Вы находитесь на странице: 1из 36

Chapter

38
Antiviral Agents and
Protease Inhibitors
P AT R I C K M. W O S T E R

Drugs Covered in This Chapter*


Acyclic nucleoside analogues Antiretroviral agents: Nonnucleoside • Fosamprenavir
• Acyclovir reverse transcriptase inhibitors • Indinavir
• Adefovir dipivoxil • Delavirdine • Lopinavir
• Cidofovir • Efavirenz • Nelfinavir
• 6-Deoxyacyclovir • Nevirapine • Ritonavir
• Famciclovir Conventional nucleoside analogues • Saquinavir
• Ganciclovir • Tipranavir
• Cytarabine
• Valacyclovir • Idoxuridine Inhibitors of viral attachment,
Agents affecting the ribosome • Ribavirin penetration, or early replication
• Methisazone • Trifluorothymidine • Amantadine
Antiretroviral agents: Nucleoside • Vidarabine • Interferon
reverse transcriptase inhibitors Fusion inhibitors • Rimantadine
• Abacavir • Enfuvirtide Neuraminidase inhibitors
• Didanosine • Maraviroc • Oseltamivir
• Emtricitabine HIV integrase inhibitors • Peramivir
• Lamivudine • Zanamivir
• Raltegravir
• Stavudine Non nucleoside analogues
HIV protease inhibitors
• Tenofovir disoproxil • Boceprevir
• Zalcitabine • Amprenavir
• Fomivirsen†
• Zidovudin • Atazanavir
• Foscarnet
• Darunavir
• Telaprevir

Abbreviations
AIDS, acquired immunodeficiency AUC, area under plasma concentration– CMV, cytomegalovirus
syndrome time curve CNS, central nervous system
ara-HX, arabinosyl hypoxanthine CCD, catalytic core domain CSF, cerebrospinal fluid
ARC, AIDS-related complex cDNA, complementary DNA D4T, stavudine
*
Drugs listed include those available inside and outside of the United States; drugs available outside of the United States are shown in italics.

Fomivirsen has been withdrawn from the U.S. market. Kaduse.com
1267
1268 PART III / PHARMACODYNAMIC AGENTS

DANA, 2-deoxy-2,3-dehydro-N- HAV, hepatitis A virus IV, intravenous


acetylneuraminic acid HBV, hepatitis B virus MHC, major histocompatibility complex
ddI, didanosine HCV, hepatitis C virus NA, neuraminidase
ddC, zalcitabine HEV, hepatitis E virus RNAi, RNA interference
DMSO, dimethylsulfoxide HHV, human herpesvirus RSV, respiratory syncytial virus
EBV, Epstein-Barr virus HIV, human immunodeficiency virus RT, reverse transcriptase
FDA, U.S. Food and Drug Administration HPV, human papilloma virus siRNA, short interfering RNAs
GI, gastrointestinal HSV, herpes simplex virus 3TC, lamivudine
HA, hemagglutinin HTLV-1, human T-cell leukemia virus 1 VZV, varicella-zoster virus
HAART, highly active antiretroviral IC50, half maximal inhibitory ZDV, zidovudine
therapy concentration

SCENARIO
Douglas Slain, PharmD, BCPS
WM is a 37-year-old white woman who is employed as a nurses’ the residents of her nursing home had an H1N1 strain of influ-
aid in an elder care nursing home. She presents to the hospital enza that has the neuraminidase-resistant H274Y mutation. This
with extreme flu-like symptoms and is having difficulty breath- mutation will drastically reduce the effectiveness of neuramini-
ing. She is placed in the intensive care unit where she ultimately dase inhibitors that are dependent on C6 hydrophobic binding.
requires intubation and mechanical ventilation to maintain her Most circulating strains have also been resistant to rimantadine.
respiratory function. WM is also given empiric broad-spectrum Treatment options to consider are investigational intravenous
antibiotics in case of bacterial pneumonia. A rapid influenza zanamivir and investigational intravenous peramivir.
test is positive. She needs anti-influenza therapy, but oral osel-
tamivir and inhaled zanamivir cannot be reliably delivered given (The reader is directed to the clinical solution and chemical analy-
her sedation, intubation, and mechanical ventilation. Many of sis of this case at the end of the chapter.)

INTRODUCTION undergo mutations, resistance to existing therapies can


develop. The discovery of new antiviral agents is thus an
Viruses are the smallest of the human infectious agents important ongoing effort in medicinal chemistry.
and range in size from about 20 nm to about 300 nm
in diameter (1,2). They contain one kind of nucleic
acid, either RNA or DNA, as their entire genome, which VIRUS STRUCTURE AND CLASSIFICATION (1,2)
codes for a variety of enzymes and other proteins used in
replication and transmission of the organism. It can be Numerous species of virus that infect bacteria, plants,
argued that a virus does not qualify as a true life form, and animals have been identified, and they exhibit a
since it is nothing more than a nucleic acid strand with remarkable range of diversity. All viruses exist as obli-
associated proteins, and it cannot move on its own power. gate cellular parasites, and as such, they do not possess
However, when it attaches itself to a host cell, it internal- the complex biochemical machinery that is charac-
izes itself and forces the host to make additional copies teristic of higher organisms. However, they do have a
of the virus, demonstrating a clear reproductive plan. defined macromolecular structure that is designed
During replication, it uses host cellular biochemicals to protect them from the environment and facilitate
and processes and, thus in a sense, takes in “nutrients” their entry into cells. The basic subunit of a virus is
in order to survive and multiply. In some cases, viruses its genome, which can be made up of either DNA or
respond to external conditions and escape the immune RNA. The nucleic acid portion of a virus can be sin-
response by integrating into the host DNA, demonstrat- gle or double stranded, and may be present in linear
ing the ability to respond to external stimuli. Although or circular form. Viral genomic DNA or RNA is often
viruses are simple organisms, they are a significant caus- associated with basic nucleoproteins and may be sur-
ative agent for numerous human diseases and, as such, rounded by a symmetrical protein known as a capsid.
represent one of the major challenges in the area of drug The capsid is made up of repeating structural units
discovery. Agents that are used clinically for a variety of known as protomers, which themselves are made up
viral diseases act by targeting processes that are specific of nonidentical protein subunits. The combination
to the virus, such as a unique viral enzyme or a neces- of the nucleic acid core and the capsid is termed the
sary process such as transcription. However, to date, no nucleocapsid, and in some cases, this comprises the
drug has been discovered that is truly curative for viral entire virus. In other cases, the nucleocapsid structure
infection. In addition, because viruses have the ability to Kaduse.com
is surrounded by a lipid-containing membrane that is
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1269

derived during viral maturation, when the virus undergoes and viral nucleic acid prevents the formation of “empty”
budding through the host cell membrane. The com- helical particles. Finally, some virus particles, such as
plete virus particle, with or without an envelope, is the large and complex poxviruses, do not exhibit cubic
termed the virion. Viral architecture can be grouped or helical symmetry, but instead form more compli-
into three types based on the arrangement of morpho- cated structures that can be spherical, brick-shaped,
logic subunits, and each virus exhibits cubic (icosahe- or ovoid. A subset of complex viruses is termed pleo-
dral) symmetry, helical symmetry, or has a complex morphic, in that they assume multiple complex mor-
structure. Icosahedral virions are symmetrical struc- phologies. Advances in the visualization of viral capsid
tures that contain 20 surfaces, each of which is an equi- structure suggest that the assembly and processing of
lateral triangle. A sufficient number of capsid structural viral proteins could serve as a new target for antiviral
units must be employed in the icosahedron to make a drug development (3).
capsid large enough to encapsulate the viral genome. Viral taxonomy is complex, and viruses are classified
Morphologic units called capsomeres are seen on the according to a number of factors, including morphol-
surface of icosahedral virus particles. These structures ogy, properties of the genome (i.e., DNA vs. RNA, single
are clusters of polypeptides, but they do not necessarily strand vs. double strand, linear vs. circular, sense vs. anti-
correspond to the chemically defined structural units. sense), physicochemical properties, structure of associ-
Some viruses arrange their structural subunits into a ated proteins, replication strategy, and so on. Viruses are
standard helical formation. In viruses with helical sym- separated into major groups called families, with names
metry, protein subunits systematically bind to the viral that end in the suffix –viridae, and then into genera
nucleic acid, ultimately forming a nucleocapsid helix. that end in –virus. Thus, pox viruses are in the family
The filamentous nucleocapsid is then coiled inside a Poxviridae, and in the genus Poxvirus. A comparison of
lipid-containing envelope. Unlike icosahedral virions, the genetic and structural features of viral families with
regular, periodic interaction between capsid protein members that can infect humans appears in Table 38.1.

