, William Joseph 148 Folks, Thomas 369
Cold Spring Harbor Symposium - Basic de Schweinitz, Emil Alexander 84 Elion, Gertrude Belle 363 foot-and-mouth disease epidemic, United Kingdom
Mechanisms in Animal Virus Biology 309 Desrosiers, Ronald C. 438 Ellerman, Vilhelm 99 495
Cole, Gerald 296 de Vries, Hugo Marie 76 Elliott, Luanne H. 472 foot-and-mouth disease vaccines, development of
Collinge, John 479 dHerelle, Flix 115 Ellis, Emory Leon 188 128
Collins, James E. 464 Diener, Theodore Otto 366 embryonating hens eggs, development as host for foot-and-mouth disease virus, discovery of 64
Colorado tick fever virus, discovery of 217 Dimitrov, Dimiter S. 379 viruses 149, 150 formaldehyde to inactivate viruses for vaccines 122
common descent, concept of 36 Dimock, William W. 162 Encyclopedia of Virology, publication of book 473 Formenty, Pierre 500, 505
comparative pathology, father of 35 Dinter, Zvonimir 257 Enders, John Franklin 110, 226, 319 Forterre, Patrick 383
compound microscope, invention of 9 disinfection, methods of 31 Engvall, Eva 364 fowl plague virus (avian influenza virus), discovery
confocal microscope, invention of 267 di Vestea, Alfonso 87, 88 Enquist, Lynn W. 483, 409 of 79
contagium vivum fluidum 63 Dixon, Frank 265, 295 enzyme-linked immunoassays (EIAs, ELISAs), fowl plague virus is an influenza virus, discovery
Convergence Model, factors in viral emergence 470 DNA cloning in phage 409 development of 364 that 257
Conzelmann, Karl-Klaus 475 DNA-dependent RNA polymerase, discovery in epidemiology, founding of 28 Fracastoro, Girolamo 8
Corey, Lawrence 363 virus 339 Epstein-Barr virus, association with Burkitts Fraenkel-Conrat, Heinz 255, 260
Cosgrove, Albert S. 313 DNA fingerprinting, development of 440 lymphoma 323 Francis, Donald 417
Cosgrove, B. P. 387 DNA, identified as the material of inheritance 207, Epstein-Barr virus, association with infectious Francis, Thomas Jr. 211, 249
Cossart, Yvonne Edna 386 234 mononucleosis 344 Franklin, Rosalind Elise 242
Coto, Celia 276 DNA of T2 phage, discovery of its nature 303 Epstein, Michael Anthony 323, 369 Frazer, Ian 498
Cotton, William 129 DNA sequencing, development of automated 445 equine abortion virus, discovery of 162 Fredricks, David N. 480
Courtois, Ghislain F. G. J. 212 DNA structure, discovery of 241, 242 equine arteritis virus, discovery of 270 French, Eric Lancelot 120
Cowdry, Edmund Vincent 55 DNA synthesis technology, development of equine herpesviruses, discovery of 162 French, George 435
cowpox virus 16 commercial 431 equine infectious anemia virus, discovery of 90 Frenkel, Herman 128
Cox, Herald Rea 150, 280 Doane, Frances 224 equine rhinopneumonitis virus, discovery of 162 Frenkel, Nitza 463
Cox, Nancy 490 Dobzhansky, Theodosius 175 erythema infectiosum, fifth disease 386 Friend, Charlotte 231
Coxsackieviruses, discovery of 222 Dochez, Alphonse Raymond 119 Essex, Max 438, 439 Frosch, Paul 64
Craig, Charles Franklin 97 Doermann, Augustus 179 estimating 50% endpoints, development of methods Gajdusek, D. Carleton 337
Creutzfeldt, Hans Gerhard 124 Doerr, Robert 102, 182, 184 for 180 Gallo, Robert C. 413
Creutzfeldt-Jakob disease, description of 124 Doherty, Peter C. 381 European Centre for Disease Prevention and Galtier, Pierre-Victor 19
Crick, Francis 241, 302 Doll, Elvis Roger 162, 270 Control (ECDC) 510 Garcia-Sastre, Adolfo 506
Crimean-Congo hemorrhagic fever virus, discovery Dollond, John 25 Evans, Alfred S. 394 Gardner, Phillip 224
of 212 Dorset, Marion 84 Evans, Martin J. 448 Gardner, Sylvia D. 367
Cuill, Jean 171 double-stranded viral RNA, discovery of 317 evolutionary science, father of 36 Garnham, Percy Cyril 151
Curran, James 417 Dowdle, Walter Reed 315, 414 experimental pathology, founding of 19 Gay, Frederick Parker 154
Curtice, Cooper 60, 61 Downs, Wilbur George 290, 321 Extended Evolutionary Synthesis, development of Gayle, Helene 417
cytology, viral disease 55 Doyle, T. 130 513 Gelderblom, Hans 385
Dalldorf, Gilbert 222 dsRNA-dependent RNA polymerase, discovery of extrinsic incubation period, mosquito 67 GenBank, development of 426
Dalton, Albert J. 246 343 Farr, William 28 gene, definition of 256
Danna, Kathleen J. 348, 380 Dubovi, Edward J. 446 Feinstone, Stephen M. 375 Genentech, Inc. 412
Darnell, James E. Jr. 358 Dulbecco, Renato 238, 248 Feldman, Joseph 295 genetically manipulated (knockout) mice,
Darwin, Charles Robert 36, 37 Dunkin, George 94 feline immunodeficiency virus, discovery of 444 development of 448
Daubney, Robert 151 Eagle, Harry 110 feline leukemia virus, discovery of 324 genetic code, deciphering of 305
Davaine, Casimir 40 Earle, Wilton R. 192 feline panleukopenia virus, discovery of 139 genetic control of enzyme and virus synthesis,
Davenport, Frederick 211 eastern equine encephalitis virus, discovery of 165 Feng, Huichen 514 discovery of mechanisms 298, 299
Davidson, Geoffrey 376 Eaton, Bryan 491 Fenner, Frank 221, 335, 354, 398, 446 genetic engineering, development of 373
DDT, discovery of the insecticidal qualities and use Ebola and Marburg viruses, reservoir host fruit ferret model for influenza 155 genetic recombination in bacteria, discovery of 213
for mosquito control 187 bats 505 Field, Anne 367, 386 genetic recombination in viruses, discovery of 215
Death in a Sailors Suit, Yellow Fever in New York Ebola disease in endangered great apes 500 Fields, Bernard Nathan 296, 436 Genetics and the Origin of Species, publication of
168 Ebola virus, discovery of 390 Fields Virology, publication of book 436 book 175
De Cock, Kevin 417 eclipse period in virus replication, discovery of 179 Fiers, Walter 396 genetics, Gregor Mendel, father of 41
de Hevesy, George 107 Eddy, Bernice 274 fifth disease, erythema infectiosum 386 genetics, rediscovery of Mendels work 76
de la Rivire, Ren Dujarric 118 Eddy, Gerald 435 filariasis 45 Gerin, John L. 400
de la Torre, Juan 454 Edelman, Gerald Maurice 285 Filterable Viruses, first major virology book 133 germ theory, father of 8, 29
Delbrck, Max Ludwig Henning 188, 210, 215 Ehrlich, Paul 49 Finlay, Carlos Juan 48, 68 Gershon, Anne 320
dendritic cells, discovery of role in immunity 378 Eigen, Manfred 454 Fire, Andrew Z. 488 Gessain, Antoine 507
Gey, George Otto 230 Helenius, Ari 379 Horne, Robert W. 282, 283, 310 immune enhancement, discovery of 401
Gibbs, Adrian 454 Hellman, Alfred 374 Horsfall, Frank Lappin Jr. 134, 223 immunofluorescence in viral pathogenesis research,
Gibbs, Clarence Joseph 337 hematology, founding of 49 Horstman, Dorothy 422 development of 229
Gibbs, E. Paul J. 446 hemorrhagic fever with renal syndrome 403 Horzinek, Marian C. 428, 446 immunological tolerance, discovery of 245
Gierer, Alfred 260 Henderson, Donald A. 398 Houghton, Michael 458 immunology, founding of 49
Gilbert, Walter 334, 399 Henderson, William 30 Howitt, Beatrice 145 immunopathology in viral diseases, discovery of
Gillespie, James R. 271 Hendra virus, discovery of 477 Howley, Peter M. 436 295
Ginsberg, Harry S. 240, 422 Henle, Friedrich Gustav Jakob 29 h-ras, cloning of first tumor-derived oncogene 421 inclusion bodies 30, 54, 55
global eradication of smallpox 398 Henle, Gertrude S. 344 Hsiung, Gueh-djen (Edith) 224 infectious bovine rhinotracheitis virus, discovery
Goad, Walter 426 Henle-Koch postulates revisited 177 Huang, Alice S. 355 of 261
Goldberger, Joseph 104, 105 Henle-Koch postulates revisited again 394 Hudson, Charles B. 176 infectious bursal disease virus, discovery of 313
Golde, David 413 Henle-Loeffler-Koch Postulates 29 Hudson, John 151 infectious canine hepatitis virus, discovery of 141
Goldsmith, Cynthia S. 472 Henle, Werner 344 Hudson, Noel 137 infectious disease sciences, Louis Pasteur, father
Goldwasser, Robert 275 Henson, James B. 265 Huebner, Robert J. 240, 349 of 33
Gomatos, Peter J. 317 hepatitis A and B, separation of two diseases 209 Hughes, James M. 414 influenza pandemic of 1918-1919 117
Gonzalez, Jean-Paul 500 hepatitis A virus, discovery of 375 Hughes, Thomas 189 influenza virus, 1918 virus sequenced /
Gonzlez-Scarano, Francisco 296 hepatitis B virus, discovery of 327 human adenoviruses, discovery of 240 reconstructed 506
Goodpasture, Ernest W. 149, 163 hepatitis B virus infection, association with human astroviruses, discovery of 387 influenza virus, discovery of 118, 119
Google, development of 489 hepatocellular carcinoma 408 human bocavirus, discovery of 511 influenza viruses, discovery of the link between
Gorgas, William Crawford 91 hepatitis C virus, discovery of 458 human coronaviruses, discovery of 328 avian and human viruses 316
Gorham, John R. 265 hepatitis C virus, in vitro replication of 508 human cytomegalovirus, discovery of 264 influenza virus hemagglutinin, determination of
Gottschalk, Alfred 218 hepatitis delta virus, discovery of 400 human diploid cell strains (WI-1 through WI-44), atomic structure of 420
Gowans, James Learmonth 279 hepatitis E virus, discovery of 429 establishment of 333 influenza virus, highly pathogenic H5N1 variant,
graded collodion membranes, to determine virion hepatocellular carcinoma, hepatitis B virus Human Genome Project 462, 501 discovery of 485
size 148 infection 408 human herpesvirus 6B (exanthem subitum), influenza virus, molecular basis for virulence 490
Granoff, Allan 473 herpes simplex virus 1, discovery of 121 discovery of 449 influenza virus, proof of viral etiology 155
Grassi, Giovanni Battista 45 herpes simplex viruses, differentiation of HSV 1 human herpesvirus 7 (exanthem subitum), innate immunity 482
Graunt, John 28 and 2 315 discovery of 463 Institute of Medicine of the U.S. National Academy
green fluorescent protein, discovery and herpes simplex virus, molecular biology and human herpesvirus 8 (Kaposi sarcoma herpesvirus), of Sciences, founding of 352
development of 318 replication, discovery of 331 discovery of 474 Institut Pasteur, founding of 57
Green, Robert Gladding 141 Hershey, Alfred Day 234 human immunodeficiency virus 1 (HIV1), discovery interferons, discovery of 268
Griffin, Diane 296 Hess, Alfred Fabian 113 of 430 internal ribosomal entry sites (IRES) in virus
Griffith, Fred 135 Hierholzer, John C. 308 human immunodeficiency virus 2 (HIV2), discovery mRNA, discovery of 450
Gross, Ludwik 231 high-containment virology laboratories, of 439 International Committee on Taxonomy of Viruses,
Guanarito virus (Venezuelan hemorrhagic fever), development of 340 human metapneumovirus, discovery of 493 founding of 335, 336
discovery of 468 highly active antiretroviral therapy (HAART), human papillomaviruses and cervical carcinoma, International Congresses for Virology 345, 346, 347
HAART (highly active antiretroviral therapy), development of 481 discovery of 384 International Human Genome Project 501
development of 481 Hilleman, Maurice 292, 320, 424 human papillomaviruses, discovery of 98 International Human Genome Sequencing
Haase, Ashley 296 Hippocrates 6 human parainfluenza viruses, discovery of 263 Consortium 462