CLINICAL SIGNIFICANCE
Antiviral agents include diverse these agents can maintain activity against viruses that have
compounds with varied actions. developed resistance to other nucleoside agents through certain
A thorough clinical understanding resistant mutations.
of most of these compounds is depen- In clinical practice, HIV antiretroviral regimens usually
dent on a basic appreciation of biochem- require a pair of nucleoside reverse transcriptase inhibitors in
istry and medicinal chemistry. Viruses are mainly composed of the regimen. It is of paramount importance that combinations
either RNA or DNA nucleic acid strands. As such, one of the of “like” nucleosides analogs (e.g., two thymidine, two cytosine)
most ideal targets for treating viral pathogens has been the inhi- are not used together as they have displayed antagonism and
bition of viral replication. Successful viral replication is depen- reduced viral load suppression. Nonnucleoside reverse transcrip-
dent on enzyme-mediated transcription of viral RNA or DNA. A tase inhibitors are not structurally related to nucleic purines and
significant number of antiviral agents, including HIV nucleoside pyrimidines; therefore, they do not act as substrates of the tar-
reverse transcriptase inhibitors, and a majority of antiherpes get enzyme.
agents are designed to be “false” substrates of viral transcrip- Protease inhibitors, which are the most potent antiretrovi-
tion enzymes. It is through competitive inhibition that these ral agents available, were the end result of coordinated chemical
agents are able to fulfill their intended effect. Their chemical design and structure-based computational analysis of the prote-
structures are similar to naturally occurring substrate nucleo- ase enzyme. With identification of the protease crystalline struc-
side purines (adenosine and guanosine) and pyrimidines (thy- ture and identification of active binding sites, scientists were
midine and cytidine). However, as they are purposely modified able to create compounds that would fit the protease enzyme
in key positions with functional groups designed to foil poly- with strong affinity, and cause an inhibition of protease func-
merase enzymes, their incorporation will lead to termination of tion. The clinical impact of these man-made drugs is regarded
replication. While these nucleoside antiviral agents are often by many as the greatest single advance in the treatment of HIV
actively carried into cells via specific nucleoside transporting infection.
proteins, they typically require triphosphorylation to become
active intracellularly. It is important to recognize that resistance Douglas Slain, PharmD, BCPS
to these antiviral medications can develop when genetic muta- Associate Professor
tions alter the ability for viral enzymes to phosphorylate the College of Pharmacy
drugs. Drugs like cidofovir and tenofovir are nucleotide analogs, West Virginia University
which means they are already monophosphorylated. As a result, Morgantown, WV
Kaduse.com
1270 PART III / PHARMACODYNAMIC AGENTS

TABLE 38.1 Characteristics of Virus Families Containing Members That Infect Humans
Family Examples Genome Capsid Size Envelope Diseases
Symmetry (nm−1)

Parvoviridae Parvovirus B19 ssDNA sense or Icosahedral 18–26 No Erythema infectiosum (fifth
antisense disease); polyarthralgia
arthritis; aplastic crisis, anemia
Papillomaviridae Human papilloma (wart) ds, circular DNA Icosahedral 55 No Warts; salivary gland
virus; polyoma virus; infection; multifocal
SV 40 leukoencephalopathy; tumors
(i.e., cervical)
Adenoviridae Multiple types dsDNA Icosahedral 70–90 No Infections of the eye and
(40 adenoviruses and respiratory tract; tumors
mastadenovirus)
Hepadnaviridae Hepadnavirus, hepatitis dsDNA, circular, Icosahedral 40–48 Yes Hepatitis B; tumors
B virus one ss region
Herpesviridae Herpes simplex I and II; dsDNA Icosahedral 150–200 Yes Eye, skin and genital infection;
varicella zoster; herpes chickenpox; shingles;
zoster; cytomegalovirus; mononucleosis; tumors
Epstein-Barr virus
Poxviridae Variola; vaccinia dsDNA Complex 230–400 Complex Smallpox; cowpox;
chickenpox; tumors
Picornaviridae Hepatitis A virus; ssRNA, sense Icosahedral 28–30 No Respiratory diseases;
poliovirus; enterovirus; gastrointestinal diseases;
rhinovirus, coxsackie polio; aseptic meningitis
virus A and B
Astroviridae Astrovirus ssRNA, sense Icosahedral 28–30 No Diarrhea in infants and
immunocompromised patients
Caliciviridae Norwalk virus ssRNA, sense Icosahedral 27–40 No Epidemic gastroenteritis
Togaviridae Rubella virus; ssRNA, sense Icosahedral 50–70 Yes Measles (rubella)
alphavirus, arbovirus
Flaviviridae Hepatitis C virus; arbo- ssRNA, sense Complex 40–60 Yes Hepatitis C; yellow fever;
virus; yellow fever virus; dengue fever; encephalitis;
dengue virus; West Nile tumors
virus
Coronaviridae Coronavirus ssRNA, sense Complex 120–160 Yes Colds; gastroenteritis in
infants; SARS
Retroviridae HIV I and II; lentivirus; ssRNA as dimer Complex 80–100 Yes AIDS; AIDS-related complex;
human T-cell breast cancer; human T-cell
lymphotropic viruses leukemia; nasopharyngeal
carcinoma
Arenaviridae Arenavirus ssRNA, antisense Complex 50–300 Yes Lassa fever; hemorrhagic
fever; choriomeningitis
Orthomyxoviridae Influenza virus A, B, C ssRNA, antisense Helical 80–120 Yes Influenza
Bunyaviridae Hantavirus ssRNA, antisense Helical 80–120 Yes Hemorrhagic fever
Rhabdoviridae Rhabdovirus; rabies ssRNA, antisense Helical 75–180 Yes Rabies; encephalitis
virus; encephalitis virus
Paramyxoviridae Syncytial virus; ssRNA, antisense Helical 150–300 Yes Mumps; measles (rubeola)
parainfluenza virus
Filoviridae Marburg virus; ssRNA, antisense Helical 80–800 Yes Marburg viral fever; ebola
Ebola virus hemorrhagic fever
Reoviridae Reovirus; rotavirus; dsRNA in 10–12 Icosahedral 60–80 No Mild respiratory and
orbivirus pieces gastrointestinal infection;
Colorado tick fever
Prion Prion proteinaceous None NA NA No Bovine spongiform
material encephalopathy; Creu
tzfeldt-Jakob disease
ds, double stranded; NA, not applicable; SARS, severe acute respiratory syndrome; ss, single stranded.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1271

VIRAL REPLICATION, CELLULAR EFFECTS, AND endocytosis, fusion of the viral envelope with the cell
PATHOGENESIS (2,4) membrane, or in some cases direct penetration of the
membrane. Following penetration of the cell, viruses
As mentioned earlier, all viruses exist as obligate intra- must be uncoated, resulting either in the naked nucleic
cellular parasites, and as such, they rely on the cellular acid or in the nucleocapsid form, which usually contains
machinery of the host for their growth, development, polymerase enzymes. After they have been uncoated,
and replication. In order to synthesize the proteins viruses are no longer infectious.
needed for viral replication, the organism must be capa- Once the virus has penetrated the cell and uncoated,
ble of producing usable mRNA in sufficient quantities to it enters a segment of its life cycle known as the eclipse
compete with host mRNA for protein synthesis. During period, the length of which varies with the type of virus.
viral replication, all of the macromolecules required by It is during this time that the virus utilizes host resources
the virus are synthesized in a highly organized sequence. to replicate and produce necessary viral proteins. Cells
The replication cycle (Fig. 38.1) begins when the intact that can support viral reproduction are termed permis-
virion binds to a host cell through electrostatic adsorp- sive, and as a result, the infection is known as productive,
tion to a specific “receptor” site. This process is known since it results in new viral particles. When new infectious
as the attachment phase. Attachment is most likely a for- viral particles are produced, host cellular metabolism may
tuitous event resulting from structural complementarity be completely directed to the production of viral prod-
between the exterior structure of the virion and a nor- ucts, resulting in destruction of the cell. In other cases,
mal cell surface structure on the host cell. For example, host cell metabolism is not dramatically altered, and the
HIV binds to the CD4 receptor on cells of the immune infected cell can survive. During viral reproduction, up
system, rhinoviruses bind ICAM-1, and Epstein-Barr virus to 100,000 new virions can be produced, and the replica-
recognizes the CD21 receptor on B cells. Recent studies tion cycle can vary from a few hours to more than 3 days.
suggest that some mammalian cells can develop proteins Some cells types, called nonpermissive, are unable to
that restrict the binding of viral capsid structures, thus support the reproduction of an infective virion, resulting
preventing entry into noninfected cells (5). When attach- in an abortive infection. Abortive infections also occur
ment has been achieved, the virion enters the penetra- when the virus itself is defective. Either situation can lead
tion phase, the process by which it gains entry into the to a latent infection, where the viral genome may persist
host cell. Penetration may occur by receptor-mediated in a surviving host cell. As will be described below, such

ADENOVIRUS Cytoplasm
1
ADSORPTION Nucleus
10 2
RELEASE PENETRATION

9 Viral DNA 3
ASSEMBLY UNCOATING
Early proteins
Viral mRNA
Viral capsid Viral DNA in
Viral protein the nucleus
Ribosomes

Transfer of capsid
proteins to nucleus 4
8
EARLY
CONDENSATION Synthesis of late TRANSCRIPTION
proteins (capsid Synthesis of early
proteins) proteins (enzymes such
Synthesis of
as DNA polymerase)
early mRNA
Synthesis of viral
DNA and late mRNA
7 5
LATE EARLY
TRANSLATION TRANSLATION

6
DNA SYNTHESIS
AND LATE
TRANSCRIPTION

FIGURE 38.1 Steps involved in the viral life cycle. (Used with permission from Brooks GF, Butel JS, Morse SA, et al.
Jawetz, Melnick and Adelberg’s Medical Microbiology, 23rd Ed. New York: McGraw-Hill Companies, 2004.)
Kaduse.com
1272 PART III / PHARMACODYNAMIC AGENTS

an infection can lead to the transformation of a cell from be translated by the host ribosome. Following the initial
normal to malignant. translation of the sense strand, it serves a second func-
tion, namely to serve as a template for the synthesis of
DNA Virus a (–) or antisense strand via an RNA-dependent RNA
polymerase. This antisense strand then can be used to
The strategies used by various viruses to replicate vary produce additional sense-strand RNAs that are infectious
widely, but all are characterized by the need to tran- and can also serve as mRNA. These progeny sense RNA
scribe mRNA that is suitable for translation of viral strands are then packaged into an intact virion prior to
proteins. There are several pathways leading to the transmission to another host cell.
required mRNA, after which the host enzymes and raw The second group of single-stranded RNA viruses,
materials are used to make viral proteins. Early viral including orthomyxoviruses, bunyaviruses, arenavi-
proteins used in replication are synthesized immedi- ruses, paramyxoviruses, filoviruses, and rhabdoviruses,
ately after infection, while late proteins used to pro- all contain an antisense RNA genome that can only be
duce the complete virion structure are synthesized after used for transcription of new RNA. All antisense RNA
viral nucleic acid synthesis. Most DNA viruses contain viruses contain an RNA transcriptase as part of their
double-stranded DNA as their genome and thus can virion, because the host cell does not have this type of
replicate using host cell machinery to produce mRNA RNA-dependent RNA polymerase. In the first round of
directly. Papillomavirus, adenovirus, and herpesvirus genome expression, a series of short sense-strand RNAs
are replicated in the host nucleus, and thus use tran- are made and then translated to form the required viral
scriptional enzymes of the host (i.e., DNA-dependent enzymes for replication. Ultimately, these enzymes are
RNA polymerase) to synthesize mRNA. This mRNA is used to produce a full-length sense RNA strand, which
then translated to form proteins needed by the virus, is then used to make multiple copies of the antisense
including enzymes (e.g., DNA-dependent DNA poly- viral genome. The progeny antisense DNA strands by
merase) used to produce progeny DNA copies. These themselves are not infectious, because they have not yet
progeny DNA strands are infectious. By contrast, pox- been packaged with the required RNA transcriptase.
viruses replicate in the cytoplasm using a mechanism When the progeny antisense RNA has been synthe-
that is not well understood, wherein the genome is ini- sized, it is packaged into an intact virion, in which form
tially transcribed by a viral enzyme in the virion core. it becomes infectious prior to transmission to another
Parvoviruses contain a single-stranded DNA genome, cell.
and must synthesize double-stranded DNA in the host The third group of RNA viruses is the retroviruses, in
nucleus prior to synthesis of mRNA and translation of which single-stranded RNA exists as a dimer of a sense
proteins. This process may or may not require a helper and antisense strand. The genomic RNA strands can
virus such as herpes simplex. The hepatitis B viral be base-paired, although the structure of this complex
genome, comprised of double-stranded DNA, contains is not well understood, or the strands can be hydrogen
numerous gaps that must be repaired using a DNA poly- bonded to other macromolecules in the virion. Retroviral
merase packaged in the virion before transcription to genomic RNA serves a single function, namely, to act as
form mRNA. a template for the formation of double-stranded viral
DNA. Host cells do not contain an enzyme that can form
RNA Virus DNA from viral RNA, and thus the virion of a retrovi-
Compared to DNA viruses, those viruses with RNA-based rus must contain a reverse transcriptase (RT) enzyme, as
genomes have evolved a wide variety of reproductive strat- well as various host tRNA molecules. Transcription of the
egies. The single-stranded RNA viruses may be divided genome begins when a complex of RT and tRNA binds to
into three groups that differ in the method by which the viral genome. A complimentary DNA strand is then
the RNA genome is utilized. In all three groups, the synthesized using one of the host tRNAs as a primer, and
RNA genome must serve two functions: to be translated the original RNA strand is digested by RNAse H, and viral
to form protein and to be replicated to form progeny ribonuclease packaged in the virion. A complimentary
RNA strands. The first group is comprised of viruses such DNA strand is then synthesized, and the resulting dou-
as picornaviruses, flaviviruses, and togaviruses that have ble-stranded DNA is translocated to the nucleus, where
an RNA genome that can be used directly as mRNA. viral enzymes incorporate genome-length viral DNA
Viral RNA that can be used as mRNA is by convention into the host genome. In some cases, the viral portion
termed (+) or sense-strand RNA. In most cases (e.g., of the genome can remain dormant for long periods, or
picornaviruses), this sense-strand RNA binds to the it may be immediately used to make progeny viral RNA,
host ribosome shortly after entering the cell, where it a process that is catalyzed by host RNA polymerase II.
is read and used to produce a single polypeptide called Transcription produces both shortened segments that
the polyprotein. The polyprotein is then processed by are used to make polyproteins and full length progeny
autocatalysis and various proteolytic enzymes to produce RNA. The polyproteins are processed to form various
the required viral proteins. In some cases (e.g., togavi- viral proteins, while the full-length RNA is packaged into
ruses), only a portion of the RNA genome is available to an infectious virion.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1273