Haddow, Alexander John 216 Hiro, Y. 113 human polyomaviruses KI, WU, MC, discovery of Ireri, Ricky 516
Hadlow, William J. 337 Hirsch, Martin S. 481 514 Ironside, James 479
Hallauer, Curt 182, 184 Hirst, George K. 211, 253 human rhinoviruses, discovery of 259 Isaacs, Alick 268
Halonen, Pekka Eljas 224, 360 His, Wilhelm 26 human rotaviruses, discovery of 376 Ishida, Nakao 244
Halstead, Scott B. 401 Hitchings, George H. 363 human rotavirus vaccines, development of 452 isopycnic density gradient ultracentrifugation,
Hamburg, Margaret 470 HIV antibody tests (EIAs), development of 437 human T-lymphotropic viruses 1 & 2, discovery invention of 272
Hammon, William McDowall 198 Ho, David 481 of 413 Israeli, Clara 111
Handbuch der Virusforschung, publication of book Hoerlein, A. B. 271 human T lymphotropic viruses 3 and 4, discovery Ivanovsky, Dmitry 62
182 Hoffmann, Jules A. 482 of 507 Jackson, Dale A. 372
Hantaan virus, discovery of 403 hog cholera virus, discovery of 84 Hunter, John 19 Jackson Memorial Laboratory 153
Haring, Clarence Melvin 145 Hogle, James M. 407 Hunt, Ronald 439 Jacob, Franois 298
Harpers Weekly 47 Holden, Margaret 154 Hurlbut, Herbert 254 Jaffe, Harold 417
Harrison, Ross Granville 96 Holland, John 454 Hwang, Lu-yu 408 Jakob, Alfons Maria 124
Harrison, Steven C. 407 Holley, Robert W. 305 Hyatt, Alex 477 Jansen, Sacharias 9
Haselkorn, Robert 422 Holloway, Ann 319 hygiene, development of methods 31 Japanese encephalitis virus, discovery of 166
Hayashi, Michitomo 166 Holmes, Ian H. 376 icosahedral structure of viruses, discovery of Jarrett, William F. H. 324
Hayflick, Leonard 333 Holmes, Kathryn 296 principles of 311 JC virus, progressive multifocal
HBsAg produced in yeast, first recombinant human Holmes, Sr., Oliver Wendell 31 IL-2, discovery/characterization of first cytokine leukoencephalopathy, discovery of 278
vaccine 424 Hood, Leroy E. 445 332 Jeffreys, Alec J. 440
Heberling, Richard L. 361 Hooke, Robert 10 immune complexes in viral diseases, discovery of Jenner, Edward 16, 17, 18
HeLa cell line, establishment of 230 Hooper, Peter 477 295 Jerne, Niels Kaj 266
Jesty, Benjamin 15 Kraneveld, F. 130 Lipman, David 484 McCordock, Howard A. 160
Johnson, Claud 163 Krugman, Saul 209 Lister, Joseph 31 McFadyean, John 75
Johnson, Karl M. 325, 340, 390, 403 Ksiazek, Thomas G. 472, 491, 502 Lister, Joseph Jackson 25, 31 McIntosh, Kenneth 224
Johnson, Richard T. 296 Kubes, Vladimir 181 Little, Clarence Cook 153 McKercher, Delbert Grant 261
Joklik, Wolfgang Karl 422 Kuo, George 458 Loeffler, Friedrich A. J. 64, 65 McNamara, Tracy 492
Journal of General Virology, publication of journal Kurata, Takeshi 449 Loeffler-Koch Postulates 29 measles vaccine, development of 319
253 kuru prion, transmission to non-human primates Lopez, Carlos 449 measles virus, discovery of 104
Journal of Virology, publication of journal 253 337 Lwenstein, A. 121 Mebus, Charles A. (Chuck) 351
Jubil de Pasteur a la Sorbonne 34 Kuypers, Hans H. G. 483 Luciw, Paul 369 Medawar, Peter Brian 245, 279
Jung, Rudolf 26 Kyasanur Forest disease virus, discovery of 269 Lumsden, W.H.R. (William) 262 Medical Virology, publication of book 354
Junin virus (Argentine hemorrhagic fever), Lacaillade, Charles Jr. 165 Luria, Salvador Edward 210 medicine, father of modern 6, 43
discovery of 276 lac operon of E. coli 334 Lwoff, Andr 227, 298, 310 Meiklejohn, Gordon 211
Kahn, Laura H. 512 La Crosse virus, discovery of 294 lymphocyte recirculation, discovery of 279 Mello, Craig C. 488
Kalfayan, Bernard 294 lactate dehydrogenase-elevating virus, discovery lymphocytic choriomeningitis (LCM) virus, Melnick, Joseph L. 329, 345, 347
Kalter, Seymour S. (Sy) 361 of 293 discovery of 172 Mendel, Gregor Johann 41
Kalyanaraman, Vaniambadi S. 413 Laemmli, Ulrich K. 342 lymphocytic choriomeningitis (LCM) virus Meredith, Courtney D. 353
Kanki, Phyllis Jean 439 Lagos bat virus 353 immunopathology 295 Merigan, Thomas C. 481
Kapikian, Albert Z. 371, 375, 376, 452 Laidlaw, Patrick Playfair 94 lysogeny, induction, discovery of 227 Merkel cell carcinoma (MC polyomavirus),
Kaplan, Bruce 512 Laigret, Jean 138 lyssaviruses, discovery of 353 discovery of 514
Kaposi sarcoma 417 lambda bacteriophage, discovery of 232 Maalin, Ali Maow 398 Merrifield, Robert Bruce 297
Kaposi sarcoma herpesvirus (HHV-8), discovery Lambert, Robert A. 111 Maass, Clara Louise 74 Merrill, Malcolm H. 165
of 474 Lam Sai Kit, Kenneth 491 MacCallum, Fred O. 209 Meselson, Matthew 272, 273
Krber, G. 180 Lancefield, Rebecca 136 Machupo virus (Bolivian hemorrhagic fever), Mesopotamian medicine 7
Kasahara, Shiro 166 Lanciotti, Robert S. 492 discovery of 325 Metchnikoff, Elie 49
Kasakura, Shinpei 332 Landsteiner, Karl 100, 101 MacKenzie, Ronald 325 Mettler Instruments AG, single-pan analytical
Kates, Joseph R. 339, 358 Lane, David P. 410 Maclachlan, N. James 446 balance, development of 220
Katz, Samuel 319 Lange, James V. 390 MacLeod, Colin Munro 207 Meyer, Harry Jr. 320
Kawakita, Yosio 307 La Scola, Bernard 503, 515 Macpherson, Ian 301 Meyer, Karl Friedrich 145, 146
Kawaoka, Yoshihiro 490 Lasher, Hiram N. 313 Madeley, C. Richard (Dick) 387 microarray technology, development of 459
Kelly, Thomas 348 Lassa virus (Lassa fever), discovery of 350 Madin, Stewart Harvey 261 microarray technology for identification of viruses
Kemp, Graham E. 353 latent period, discovery of 248 Mahieux, Renaud 507 497
Kendrew, John Cowdery 233 Laveran, Charles Louis 45 Mahy, Brian W. J. 473 microbead-based virus detection technology,