Virus Protein vesicles that are stored in between the inner and outer
In addition to replication of the viral genome, a number aspect of the cell membrane. Egress of the herpesvirus
of other structures associated with the complete virion vesicle always occurs through destruction of the host cell.
can also be made. A number of viral proteins may be syn- Virus Pathogenesis
thesized that have important functions in the structure,
transmission, and survival of the virus. These proteins can A complete discussion of viral pathogenesis is beyond the
protect the genetic material in the virus from destruc- scope of this chapter. However, in general it may be con-
tion by nucleases, participate in the attachment process, sidered that the symptoms of a viral infection arise from
and provide structure and symmetry to the virion. In the response to viral replication and cell injury in the host.
addition, in certain cases where the virus requires an These responses range from asymptomatic or subclinical
enzymatic process for which there is no host enzyme, a to severe clinical manifestations, and may be either local
virion may include enzymes such as RNA polymerases or or systemic. Understanding the biochemical events that
an RT. Some viruses require a lipid envelope that con- produce viral diseases can aid in the design of effective
tains transmembrane proteins specifically coded for by and specific therapies. Not surprisingly, viral pathogen-
the virus and that envelops the genetic material during esis occurs in distinct steps: 1) viral entry into the host
viral budding. Viral envelopes contain glycoproteins that and primary viral replication; 2) viral spread; 3) cellular
are involved in cell recognition during attachment to the injury and host immune response; 4) viral clearance or
host cell. These glycoproteins often reflect the composi- establishment of persistent infection; and 5) viral shed-
tion of glycoproteins in the host cell. They are a determi- ding. Most viruses enter the host through the respiratory
nant of the antigenic nature of viruses, and thus facilitate or gastrointestinal tract, but may also penetrate the skin,
recognition by the immune system of the host. However, urogenital tract, or conjunctiva. In a few cases, virus par-
depending on their composition, they can also help the ticles can enter through direct injection (e.g., HIV and
virus elude neutralization by the immune system. hepatitis) or through insect bites (arboviruses). When
a local infection occurs, the virus replicates near the
site of entry, and the underlying tissue is not affected.
Cellular Egress However, some viruses are able to migrate to other sites,
Viruses use one of two strategies for exiting infected cells. usually through the bloodstream or lymphatic system, to
Nonenveloped viruses (picornaviruses, rheoviruses, etc.) produce systemic infections. When infection occurs at
complete their maturation by assembling into their corre- a remote site, most viruses demonstrate tissue or organ
sponding virion within the cell nucleus or the cytoplasm. preference (e.g., herpesvirus localizes in nerve ganglia,
For example, picornaviruses assemble by clustering 60 and the rabies virus migrates to the CNS). Localization
copies of each of three viral proteins, called VP0, VP1, of a virus in a particular tissue can be the result of cell
and VP3, into a structure called a procapsid. Viral RNA receptor specificity or can arise because a virus may be
is then packaged into the procapsid, and proteolytic activated by proteolytic enzymes in a specific cell type.
cleavage of VP0 produces two new viral proteins called Clinical disease develops through a complex series of
VP2 and VP4. The resulting conformational change pro- events when virus-infected cells are destroyed or their
duces a stable and symmetrical structure that shields the function is impaired, and some symptoms such as mal-
genome from degradation by host nucleases. In most aise and anorexia can result from host responses such as
cases, destruction of the host cell is required when the cytokine release. Disease-mediated damage may become
virion exits. Enveloped viruses (all antisense RNA viruses, chronic when cell types that do not regenerate (e.g.,
togaviruses, flaviviruses, coronaviruses, hepadnaviruses, brain tissue) are involved. Ultimately, the host either
herpesviruses, and retroviruses) contain proteins that succumbs to the infection; develops a chronic, latent, or
carry signal sequences and markers that cause them to subclinical infection; or completely recovers. In chronic
be inserted into the inner and outer surface of the host infections, the virus can be continuously detected, at low
cell cytoplasmic membrane. Viral proteins on the outer levels, and either mild or no clinical symptoms may pres-
surface are glycosylated using host enzymes and then dis- ent. By contrast, latent infections are those in which the
place host cell surface proteins and collect into patches. virus persists for extended periods of time in an inactive
Viral nucleocapsids recognize proteins on the inner sur- form or a location not exposed to the immune response.
face of the membrane, where they bind and are engulfed Intermittent flare-ups of clinical disease can occur, during
by the patch area of the membrane. The completed which time infectious virus can be detected. Subclinical
virion exits the cell by budding and release into the extra- infections are those that give no overt sign of their pres-
cellular space. Viral egress can have a variety of effects ence. Humoral and cell-mediated immunity, interferon
on the host cell, ranging from destruction of the cell to and other cytokines, and other host defense factors,
minimal noncytolytic effects. Herpesviruses differ from depending on the type of virus, are common mediators
other enveloped viruses in the manner in which they of recovery, and begin to develop very soon after infec-
form their envelope. The nucleocapsid is formed in the tion. Infiltration with mononuclear cells and lymphocytes
nucleus, and final maturation of the virion occurs only is responsible for the inflammatory reaction in uncom-
on the inner surface of the host cell membrane, forming plicated viral lesions. Virus-infected cells can be lysed by
Kaduse.com
1274 PART III / PHARMACODYNAMIC AGENTS

T lymphocytes through recognition of viral polypeptides Mature HIV virions are spherical and consist of a lipid
on the cell surface, and humoral immunity protects the bilayer membrane surrounding a nucleocapsid that con-
host against reinfection by the same virus. Neutralizing tains genomic RNA, a viral protease, RT, an integrase,
antibodies that are directed against capsid proteins can and some other cellular factors. The HIV life cycle is
prevent viral infection by disrupting viral attachment or depicted in Figure 38.2, and begins when the viral extra-
uncoating. Interestingly, viruses have evolved a variety of cellular protein gp120 attaches to the CD4 receptor on
survival tactics that serve to suppress or evade the host T lymphocytes. Following attachment, the viral envelope
immune response. Because viruses are obligate intracel- and host cell membrane are fused, and the nucleocap-
lular parasites, a method of transmission from one host sid is released into the cytoplasm. The nucleocapsid is
to another is required for survival of the species. Thus, uncoated, and the resulting RNA serves as a substrate
during an active infection, shedding of the infectious for RT, producing a proviral double-stranded DNA that
virion into the environment is a required step in the life migrates to the nucleus and is incorporated into host
cycle of the virus and ensures transmission of the virus to DNA by integrase. This DNA is not expressed in resting
new hosts. Shedding usually occurs at the same site where
the infection was initiated and can occur at various stages
of the disease course.

1. Virus attach and fusion


VIRAL DISEASES (6,7)
HIV
The human immunodeficiency virus (HIV-1) was first RT
identified in 1979 and was found to be the cause of 2. Reverse transcription IN
acquired immunodeficiency syndrome (AIDS) in 1981
(8). Since that time, AIDS had become a serious world-
wide epidemic that continues to expand. The Joint
United Nations Program on HIV/AIDS estimated that by 3. Integration
the end of 2005, a total of 40.3 million people worldwide
were living with HIV/AIDS, the majority having been 4. Transcription
viral transcripts
infected by heterosexual contact. After four decades,
the number of infected individuals has begun to drop
slightly, and that number stands at 33.3 million in 2010. p57, Gag-Pol genomic
Only 5 million of these patients are receiving treatment RNA
for the disease. In 2005, more than 3.1 million people
5. Translation
died of AIDS, and 4.9 million new cases of HIV were diag-
nosed, including more than 700,000 children (9). The
gp160
number of new infections dropped to 2.9 million in 2009,
and annual deaths decreased to 1.8 million (10). The
incidence of the disease varies by location, with sub-Saharan
Africa having the highest incidence of the disease. In
the Third World, HIV infection is often comorbid with PR
neglected tropical diseases (trypanosomiasis, leishmani-
6. Assemble, budding
asis), malaria, and tuberculosis, causing a higher level of and maturation
mortality (11). Because it is sexually transmitted, a high
percentage of infected individuals are young adult work-
FIGURE 38.2 Replicative cycle of HIV. (1) The virus gp120 pro-
ers, and as such, the disease has a significant economic
tein binds to CD4, resulting in fusion of the viral envelope and
impact in some regions. In addition, infected mother-to-
the cellular membrane and the release of viral nucleocapsid into
fetus transmission occurs between 13% and 40% of the
the cytoplasm. (2) The nucleocapsid is uncoated, and viral RNA is
time. Although a variety of drugs have been developed reverse transcribed by reverse transcriptase (RT). (3) The resulting
for treating AIDS patients, none have proven successful double-stranded proviral DNA migrates into the cell nucleus and
in curing the disease. The basic difficulty experienced is integrated into the cellular DNA by integrase (IN). (4) Proviral
with this viral infection is the ability of virus to mutate, DNA is transcribed by the cellular RNA polymerase II. (5) The
leading to rapid drug resistance. mRNAs are translated by the cellular polysomes. (6) Viral proteins
The HIV-1 genome consists of two identical 9.2-kb and genomic RNA are transported to the cellular membrane and
single-stranded RNA molecules within the virion, each assemble. Immature virions are released. Polypeptide precursors
of which contains information for only nine genes. are processed by the viral protease (PR) to produce mature viral
Following infection of the host cell, the persistent form particles. (Used with permission from Tyler KL, Fields BN. Fields
of the HIV-1 genome is proviral double-stranded DNA. Virology, 2nd Ed. New York: Raven Press, 1990:191–239.)
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1275

T lymphocytes, but when the cell is activated, proviral simplex virus (HSV) types 1 and 2, varicella-zoster virus,
DNA is transcribed by host RNA polymerase II. The cytomegalovirus, Epstein-Barr virus, human herpesvi-
viral RNA and proteins are transported to the cell mem- ruses 6 (HHV-6) and 7 (HHV-7), and Kaposi sarcoma–
brane, where they assemble, form a viral bud, and are associated herpesvirus HHV-8 (also known as KSHV).
released from the lymphocyte membrane. This produces Herpesviruses are large viruses that have identical mor-
an immature virion, and processing of the viral surface phology and consist of a core of linear, double-stranded
proteins by HIV protease then affords the mature, infec- DNA surrounded by a protein coat that exhibits icosahe-
tious virus. dral symmetry and has 162 capsomeres. The genome is
About 50% of HIV infections are asymptomatic, large and encodes at least 100 different proteins, includ-
whereas the other 50% produce flu-like symptoms within ing polypeptides involved in viral structure, the viral enve-
4 weeks. During this initial phase, viral titers are very high lope, and enzymes involved in nucleic acid metabolism,
and decline as specific antibodies are formed. The latent DNA synthesis, and protein regulation. Oral cold sores
period is mediated by factors that are not well understood and genital herpes infections are caused by HSV1 and
and can last several years. During this time, viral replica- HSV2, respectively. These viruses can establish a latent
tion continues, and the level of CD4-positive T lympho- infection in the ganglia of nerves that supply the site of
cytes steadily declines. Eventually, the patient immune the primary infection, and the latent disease is reacti-
system becomes compromised, resulting in a variety of vated by a number of stress factors. It is estimated that
opportunistic infections. RT sometimes makes mistakes virtually 100% of adult humans are infected with HSV1,
reading the viral RNA sequence, leading to mutations although many infections are subclinical and asymptom-
in the virus and changes in the structure of the surface atic. The varicella virus is the cause of chickenpox, and
proteins. Thus vaccines, which induce the production of the herpes zoster virus is responsible for shingles. Human
antibodies that recognize and bind to very specific viral cytomegalovirus rarely causes disease in healthy people,
surface molecules, are unlikely to be effective in HIV but when infection occurs in adulthood, it may cause
therapy. As will be discussed below, other viral processes, an infectious mononucleosis-like illness. Primary infec-
such as reverse transcription and proteolytic processing, tion with the Epstein-Barr virus is the cause of infectious
are viable targets for small-molecule therapy. mononucleosis, and this virus is thought to be a factor in
Kaposi sarcoma is a common complication of AIDS the development of Burkitt lymphoma and other malig-
and has been shown to arise from a complex interaction nancies. HHV-6 is thought to cause roseola and mono-
between HIV and the human herpesvirus 8 (HHV-8). nucleosis, whereas HHV-7 is probably not involved in any
This disease presents as a reddish or purple lesion on var- human diseases. The role of HHV-8 in the pathogenesis
ious areas of the skin, and in advanced state may involve of Kaposi sarcoma has been discussed earlier.
the lungs or gastrointestinal tract. HHV-8 does not work
alone, but rather in combination with a patient’s altered Hepatitis (2)
response to cytokines and the HIV-1 transactivating pro- Viral hepatitis is a systemic disease, but primarily involves
tein Tat, which promotes the growth of endothelial cells. the liver. Hepatitis A virus (HAV) is responsible for infec-
HHV-8 can then encode interleukin-6 viral proteins, tious hepatitis, hepatitis B virus (HBV) is associated with
specific cytokines that stimulate cell growth in the skin. serum hepatitis, and hepatitis C virus (HCV) is a common
HHV-8 destroys the immune system further by directing cause of posttransfusion hepatitis. Another viral agent,
a cell to remove the major histocompatibility complex hepatitis E virus (HEV), causes an enterically transmit-
(MHC-1) proteins that protect it from invasion. These ted form of hepatitis. On occasion, disease can arise from
proteins are then transferred to the interior of the cell hepatic infection with yellow fever virus, cytomegalovirus,
and are destroyed, leaving the cell unguarded and vul- Epstein-Barr virus, herpes simplex virus, rubella virus,
nerable to pathogens that would normally be cleared by and the enteroviruses. Viral hepatitis usually involves
the immune system. It has recently been discovered that acute inflammation of the liver, fever, nausea, vomiting,
xCT, the 12-transmembrane light chain of the human and jaundice, and all forms of hepatitis produce identi-
cystine/glutamate exchange transporter system, serves cal histopathologic lesions in the liver during acute dis-
as a receptor for internalization of HHV-8 (12). ease. HAV is a member of the picornavirus family and
carries a single-stranded RNA genome; only one strain of
Herpesvirus (2) the virus exists. The onset of HAV hepatitis occurs within
The herpesvirus family contains several of the most 24 hours, in contrast to the slower onset of clinical symp-
important human pathogens, which are responsible for toms with HBV and HCV infection. Complete recovery
causing a spectrum of common diseases. A common and occurs in most HAV cases, and chronic infection never
significant feature of the herpesviruses is their ability occurs. HBV is classified as a hepadnavirus with a double-
to establish lifelong persistent infections in their hosts stranded circular DNA genome. The outcome of HBV
and to undergo periodic reactivation. Herpesviruses pos- infection ranges from complete recovery to progression
sess a large number of genes, and some of the resulting to chronic hepatitis and, rarely, death. HBV establishes
proteins are targets for antiviral chemotherapy. The her- chronic infections, especially in infants; 80% to 95% of
pesviruses that commonly infect humans include herpes infants and young children infected with HBV become
Kaduse.com
1276 PART III / PHARMACODYNAMIC AGENTS

chronic carriers and are at high risk of developing hepa- NA is an enzyme that removes sialic acid from surface
tocellular carcinoma. In adults, 65% to 80% of infections glycoproteins during viral maturation and is required
are asymptomatic, and 90% to 95% of all patients recover to produce infectious particles and lower the viscosity
completely. HCV is a positive-stranded RNA flavivirus of the mucin layer of the respiratory tract. Influenza
and exists in at least six major genotypes. Most cases of virus spreads through airborne droplets or contact with
posttransfusion hepatitis are caused by HCV, and these contaminated hands or surfaces and has an incubation
infections are usually subclinical with minor elevation of period that varies from 1 to 4 days. However, transmission
liver enzymes and a low incidence of jaundice. However, of the virus begins to occur 24 hours prior to the onset of
70% to 90% of HCV patients develop chronic hepatitis, symptoms. Interferon is detectable in respiratory secre-
and many are at risk of progressing to chronic active tions at the onset of symptoms, and the host response
hepatitis and cirrhosis decades later. HEV is transmitted to interferon contributes to recovery. Antibodies and
enterically and occurs in epidemic form in developing other cell-mediated responses are seen 1 to 2 weeks after
countries, where water supplies are sometimes contam- infection. It is well established that secondary infections
inated with feces. The disease is more severe in adults from other viruses or bacteria can occur, and Reye syn-
than in children, who are usually asymptomatic. drome, an acute encephalopathy occurring in children
and adolescents, is a rare complication of influenza B,
Influenza (2) influenza A, and herpesvirus varicella-zoster infections.
Respiratory illnesses commonly known as colds and flu The chances of contracting Reye syndrome are increased
account for more than half of all acute illnesses in the when salicylates are used in children suffering from influ-
United States each year. Influenza viruses belong to enza and related respiratory diseases.
the Orthomyxoviridae family and are a major source of
morbidity and mortality caused by respiratory disease. Tumor Viruses (2,13–17)
Outbreaks of infection can occur in worldwide epi- Viruses are etiologic factors in the development of sev-
demics that have resulted in millions of deaths world- eral types of human tumors, most notably cervical can-
wide. Genetic mutations often cause antigenic changes cer and liver cancer. At least 15% of all human tumors
in the structure of viral surface glycoproteins, making worldwide have a viral cause. Tumor viruses can be found
influenza viruses extremely difficult to control. Three in both the RNA and DNA virus kingdoms (18). The
immunologic types of influenza viruses are known and list of human viruses presently known to be involved in
are termed influenza A, B, and C. Antigenic changes are tumor development includes four DNA viruses (Epstein-
very common in the type A group of influenza viruses, Barr virus [EBV], certain papilloma viruses, HBV, and
which are responsible for the majority of influenza epi- the Kaposi sarcoma herpesvirus HHV-8) and two RNA
demics. Influenza type B undergoes more infrequent viruses, (human T-cell leukemia virus 1 [HTLV-1] and
antigenic changes and is less often the cause of an influ- HCV). Nearly every case of cervical cancer is known to
enza epidemic, whereas influenza type C is antigenically be caused by human papillomavirus (HPV), a fact that
stable and causes only mild illness in immunocompetent spurred the development of the HPV vaccines Gardasil
individuals. The viruses carry a single-stranded, negative- and Cervarix (19). Tumor viruses alter cellular behav-
sense RNA genome that has eight segments in influenza ior through the use of a small amount of genetic infor-
A and B. Influenza C viruses contain only seven segments mation, using two general strategies. The tumor virus
of RNA and lack a neuraminidase gene (see below). The either introduces a new “transforming gene” into the cell
complete virion in each type contains nine different (direct-acting), or the virus alters the expression of a pre-
structural proteins. A nucleoprotein associates with viral existing cellular gene or genes (indirect-acting). In both
RNA to form a ribonucleoprotein structure that makes cases, normal regulation of cellular growth processes is
up the viral nucleocapsid. Three other large proteins are lost. Viruses alone cannot act as carcinogens, and other
bound to the viral ribonuclear protein and are respon- events are necessary to disable regulatory pathways and
sible for RNA transcription and replication. A matrix checkpoints in order to produce transformed, malignant
protein is also included in the virion that forms a shell cells. The processes used in the transformation of host
underneath the viral lipid envelope and comprises about cells by human tumor viruses are very diverse.
40% of all viral protein. Cellular transformation may be defined as a stable,
The influenza virion structure also includes a lipid heritable change in the growth control of cells that
envelope derived from the host cell. This envelope con- results in tumor formation. Transformation from a nor-
tains two viral surface glycoproteins called hemagglutinin mal to a neoplastic cell generally requires the retention
(HA) and neuraminidase (NA). Mutations cause anti- of viral genes in the host cell. In the majority of cases,
genic changes in the structure of these two surface gly- this is accomplished by the integration of certain viral
coproteins, and thus, they are the main determinants of genes into the host cell genome. Retroviruses incorpo-
antigenicity and host immunity. The ability of the virus to rate their proviral DNA, formed through the action of
change the structure of HA on the virus surface is primar- reverse transcriptase, into host cell DNA. By contrast,
ily responsible for the continual evolution of new strains, DNA tumor viruses integrate a portion of the DNA of the
sometimes leading to subsequent influenza epidemics. viral genome into the host cell chromosome.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1277