Khorana, Har Gobind 305 Laver, William Graeme 316 Maitland, Hugh Bethune 110 development of 487
Kilbourne, Edwin 211 Layton, Marcelle 492 Maitland, Mary Cowan 110 microbiology, co-founder of 44
Kilham, Lawrence 281 Lazear, Jesse William 68, 72 Maizel, Jacob V. Jr. 342 microbiology, Louis Pasteur, father of 33
Kim, Jungsuh 467 L cell line, L929 cloned cell line, development of Maiztegui, Julio I. 435 Micrographia, publication of book 10
Kinyoun, Joseph J. 56 192 malaria, etiologic agent 45 microorganism, first depiction of 10
Kissling, Robert E. 275, 338 Leader, Robert 265 Manson, Patrick 45 microscope, simple (single lens), invention of 11
Kit, Saul 442 Lebailly, Charles 118 Maramorosch, Karl 237 microscopes, improvements in design 25
Kjeldgaard, Niels 227 Lederberg, Esther Miriam Zimmer 232 Marburg virus, discovery of 338 microtiter system for virologic and serologic assays,
Kleinschmidt, Albrecht Karl 284 Lederberg, Joshua 213, 236, 470 Mareks disease virus, discovery of 341 invention of 252
Klenk, Ernst 218 Lee, Ho Wang 403 Marine Hospital Service 23 microtome, invention of 26
Klug, Aaron 311 Lee, K. M. 271 marine mammal morbilliviruses, discovery of 453 Miescher, Johan-Friedrich 42
Knipe, David M. 436 Lee, Pyung Woo 403 marine viral metagenomics 461 Miller, Jacques 300
knockout (genetically manipulated) mice, Lemon, Stanley M. 508 Marion, Andre F. 431 Miller, Mabel 217
development of 448 Lennette, Edwin H. 136 Marshel, James H. 483 Milovanovic, Milan 319
Knoll, Max 157, 169 Lpine, Pierre 249 Martini, Gustav 338 Milstein, Csar 382
Koch, Heinrich Hermann Robert 44, 65 Lerner, Richard 295 Materia Medica 7 mimivirus, discovery of 503
Kochs Postulates 29, 44 Leroy, Eric M. 500, 505 Mathis, Constant 138 Mims, Cedric 229, 296
Kochs Postulates, revisited 177 Leslies Illustrated Newspaper 47, 168 Matsumoto, Seiichi 322 Minot, Charles Sedgwick 26
Kochs Postulates, revisited again 394 Letvin, Norman L. 438 Matteucci, Mark 431 Minsky, Marvin Lee 267
Kochs Postulates, revisted yet again (sequence- Levaditi, Constantin 101 Matthaei, J. Heinrich 305 Mitus, Anna 319
based criteria) 480 Levine, Arnold J. 410 Matthews, Richard Ellis Ford 335 Miyamoto, Kaneatsu 322
Khler, Georges 382 Levkovich, Elizabeth N. 178 Maxam, Allan 399 Mizutani, Satoshi 356
Koonin, Eugene 454 Levy, Jay 417 Mayer, Adolf Eduard 53 MMWR 304, 417
Koprowski, Hilary 280 Lincoln, Abraham 39 Mayr, Ernst Walter 190 modern evolutionary synthesis, developmnet of
Kornberg, Arthur 288 Lindbergh, Charles 109 McAuslan, Brian R. 339 190, 513
Kossel, Ludwig Karl Martin Leonhard Albrecht 42 Lindenmann, Jean 268 McCarthy, Kevin 319 Mogabgab, William J. 259
Kozloff, Lloyd 253 Linnaeus, Carolus 13 McCarty, Maclyn 207 molluscum contagiosum virus 30
Kraft, Lizbeth M. 308 Lipkin, W. Ian 492, 497 McClintock, Barbara 191 Moloney, John B. 231
Monath, Thomas P. 350, 512 New York City Department of Health, founding Patrick Playfair Laidlaw 155 Program to Monitor Emerging Diseases (ProMED),
monoclonal antibodies, development of 382 of 24 Pauling, Linus Carl 233 development of 471
Monod, Jacques 298 New York City Department of Health microbiology Paul, John Rodman 189 protein structure, X-ray crystallography, discovery
Montagnier, Luc 430 laboratory, founding of 58 Payne, James 341 of 233
Montagu, Lady Mary Wortley 12 Nichol, Stuart T. 472, 505 Pedersen, Niels C. 444 Pruden, Mitchell 28
Montgomery, Robert Eustace 116, 123 Nicolau, Stefan S. 140 Peebles, Thomas Chalmers 319 Prusiner, Stanley B. 425
Moore, Patrick S. 474, 514 Nicolle, Charles 118 Peiris, Malik 502 Prusoff, William H. 289
Morbidity and Mortality Weekly Report, Nicolle, Maurice 82, 95 Pellett, Philip E. 449 pseudorabies vaccine, development of 442
publication of 304 Nipah virus, discovery of 491 Pelon, William 259 pseudorabies virus 80
Morgan, Councilman 379 Nirenberg, Marshall W. 305 Penhoet, Edward E. 424 Ptashne, Mark 334
Morgan, Thomas Hunt 103 Nishizawa, Tsutomu 486 Pereira, Helio 330 PubMed (NCBI), development of 484
Morris, J. Anthony 258 Nisonoff, Alfred 285 Perlmann, Peter 364 puerperal fever 31
Morse, Stephen S. 470, 471 Nobel Prizes pertinent to virology, publication of Perutz, Max Ferdinand 233 Purcell, Robert H. 375
Moss, Bernard 388, 433 papers and book 499 Peters, C.J. 472 quasispecies, development of concept of 454, 456
mouse, development for virus research 143, 144 nonhuman primate virology, development of 361 Phage Course at Cold Spring Harbor Laboratory, rabbit (Shope) papillomavirus, discovery of 156
mouse mammary tumor virus, discovery of 153 noroviruses, discovery of 371 founding of 210 rabbit (Shope) papillomavirus, progression to
Muckenfuss, Ralph S. 159 Norrby, Erling Carl Jacob 330, 499 pH meter, invention of 164 carcinoma 173
Muench, Hugo 180 Norwalk virus, discovery of 371 phocine distemper virus, discovery of 453 rabies 7, 85
Mugrage, Edward 217 Nothing in Biology Makes Sense Except in the Light phylogeny based on mRNA gene sequences 402 rabies immunofluorescence diagnostic method,
Mller, Gerd B. 513 of Evolution 175 Pickels, Edward 225 development of 275
Mller, Paul Hermann 187 Nowell, Peter 332 Pigliucci, Massimo 513 rabies-like viruses, discovery of 353
Mullis, Kary B. 434 nucleic acids, discovery of 42 Pintard, John 24 rabies, oral vaccination of wildlife 370
mumps virus, discovery of 163 nucleoprotein, RNA, tobacco mosaic virus, Pirie, Norman Wingate 170 rabies vaccine, development of 50
murine cytomegalovirus, discovery of 250 discovery of 170 Pirosky, Ignacio 276 rabies virus, discovery of 87, 88
murine leukemia and lymphoma viruses, discovery Oatley, Charles 169 plaque assay, virus quantification, development of rabies virus, genotyping / mapping of wildlife
of 231 Ochoa de Albornoz, Severo 288 238 variants 469
murine parvoviruses, discovery of 281 Offit, Paul A. 452 Plotkin, Stanley 320 rabies virus, morphology/morphogenesis of 322
murine polyoma virus, discovery of 274 Okamoto, Hiroaki 486 Plowright, Walter 291, 359, 516 Racaniello, Vincent 418
Murphy, Frederick A. 308, 338, 353, 390, 391, 414, Oker-Blom, Nils 345 Pneumocystis carinii pneumonia 417 radioimmunoassays, invention of 287
422, 446 Oldstone, Michael B. A. 295, 427 Poiesz, Bernard J. 413 radioisotopic labeling, development of 107
Murphy, James Bumgardner 243 Olitsky, Peter Kosciusko 119, 129 Polio Hall of Fame, Warm Springs, Georgia 186 Ramon, Gaston 122
Murray, Andrew W. 432 One Health / One Medicine Initiative 512 polio, planned global eradication of 517 Raoult, Didier 503, 515
Murray, Keith 477 One Medicine, concept of 43 polio vaccine, attenuated live-virus, development rapid sequencing of DNA, development of the
Murray Valley encephalitis virus, discovery of 120 one-step growth experiment 188 of 280 technology for 399
myxoma virus 66, 150 On the Origin of Species 37 polio vaccine, inactivated virus, development of rapid virus diagnostics linked to clinical care 360
Nahmias, Andr J. 315 optimum constellation of genes determines 249 rate-zonal ultracentrifugation, invention of 314
Nairobi sheep disease virus, discovery of 116 virulence, development of concept 393 poliovirus, adaptation to cotton rats and mice 185 rational antiviral drug design 363
Nathans, Daniel 348, 380 Orenstein, Walter A. 320 poliovirus, complementary DNA synthesized in Rauscher, Frank J. Jr. 231
Nathanson, Neal 296 origin of viruses, development of modern concepts vitro 496 Rayer, Pierre-Franois-Olive 40
National Center for Biotechnology Information of 454, 455 poliovirus, complete genomic sequence of 418 recombinant DNA technology, application of 373
(NCBI) and its public databases, development orthoreoviruses, discovery of 247 polioviruses, discovery of 100 recombinant-DNA technology, development of 372
of 451 Osler, William 43 poliovirus, infectious clone, development of 418 recombinant live-virus vaccine, development of
National Center for Infectious Diseases (CDC), Osterhaus, Albert D.M.E. 453, 493, 502 Pollack, Robert 374 first 442
founding of 414 Oxman, Michael N. 374 polyacrylamide gel electrophoresis, development Reed, Lowell 180
National Institute of Allergy and Infectious Ozawa, Yoshihiro 516 of 342 Reed, Walter 68, 69, 73, 74
Diseases, founding of 56 p53 tumor suppressor protein in SV40 cells, polymerase chain reaction (PCR), invention of 434 Reeves, William Carlisle 120, 198, 290
National Institutes of Health, founding of 56 discovery of 410 Poon, Leo 502 Relman, David A. 480
natural history of arboviruses in vectors 198 Padgett, Billie L. 278 Popper, Erwin 100 Remlinger, Paul Antoine 87, 95
natural selection, concept of 36 Page, Irvine 352 porcelain ultrafilter, development of 51 reoviruses, discovery of 247
negative contrast electron microscopy, invention Page, Larry 489 porcine circovirus, discovery of 385 replication of DNA and RNA, discovery of
of 282 Palese, Peter 506 porcine reproductive and respiratory syndrome mechanisms 288
Negri, Adelchi 85, 95 Panama Canal 91, 92, 93 virus, discovery of 464 representational difference analysis (RDA) 467
Negri body 85 Panum, Peter Ludwig 28 Porterfield, James 306 repressor genes, discovery of 334
Negri body, discovery of nature of 322 Paoletti, Enzo 433 Porter, Rodney R. 285 respiratory syncytial virus, discovery of 258
neuronal pathway tracers, viruses as 483 papillomavirus vaccines, development of 498 Prangishvili, David 383 restriction endonucleases, discovery of 348
Newcastle disease virus, discovery of 130 Parkman, Paul 320 Preston, Richard 478 restriction enzyme map of SV40 virus DNA 380
New, Emerging and Re-emerging Infectious Parodi, Armando S. 276 Price, W.H. 259 retroviral oncogenes, discovery of cellular origin
Diseases, development of concept of 470 Parrish, Colin R. 404 prion disease 124 of 392
Newhouse, Verne 350 parvovirus B-19 (fifth disease), discovery of 386 prions, etiologic agents of the spongiform retroviruses, morphology/morphogenesis/
Pasteur, Louis 33, 50, 57 encephalopathies 425 classification of 246
reverse genetics, development of 405 Rowe, Wallace P. 240, 264, 329 Shah, Keerti 362 Spence, Leslie 321
reverse genetics for negative-strand RNA viruses Rowson, Kenneth Edmund 293 Sharp, Phillip A. 377, 397 Spinco Model E analytical ultracentrifuge,
475 rubella virus, discovery of 113 Shatkin, Aaron J. 343, 388 development of 219
reverse transcriptase in retroviruses, discovery of Ruck, Brian 376 sheeppox virus 81 Spinco Model L preparative ultracentrifuge,
356 Ruckle, Gisele 319 Shelokov, Alexis 290 development of 225
Reyes, Gregory 467 Rueckert, Roland R. 407 Shimomura, Osamu 318 spontaneous generation, refutation of concept 14
rhinoviruses, discovery of 259 Rupprecht, Charles E. 469 Shope, Richard Edwin 134, 147, 156 Sputnik, virophage, discovery of 515
ribavirin, broad spectrum antiviral drug 357 Rush, Benjamin 21, 22 Shope, Robert Ellis 306, 353, 368, 468, 470 Stahl, Frank W. 272, 273
ribozymes, catalytic RNAs 416 Ruska, Ernst 157, 183 Shortridge, Kennedy F. 485 Stanley, Wendell Meredith 167, 235