All RNA tumor viruses are members of the retrovi- features. Following exposure, prions accumulate in
rus family and belong to one of two classes (14,15,17). lymphoid tissue such as the spleen, lymph nodes, and
Class I RNA viruses are direct-transforming and carry tonsils and in Peyer’s patches (specialized lymphoid fol-
an oncogene obtained through accidental incorpora- licles located in the submucosa of the small intestine).
tion from the host cell. No class I RNA viruses are known This accumulation of the infectious agent is necessary
to produce tumors in humans. Class II or chronic RNA for invasion of the CNS. In humans, the incubation
tumor viruses are weakly transforming and do not carry period of the disease can vary between 18 months and
host cell–derived oncogenes. The two known cancer- 40 years. Prions appear to be variant, improperly folded
causing retroviruses in humans act indirectly. They often versions of a normal cellular protein called PrPc, a 30-
act by inserting their proviral DNA into the immediate to 35-kd protein with two sites for N-glycosylation that is
neighborhood of a host cellular oncogene. The human anchored in the neuronal cell membrane. PrPc protein
adult T-cell leukemia virus HTLV-1 acts in this man- contains three α-helices, a short β-pleated sheet region,
ner, thus increasing the number of preneoplastic cells and a long, unstructured portion that comprises half of
and facilitating secondary cellular changes leading to the molecule. The variant, infectious form of the protein
transformation. is known as PrPsc and is produced autocatalytically from
DNA tumor virus strains exist among the papilloma-, PrPc. Prion diseases are always fatal, with no known cases
polyoma-, adeno-, herpes-, hepadna-, and poxvirus groups of remission or recovery. The host shows no inflamma-
(16). DNA tumor viruses encode viral oncoproteins that tory response or immune response and no production
are important for viral replication, but also affect cellular of interferon, and there is no effect on host B-cell or
growth control pathways. For example, inactivation of Rb T-cell function. At present, there are no effective agents
and the p53 pathway by viral transforming proteins is a to treat prion diseases.
common strategy used by papillomaviruses and adenovi-
ruses. As mentioned earlier, all DNA tumor viruses kill
their host cell when the infectious virion is released to VIRAL CHEMOTHERAPY
infect other cells. Thus, transformation and tumorigenic- General Approaches (23,24)
ity are entirely dependent on a host cell interaction with
The principles involved in the design of antiviral agents
the virus that does not involve viral spread to other cells,
are similar to those used in the design of all chemothera-
and cells transformed by DNA tumor viruses depend
peutic agents. Drugs in this category are targeted to some
on the continued expression of the virally encoded
process in the virus that is not present in the host cell.
oncogene.
The earliest examples of antiviral agents did not achieve
Recent studies have revealed that the human tumor
this goal, and these drugs were toxic at therapeutic lev-
viruses EBV, HHV-8, HPV, HBV, HCV, and HTLV-1
els or had a limited spectrum of activity. A variety of fac-
express proteins that are targeted to the mitochondria
tors make the design of effective antiviral agents difficult,
(13). Because the mitochondria play a critical role in
including their ability to undergo antigenic changes, the
energy production, cell death, calcium homeostasis,
latent period during which there are no symptoms, and
and redox balance, these proteins have profound effects
their reliance on host enzymes and other processes. This
on host cell physiology. Further study of these proteins
problem is compounded by the fact that host immunity
and their interactions with mitochondria will aid in the
is not well understood and that symptoms of viral infec-
understanding of the mechanisms of viral replication
tion may not appear until replication is complete and
and tumorigenesis and could reveal important new tar-
the viral genome has been incorporated into infected
gets for antitumor therapy.
cells. Nonetheless, the continuing identification of new
Prion Diseases (20–22) targets for antiviral agents provides new avenues for the
discovery of small-molecule therapies. The following sec-
Although they are not viruses, the infective proteins tion includes information on currently marketed antivi-
known as prions have sufficient similarities to viruses ral compounds that have been designed in eight general
to warrant their discussion in this chapter. Prions are areas:
small proteins that have been shown to cause a variety
of transmissible spongiform encephalopathies, which 1. Agents that disrupt virus attachment to host cell
are rare neurodegenerative disorders typified by symp- receptors, penetration, or uncoating.
toms in the central nervous system (CNS) such as spon- 2. Agents that inhibit virus-associated enzymes such
giform changes, neuronal loss, glial activation, and the as DNA polymerases and others.
accumulation of amyloid aggregates of an abnormally 3. Agents that inhibit viral transcription.
folded host protein. Human prion diseases include 4. Agents that inhibit viral translation.
kuru, Creutzfeldt-Jakob disease (variant, sporadic, famil- 5. Agents that interfere with viral regulatory proteins.
ial, and iatrogenic), Gerstmann-Sträussler-Scheinker 6. Agents that interfere with glycosylation, phos-
syndrome, and fatal familial insomnia. The disease in phorylation, sulfation, and so on.
cattle known as bovine spongiform encephalopathy and 7. Agents that interfere with the assembly of viral
the related disease scrapie exhibit similar pathologic proteins.
Kaduse.com
1278 PART III / PHARMACODYNAMIC AGENTS

8. Agents that prohibit the release of viruses from broad-spectrum antiviral activity, completely inhibit
cell surface membranes. viral replication, maintain efficacy against mutant viral
strains, and reach the target organ without interfering
The remainder of this chapter deals primarily with small-
with normal cellular processes or the immune system
molecule antiviral agents that have been approved by
of the host.
the U.S. Food and Drug Administration (FDA) and are
clinically effective in the treatment of viral infection.
Immunizing biologic agents such as vaccines, as well
as antineoplastic agents with antiviral activity, are not ANTIVIRAL AGENTS (23,24)
covered. Some compounds used primarily in the treat-
ment of bacterial infections, such as rifampicin, bleo-
Agents Inhibiting Virus Attachment, Penetration,
mycin, doxorubicin, and actinomycin, also inhibit viral and Early Viral Replication (Table 38.2)
replication. However, these antibiotics do not affect Amantadine
the transcription or translation of viral mRNA and are MECHANISM OF ACTION Amantadine hydrochloride
only effective in high concentrations. Therefore, such (1-adamantanamine hydrochloride) is a symmetrical tri-
antibiotics are not commonly used for viral infections. cyclic primary amine that inhibits penetration of RNA
There is a continuing need for new antiviral agents, virus particles into the host cell (Fig. 38.3)(25). It also
primarily because viral infections are not curable after inhibits the early stages of viral replication by blocking
the virus invades the host cell and begins to replicate. the uncoating of the viral genome. Recent studies suggest
Vaccines are effective, but they are only able to pre- that amantadine inhibits viral replication by blocking the
vent an infection, and only in cases where specific influenza A virus M2 proton-selective ion channel.
virus strains are involved. For example, immunization
against influenza, which is a yearly routine in many CLINICAL APPLICATION Amantadine is clinically effective
parts of the world, can only provide immunity against in preventing and treating all A strains of influenza,
specific strains that are represented in that prepara- particularly A2 strains of Asian influenza virus and, to
tion. New virulent strains may arise from nonhuman a lesser extent, German measles (rubella) or togavirus.
sources, such as the so-called swine flu or avian flu It also shows in vitro activity against influenza B, para-
viruses, and currently available vaccines would have no influenza (paramyxovirus), respiratory syncytial virus
effect against these new strains. With regard to small- (RSV), and some RNA viruses (murine, Rous, and Esh
molecule antiviral agents, the ideal drug would have sarcoma viruses). Many prototype influenza A viruses

TABLE 38.2 Antiviral Agents Interfering with Cellular Penetration and Early Replication
Generic Name Trade Name Spectrum of Activity Dosage Form

Amantadine Symmetrel Influenza A Cap (100 mg), syrup (50 mg/5 mL)

Rimantadine Flumadine Influenza A Cap (100 mg)

Interferon α-2a Roferon A Chronic hepatitis, CMV, HSV, Injectable (3, 5, 10, 18, 25, and 50
Alferon papillomavirus, rhinovirus, million units/mL)
Intron and others
Wellferon

Interferon α-2b Interon A Chronic hepatitis B and C, Injectable (3, 5, 10, 18, 25, and 50
many other viruses million units/mL)

γ-Interferon Actimmune Chronic hepatitis B and C, Injectable (100 mcg/0.5 mL)


many other viruses

Tilorone Amixin IC Induces the production of Variable single doses given by


interferons injection

Zanamivir Relenza Influenza A and B Inhaled powder (5 mg)

Oseltamivir Tamiflu Influenza A and B Cap (75 mg)

Peramivir Experimental H1N1 600 mg daily by IV administration

Enfuvirtide Fuzeon HIV Adult: 90 mg SQ BID


Age 6–16: 2 mg/kg SQ BID

Maraviroc Selzentry HIV 300 mg orally twice daily

BID, twice a day; Cap, capsule; SQ, subcutaneous.


Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1279

NH2
H2N CH3 for 1 week after influenza A vaccination. A virus resis-
tant to amantadine has been obtained in cell culture
HCl HCl
and from animals, but these reports are not confirmed
in humans.
Amantadine Rimantadine
Rimantadine
FIGURE 38.3 Agents that inhibit virus attachment, penetration, MECHANISM OF ACTION Rimantadine hydrochloride
and early viral replication. (α-methyl-1-adamantanemethylamine hydrochloride)
is a synthetic adamantane derivative that is structurally
and pharmacologically related to amantadine (Fig. 38.3)
of different human subtypes (H1N1, Fort Dix, H2N2, (27,28). It appears to be more effective than amantadine
Asian type, and H3N2, Hong Kong type) are also inhib- hydrochloride against influenza A virus with fewer CNS
ited by amantadine hydrochloride in vitro and in ani- side effects. Rimantadine hydrochloride is thought to
mal model systems. If given within the first 48 hours of interfere with virus uncoating by inhibiting the release
onset of symptoms, amantadine hydrochloride is effec- of specific proteins. It may act by inhibiting RT or the
tive in respiratory tract illness resulting from influenza synthesis of virus-specific RNA but does not inhibit virus
A but not influenza B virus infection, adenoviruses, adsorption or penetration. It appears to produce a viru-
and RSV. static effect early in the virus replication. It is used widely
in Russia and Europe.
PHARMACOKINETICS Amantadine is well absorbed orally,
and the usual dosage for oral administration is 100 mg CLINICAL APPLICATION Rimantadine hydrochloride has
twice daily. A 100-mg oral dose produces blood serum activity against most strains of influenza A including
levels of 0.3 mg/mL within 1 to 8 hours. Maximum tis- H1N1, H2N2, and H3N2 but has no activity against influ-
sue concentration is reached in 48 hours when a 100-mg enza B virus. It is used for prevention of infection caused
dose is given every 12 hours. In healthy adults receiv- by various human, animal, or avian strains of influenza A
ing 25-, 100-, and 150-mg doses of the drug twice daily, virus in adults and children. The side effects are night-
steady-state trough plasma concentrations were 110, 302, mares, hallucinations, and vomiting. The most common
and 588 mg/mL, respectively. Usually, no neurotoxic- side effects of rimantadine are associated with the CNS
ity is observed if the plasma level of amantadine is no and GI tract.
more than 1.00 mg/mL. Amantadine crosses the blood–
brain barrier and is distributed in saliva, nasal secretions, PHARMACOKINETICS Rimantadine is metabolized in the
and breast milk (26). Approximately 90% of the drug is liver, and about 90% is excreted in the urine within
excreted unchanged by the kidney, primarily through 24 hours. The major urinary metabolites are ring-
glomerular filtration and tubular secretion, and there hydroxylated derivatives, a small percentage of which
are no reports of metabolic products. Acidification of are glucuronidated. The half-life of rimantadine in
urine increases the rate of amantadine excretion. The adults ranges from 24 to 36 hours. Over 90% of riman-
half-life of the drug is 15 to 20 hours in patients with nor- tadine doses are absorbed in 3 to 6 hours. Steady-state
mal renal function. plasma concentrations range from 0.10 to 2.60 mg/mL
at doses of 3 mg/kg/d in infants to 100 mg twice daily
SIDE EFFECTS Generally, the drug has low toxicity at ther- in the elderly. Nasal fluid concentrations of rimanta-
apeutic levels but may cause severe CNS symptoms such dine at steady-state were 1.5 times higher than plasma
as nervousness, confusion, headache, drowsiness, insom- concentration.
nia, depression, and hallucinations. Gastrointestinal
(GI) side effects include nausea, diarrhea, constipation, Interferon (29,30)
and anorexia. Convulsions and coma occur with high Isaacs and Lindenmann discovered interferon in 1957.
doses and in patients with cerebral arteriosclerosis and When they infected cells with viruses, interference
convulsive disorders. Chronic toxicity with amantadine with the cellular effects of viral infection was observed.
is unexpected since few side effects have been expe- Interferon was subsequently isolated and found to pro-
rienced when the drug has been used for long-term tect the cells from further infection. When interferon
therapy for Parkinson disease. Some serious reactions, was administered to other cells or animals, it displayed
however, include depression, orthostatic hypotension, biologic properties such as inhibition of viral growth,
psychosis, urinary retention, and congestive heart fail- cell multiplication, and immunomodulatory activities.
ure. Amantadine hydrochloride should be used with The results led to the speculation that interferon may
caution in patients who have a history of epilepsy, severe be a natural antiviral factor, possibly formed before anti-
arteriosclerosis, liver diseases, and eczematoid derma- body production, and may be involved in the normal
titis. Because amantadine does not appear to interfere mechanism of resistance displayed against viral infec-
with the immunogenicity of inactivated influenza A virus tion. Some investigators relate interferon to the poly-
vaccine, patients may continue the use of amantadine peptide hormones and suggest that interferon functions
Kaduse.com
1280 PART III / PHARMACODYNAMIC AGENTS