Rice, Charles M. 508 Ruska, Helmut 127, 183 shotgun cloning/sequencing, invention of 466 Stark, George R. 411
Richardson, Carol 441 Russian Spring-Summer encephalitis virus, Shuda, Masahiro 514 St. Clair, Marty 447
Rico-Hesse, Rebecca 468 discovery of 178 Sickles, Grace 222 Steck, Franz 370
Riffat-Bay 87 Rutter, William J. 424 Siegert, Rudolf 338 Steinhardt, Edna Harde 111
Rift Valley fever virus, discovery of 151, 152 Sabattini, Marta 276 Sigurdsson, Bjrn 251 Steinman, Ralph A. 378
Riley, Vernon Todd 293 Sabi virus (Brazilian hemorrhagic fever), discovery simian foamy virus, infection in humans 369 Stent, Gunther 210
rinderpest 44 of 476 simian immunodeficiency viruses (SIVs), discovery Sternberg, George Miller 59
rinderpest, global eradication of 516 Sabin, Albert Bruce 154, 247, 280 of 438 Stewart, Sarah Elizabeth 274
rinderpest vaccine, attenuated-live virus, Sabin, Florence Rena 126 Siminovitch, Louis 227 St. Louis encephalitis virus, discovery of 159
development of 291 Salas, Rosalba A. 468 Simon, Charles Edmund 133 Stoker, Michael George Parke 301
rinderpest virus, discovery of 82 Salk, Jonas 249 Simpson, David I. 353 Stokes, Adrian 137
Rios, Francisco A. 181 Salzman, Norman 253 Sindbis virus, discovery of 254 Studdert, Michael J. 162, 446
Rivers, Thomas Milton 131, 132, 133, 134, 172, 177, Sambrook, Joseph 377 Singer, Beatrice A. 260 subacute sclerosing panencephalitis, discovery of
223 Sanarelli, Giuseppe 66 Singer, Maxine 256, 374 etiology 326
Rivers, Thomas Milton, criteria for proof of viral sandfly (phlebotomus) fever viruses, discovery of single burst experiment 188 Summers, Max D. 422
disease causation 177 102 single cell burst size, discovery of 248 Suttle, Curtis 461, 503, 515
Rizzetto, Mario 400 Sanford, Katherine K. 192 single nucleotide polymorphisms (SNPs), detection SV40 virus as cause of human tumors, controversy
RNA-dependent DNA polymerase in retroviruses, Sanger, Frederick 396, 399 of 440 362
discovery of 356 SARS coronavirus, discovery of 502 single stranded DNA viral genome, discovery of SV40 virus (simian virus 40), discovery of 292
RNA-dependent RNA polymerase in VSV, Sawyer, Wilbur Augustus 134, 137 286 Svedberg, Theodor 125
discovery of 355 scanning electron microscope, invention of 169 Sin Nombre virus (hantavirus pulmonary Swanepoel, Robert 500, 505
RNA interference (RNAi), discovery of 488 scanning tunneling electron microscope, invention syndrome), discovery of 472 Swanson, Robert A. 412
RNA splicing and split genes, discovery of 397 of 419 Sinsheimer, Robert L. 286 Sweet, Benjamin 292
Robbins, Frederick Chapman 110, 226 Schaeffer, Priscilla 422 Sipe, Jean D. 343 swine flu vaccination episode of 1976 395
Roberts, Richard J. 397 Schfer, Werner 257 Skehel, John J. 420 swine influenza virus, discovery of 147
Robinson, Harriet 296 Scherer, William 290 Slenczka, Werner 338 sylvan/jungle yellow fever cycle, discovery of 161
Robinson, Marion 262 Schleiden, Matthias 27 slow viruses, development of the concept of 251 Symons, Robert H. 372
Robinson, Roslyn Q. 338 Schmaljohn, Connie S. 368 smallpox 15, 16 Szmuness, Wolf 408
Robins, Roland K. 357 Schmidt, Nathalie J. 136 smallpox, global eradication of 398 Szostack, Jack W. 432
Rockefeller Foundation Virus Laboratory, founding Schneweis, Karl-Eduard 315 Smithburn, Kenneth C. 189, 216 Tacaribe virus, discovery of 321
of 134 Schoening, Harry 129 Smith, Geoffrey 296 Takahashi, Michiaki 320
Rockefeller Institute for Medical Research, Scholtissek, Christoph 393 Smith, Hamilton 348, 466 Takatsuki, Kiyoshi 413
founding of 77 Schramm, Gerhard 260 Smith, Hugh Hollingsworth 168 Taktsy, Gyula 252
Rockefeller, John D. Jr. 77 Schrdinger, Erwin 208 Smithies, Oliver 448 Talmage, David 266
Rockefeller, John D. Sr. 77 Schuster, Peter 454 Smith, Jean S. 469 Tamm, Igor 223, 317
Rodriguez, F.R. 269 Schuurs, Anton 364 Smith, Kendall 332 Taq polymerase 434, 457
Roeder, Peter 516 Schwabe, Calvin W. 512 Smith, Lloyd M. 445 Tatum, Edward Lawrie 213
Rohrer, Heinrich 419 Schwann, Theodor 27 Smith, Margaret Gladys 250, 264 Taubenberger, Jeffery K. 506
Roizman, Bernard 331, 440 scrapie prion, discovery of transmissibility of 171 Smith, Theobald 60 Taylor, Grant 312
role of monocytes in infections 126 SDS polyacrylamide gel electrophoresis, Smith, Wilson 155 Taylor, Richard Moreland 254, 290, 306
Rollin, Pierre E. 472, 500, 505 development of 342 Smolinski, Mark 470 Taylor, William 516
Rosenberg, Barbara 471 Sellards, Andrew 138 Snow, John 28 Temin, Howard M. 356
Rossman, Michael G. 407 semi-conservative mode of replication of DNA, solid phase synthesis technology, development of Ten Broeck, Carl 165
Ross, Ronald 45 discovery of 273 297 ter Meulen, Volker 296
Ross, R.William 262 Semmelweis, Ignaz Philipp 31 Song, Jin-Won 368 Terpstra, Cathrinus 464
rotaviruses, discovery of human 351 Sencer, David J. 391, 395 Sonnenberg, Nahum 450 Tesh, Robert B. 359, 468
Rothberg, Jonathan M. 509 Sendai virus (murine parainfluenza virus 1), Soper, Fred L. 161 Texas fever 60
Rott, Rudolf 393 discovery of 244 southern blotting, development of 389 The Hot Zone, publication of book 478
Rous, Francis Peyton 106, 156, 173 sequence-independent, single-primer amplification Southern, Edwin Mellor 389 Theiler, Arnold 83
Rous sarcoma virus, discovery of 106 (SISPA) 467 Spallanzani, Lazzaro 14 Theiler, Max 134, 143, 168
Roux, Pierre-Paul-Emile 50 Sever, John L. 252, 326 specificity of disease causation 33 Thompson, Wayne H. 294
Thottapalayam virus, discovery of 368 U.S. National Center for Biotechnology Information virology, Louis Pasteur, father of 33 WHO Global Polio Eradication Initiative 517
Thuillier, Louis 50 (NCBI), development of PubMed 484 Virology, publication of journal 253 Wigler, Michael H. 421, 467
thymus, role in cellular immunity, discovery of 300 U.S. National Library of Medicine 451 virophage, Sputnik, discovery of 515 Wildy, Peter 253, 335, 345
tick-borne encephalitis virus, discovery of 178 U.S. Public Health Service, founding of 23 Virosphere 402 Wiley, Don C. 420
Tignor, Gregory H. 353 vaccination 15, 16 virus entry mechanisms, discovery of 379 Wilkins, Maurice Hugh 241
Tischer, Ilse 385 vaccinia virus 111 viruses as neuronal pathway tracers 483 Wilkinson, Lise 406
Tiselius, Arne Wilhelm Kaurin 89 vaccinia virus, discovery of 54, 95 viruses in the ocean 461 Williams, Elizabeth S. 415
tobacco mosaic disease 53 vaccinia virus recombinants, for vectored viral viruses of Archaea, discovery of 383 Williams, Robley Cook 255
tobacco mosaic virus, crystallization of 167 vaccines 433 viruses replicating in both plants and insects, Will, Robert 479
tobacco mosaic virus, discovery of 62, 63 Valentine, Robin C. 330 discovery of 237 Wilson, Ian A. 420
tobacco mosaic virus, infectivity of RNA, discovery Valenzuela, Pablo 424 virus evolution, development of modern concepts Wimmer, Eckard 418, 450, 496
of 260 Valle, Henri 90, 128 454 Witkowski, Jan A. 96, 108, 109
tobacco mosaic virus, purification and van den Hoogen, Bernadette 493 virus, first visualization 54 Witkowski, Joseph 357
crystallization of 167 van der Groen, Guido 390 Virus Laboratory, University of California Berkeley, Woese, Carl Richard 402
tobacco mosaic virus, reconstitution from protein van Leeuwenhoek, Anton 11 founding of 235 Wollman, lie Leo 179
and RNA 255 van Regenmortel, Marc H. V. 460, 473 virus-like particles 498 Wollman, Elisabeth 179
Todaro, George 349 van Weemen, Bauke 364 virus neutralization, discovery of 59 Wollman, Eugne 179
Toll-like receptors 482 variant Creutzfeldt-Jakob disease (vCJD), BSE prion virus quantification by plaque assay, development Woodall, John P. 212, 471
toroviruses, discovery of 428 479 of 238 Woode, Gerald N. 428
torque teno virus, discovery of 486 varicella-zoster virus, discovery of 127 virus species, definition of 460 Woodruff, Alice Miles 149
Torsvik, Vigdis 383 variolation 12 visualization of viral DNA by electron microscopy Work, Telford Hindley 254, 269, 290
Tournier, Paul 310 variola virus, discovery of 54, 95 284 World Wide Web, development of 465
Towner, Jonathan S. 505 Varmus, Harold E. 392 Vogt, Marguerite 238, 248 Wright, Arthur 154
transduction: DNA transfer by bacteriophage, Venezuelan equine encephalitis virus, discovery Volberding, Paul 417 Wyckoff, Ralph Walter 181
discovery of 236 of 181 von Ardenne, Manfred 169 Yabe, Yoshiro 312
transformation in bacteria, discovery of 135 Venter, J. Craig 461, 466 von Behring, Emil Adolf 49 Yalow, Rosalyn Sussman 287
transmissible spongiform encephalopathy, scrapie Verge, Jean-Louis-Armand 139 von Borries, Bodo 183 Yamamoto, Janet K. 444
171 Vero cell line, establishment of 307 von Prowazek, Stanislaus 95 Yamanishi, Koichi 449
transmission electron microscope, invention of vesicular exanthema virus of swine, discovery of Wagner, Robert 253 Yamanouchi, T. 118
157, 158 174 Wakita, Takaji 508 Yanagihara, Richard 368
transovarial transmission of viruses by arthropods vesicular stomatitis viruses, discovery of 129 Waldmann, Otto 128 Yasumura, Yosihiro 307
359 Veterinary Virology, publication of book 446 Walker, Duard Lee 278 yeast artificial chromosomes, cloning of large DNA
transposable elements, transposons, development Villarreal, Luis 454 Walsh, Peter 500 fragments 432
of the concept of 191 Villerm, Louis-Ren 28 Wandeler, Alexander 370 yellow fever 46, 48, 67, 91, 92, 93
Traub, Erich 172 Vinograd, Jerome 219, 272 Wang, David 514 yellow fever epidemic, Philadelphia 21
Traum, Jacob 129, 174 Viral and Rickettsial Infections of Man, publication Ward, Richard 452 yellow fever epidemics in U.S. 47
Tree of Life, based on mRNA gene sequences 402 of book 223 Waring blender experiment 234 yellow fever jungle/sylvan cycle, discovery of 161
Trentin, John J. 312 viral damage to cellular homeostasis, development Waterson, Anthony Peter 406 yellow fever vaccine, development of 168
triplet coding strategy of DNA, discovery of 302 of concept 427 Watson, James Dewey 241 yellow fever virus 68, 69, 72
trypsinization for dispersal of cells in culture 114 viral diagnostic instruments, development of high Watts, Douglas M. 359 yellow fever virus, discovery of 68, 69, 70, 71
Tsien, Roger 318 throughput 487 Weaver, Scott 454 yellow fever virus, transmission to rhesus macaques
tuberculosis 44 viral diagnostics linked to clinical care 360 web-based surveillance tools (Google), 137, 138
tumors in hamsters induced by human viral disease diagnostics, broad application of development of 489 York, Charles J. 261
adenoviruses, discovery of 312 methods 224 Webb, Patricia A. 325, 390 Yoshida, Mitsuaki 413
Tumpey, Terrence M. 506 viral disease diagnostics, founding of 136 Webster, Leslie Tillotson 143, 159 Youngner, Julius 249
Twort, Frederick William 115 viral disease pathogenesis research, founding of Webster, Robert G. 316, 473, 485, 490 Young, Stuart 415
Tyrrell, David Arthur John 328 221 Weigle, William 295 Zaitlin, Milton 422
Uganda Virus Research Institute 216 viral genomes, sequencing of MS2 and 174 396 Weinberg, Robert A. 421 Zeiss, Carl 26
Ugolini, Gabriella 483 viral oncogene hypothesis, development of 349 Weissenbacher, Mercedes 276 Zhdanov, Victor 345
ultracentrifuge, development of 125 viral pathogenesis research, founding of the modern Weissmann, Charles 405 Zhou, Jian 498
ultrafilterable virus, concept of 53 era 296 Weller, Thomas Huckle 110, 127, 226, 264 zidovudine, first anti-HIV drug 447
uncultivable viruses, development of techniques to viral receptors on target cells, discovery of 218 Wensvoort, Gert 464 Zilber, Lev Alexandrovich 178
find 467 viral vaccines based upon innovative technologies western blotting, invention of 411 Zillig, Wolfram 383
Urbani, Carlo 502 443 western equine encephalitis virus, discovery of 145 Zinder, Norton 236
U.S. Army Yellow Fever Commission 73 viral vaccines, development, promulgation and West Nile virus, discovery of 189 Zinke, Georg Gottfried 19
U.S. Centers for Disease Control and Prevention commercialization of 320 West Nile virus in the United States 492 Zinkernagel, Rolf M. 381
202, 304, 414 Virchow, Rudolf Ludwig Karl 35 What is Life?, publication of book 208 zur Hausen, Harald 384
U.S. Communicable Disease Center 304, 340 viroids, discovery of 366 White, David Ogilvie 354 Zu Rhein, Gabriele M. 278
U.S. National Academy of Sciences, founding of 39 virology, establishment as a distinct field of science Whitfield, Sylvia G. 390 Zworykin, Vladimir 169
131 Whitley, Richard J. 363
The Author Frederick A. Murphy is a professor in the Department of the American Committee on Arthropod-Borne Viruses,
Pathology, University of Texas Medical Branch (UTMB), American Society of Tropical Medicine and Hygiene; the
Galveston, Texas. He holds a BS and DVM from Cornell University of California Davis Medal; the K.F. Meyer Gold
University and a PhD from the University of California, Davis. Headed Cane from the American Veterinary Epidemiology
He has served as Dean and Distinguished Professor, School of Society and American Veterinary Medical Association; and an
Veterinary Medicine, and Distinguished Professor, School of Honorary Fellowship from the John Curtin School of Medical
Medicine, University of California, Davis. Before that he served Research, the Australian National University. At UTMB he is a
as Director of the Division of Viral and Rickettsial Diseases and member of the Institute for Human Infections and Immunity,
then Director of the National Center for Infectious Diseases, the Center for Biodefense and Emerging Infectious Diseases,
Centers for Disease Control, Atlanta. He is a member of the the Galveston National Laboratory, and the McLaughlin
Institute of Medicine of the U.S. National Academy of Sciences Endowment for Infection and Immunity. His professional
(where he has served on 10 committees) and is a member of interests, in association with many colleagues, include the
the Deutschlands Nationale Akademie der Wissenschaften pathology and epidemiology of highly pathogenic viruses/
(German National Academy of Sciences). His honors include viral diseases: (1) Rabies: long running studies leading to the
an honorary Doctor of Medicine and Surgery from the identification of more than 25 viruses as members of the virus
University of Turku, Turku, Finland; an honorary Doctor of family Rhabdoviridae, identification and characterization
Science from the University of Guelph, Ontario, Canada; an of the first rabies-like viruses, and major studies of rabies
honorary Doctor of Veterinary Medicine from the University pathogenesis in experimental animals, including the initial
of London, United Kingdom; an honorary Doctor of Science descriptions of infection events in salivary glands and in
from University College Dublin, Ireland; the Presidential Rank muscle. (2) Arboviruses: long running studies of alphaviruses,
Award of the U.S. Government; the PennVet World Leadership flaviviruses and bunyaviruses with the initial proposal for the
Award from the University of Pennsylvania; the Distinguished establishment and naming of the virus family Bunyaviridae,
Microbiologist Award from the American College of Veterinary and characterization of reo-like viruses culminating in the
Frederick A. Murphy Microbiologists; the Richard Moreland Taylor Award from establishment and naming of the virus genus Orbivirus. (3) Viral
hemorrhagic fevers: long running studies leading to the initial
discovery of Marburg and Ebola viruses, and characterization
of several other hemorrhagic fever viruses, culminating in the
establishment and naming of the virus families Arenaviridae
(e.g., Lassa and Machupo viruses) and Filoviridae (Marburg
and Ebola viruses), and elucidation of the pathology and
pathogenesis of the diseases in humans and experimental
animals caused by these exceptionally virulent agents. (4)
Viral encephalitides: long running studies of the pathogenesis
of neurotropic viruses in experimental animals, including
alphaviruses, flaviviruses, bunyaviruses, enteroviruses,
paramyxoviruses, herpesviruses, and others. He has been
a leader in advancing the concept of new and emerging
infectious diseases and new and emerging zoonoses, the
concept which has reenergized the infectious disease research
sciences. Most recently his interests have included the threat
posed by bioterrorism and national efforts in prevention of this
threat.

1998: John, Terence, Irene,


Fred, Rick and Tim Murphy

Вам также может понравиться