in cell-to-cell communication by transmitting specific (skin cancer), and Kaposi sarcoma (cancer associated
messages. Recently, antitumor and anticancer proper- with AIDS). Both β- and γ-interferons and interleukin-2
ties of interferon have evoked worldwide interest in the may be commercial drugs of the future for the treatment
possible use of this agent in therapy for viral diseases, of cancers and viral infections, including genital warts
cancer, and immunodeficiency disorders. Host cells in and the common cold.
response to various inducers synthesize interferons.
PHARMACOKINETICS The pharmacokinetics of interferon
INTERFERON STRUCTURE Interferon consists of a mixture are not well understood. Maximum levels in blood after
of small proteins with molecular weights ranging from intramuscular injection were obtained in 5 to 8 hours.
20,000 to 160,000 daltons. They are glycoproteins that Interferon does not penetrate well into cerebrospinal
exhibit species-specific antiviral activity. Human interfer- fluid (CSF). Oral administration of interferon does
ons are classified into three types: alpha (α), beta (β), not indicate a detectable serum level, and as such, oral
and gamma (γ) (31). The α-type is secreted by human administration is clinically ineffective. After intramus-
leukocytes (white blood cells, non–T lymphocytes), and cular or subcutaneous injection, drug concentration in
the β-type is secreted by human fibroblasts. Lymphoid the plasma is dose related. Clinical use of interferon is
cells (T lymphocytes), which either have been exposed limited to topical administration (nasal sprays) for pro-
to a presensitized antigen or have been stimulated to phylaxis and treatment of rhinovirus infections. Adverse
divide by mitogens, secrete α-interferon. γ-Interferon is reactions and toxicity include influenza-like syndrome of
also called “immune” interferon. Interferons are active fever, chills, headache, myalgias, nausea, vomiting, diar-
in extremely low concentrations. rhea, bone marrow suppression, mental confusion, and
behavioral changes. Intranasal administration produces
MECHANISM OF ACTION Although, interferons are mediators mucosal friability, ulceration, and dryness. Two long-
of immune response, different mechanisms for the antiviral acting forms of interferon have recently become available.
action of interferon have been proposed. α-Interferon The addition of polyethylene glycol to the formulation of
possesses broad-spectrum antiviral activity, and acts on interferon α-2a and interferon α-2b affords an injectable
virus-infected cells by binding to specific cell surface form of each drug that has an extended half-life.
receptors. It inhibits the transcription and translation of Because viruses were found to induce the release of
mRNA into viral nucleic acid and protein. Studies in cell interferon, efforts were made to induce the produc-
free systems have shown that the addition of adenosine tion or release of interferon in humans by the admin-
triphosphate and double-stranded RNA to extracts of istration of chemical “inducers” (34). Various small
interferon-treated cells activates cellular RNA proteins molecules (substituted propanediamine) and large poly-
and a cellular endonuclease. This activation causes the mers (double-stranded polynucleotides) were used to
formation of translation inhibitory protein, which termi- induce interferons. Statolon, a natural double-stranded
nates production of viral enzyme, nucleic acid, and struc- RNA produced in Penicillium stoloniferum culture, and a
tural proteins (32). Interferon may also act by blocking double-stranded complex of polyriboinosinic acid and
synthesis of a cleaving enzyme required for viral release. polyribocytidylic acid (poly I:C) have been used as non-
viral inducers for releasing preformed interferons. A
CLINICAL APPLICATION Interferon has been tested for use in modification of poly I:C stabilized with poly-l-lysine and
chronic HBV infection, herpetic keratitis, herpes genitalis, carboxymethylcellulose (poly ICLC) has been used exper-
herpes zoster, varicella-zoster, chronic hepatitis, influenza, imentally in humans. Clinically, it prevented coryza when
and common cold infections. Other uses of interferon are used locally in the nose and conjunctival sacs. This sub-
in the treatment of cancers, such as breast cancer, lung car- stance was found to be a better interferon inducer than
cinoma, and multiple myeloma. Interferon has had some poly I:C. Another interferon inducer is Ampligen, a poly-
success when used as a prophylactic agent for cytomegalo- nucleotide derivative of poly I:C with spaced uridines. It
virus (CMV) infection in renal transplant recipients. The has anti-HIV activity in vitro and is an immunomodulator.
scarcity of interferon and the difficulty in purifying it have Other chemical inducers, such as pyran copolymers,
limited clinical trials. Supplies have been augmented by tilorone, diethylaminoethyl dextran, and heparin, have
recombinant DNA technology, which allows cloning of also been used. Tilorone is an effective inducer of inter-
the interferon gene (33), although the high cost still hin- feron in mice but is relatively ineffective in humans.
ders clinical application. The FDA has approved recom- Initial use of interferon and its inducers instilled intrana-
binant interferon α-2a and α-2b and γ-interferon for the sally after rhinovirus exposure was successful in the pre-
treatment of hairy cell leukemia (a rare form of cancer), vention of respiratory diseases.
AIDS-related Kaposi sarcoma, and genital warts (condy-
O
loma acuminatum). Subcutaneous injection of recombi-
nant interferon α-2b has been approved for the treatment N O O N
of chronic HCV. Some foreign countries have approved
α-interferon for the treatment of cancers such as multiple
myeloma (cancer of plasma cells), malignant melanoma Tilorone
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1281

The clinical success of interferon and its inducers has not potential means of inhibiting the spread of viral infections.
yet been established, although they may play a significant X-ray crystallography of NA has shown that while the
role in cell-mediated immunity to viral infections and amino acid sequence of NA from various viruses is con-
cancer. Disadvantages of interferon use include unac- siderably different, the sialic acid binding site is quite
ceptable side effects, such as fever, headache, myalgias, similar for type A and B influenza viruses. In addition,
leukopenia, nausea, vomiting, diarrhea, hypotension, it is believed that the hydrolysis of sialic acid proceeds
alopecia, anorexia, and weight loss. through an oxonium cation–stabilized carbonium ion
as shown in Figure 38.4. Mimicking the transition state
Neuraminidase Inhibitors (35) with novel carbocyclic derivatives of sialic acid has led
Virus Activation to the development of transition state–based inhibitors
(36). The first of these compounds, 2-deoxy-2,3-dehydro-
The role played by the surface glycoproteins HA, an
N-acetylneuraminic acid (DANA; Fig. 38.5), was found to
enzyme important for viral binding to host cell recep-
be an active neuraminidase inhibitor but lacked specific-
tors via a terminal sialic acid residue, and NA, an enzyme
ity for viral neuraminidase. Upon determination of the
involved in various aspects of activation of influenza
crystal structure of neuraminidase, more sophisticated
viruses, was discussed earlier. Freshly shed virus particles
measurements of the binding site for sialic acid led to the
are coated with sialic acid residues. NA is found in both
development of zanamivir and, later, oseltamivir.
influenza A and B viruses and is thought to be involved
in catalytically cleaving glycosidic bonds between terminal Specific Drugs
sialic acid residues and adjacent sugars on HA. The cleav-
ZANAMIVIR Crystallographic studies of DANA bound to
age of sialic acid bonds facilitates the spread of viruses by
NA defined the receptor site to which the sialic acid por-
enhancing adsorption to cell surface receptors, and thus
tion of the virus binds. These studies suggested that substi-
increases the infective level of the virus. In the absence of
tution of the 4-hydroxy with an amino group or the larger
sialic acid cleavage from HA, viral aggregation or inappro-
guanidino group should increase binding of the inhibitor
priate binding to HA will occur, interfering with the spread
to NA. The 4-amino derivative was found to bind to a glu-
of the infection. NA also appears to play a role in prevent-
tamic acid (Glu 119) in the receptor through a salt bridge,
ing viral inactivation by respiratory mucus (Fig. 38.4).
whereas the 4-guanidino derivative was able to form both
Mechanism of Action a salt bridge to Glu 119 and a charge–charge interaction
with a glutamic acid at position 227. The result of these
Since NA plays such an important role in the activa-
substitutions was a dramatic increase in binding capacity
tion of newly formed viruses, it is not surprising that the
of the 4-amino and 4-guanidino derivatives to NA, lead-
development of NA inhibitors has become an important
ing to effective competitive inhibition of the enzyme. The
result has been the development of the 4-guanidino ana-
HO
log as zanamivir (Fig. 38.5), which has become an effec-
OH COOH tive agent against influenza A and B virus.
HO
H3C NH O O sugar-GP
HO
O HO OH HO OH
NA H
1 H
HO H 6 O HO H O
N COOH N COOH
H3C C 5 2 H 3C C
HO O HO 4 3
O HN
HO
OH
HO COOH OH HN NH2
H3C NH O HO O COOH HO
HO H3C NH DANA Zanamivir
O HO OH
O HO
O
Transition state H2N
COOH
OH
HO

Sialic acid
HO
OH COOH OH
HO O H H
H3C NH O O H + Sugar GP HN COOH
HO HN COOC2H5
H3C O
O
H3C HN
O H2N H3PO4 NH
N-Acetylsialic acid H2N

FIGURE 38.4 Neuraminidase-catalyzed removal of a sialic acid Peramivir


Oseltamivir phosphate
residue from a glycoprotein chain. GP, glycoprotein; NA, neur-
aminidase. FIGURE 38.5
Kaduse.com
Neuraminidase inhibitors based on sialic acid.
1282 PART III / PHARMACODYNAMIC AGENTS

Zanamivir is effective when administered via the nasal, O H O H


intraperitoneal, and intravenous routes but is inactive
HN COOC2H5 HN COOH
when given orally (Table 38.2). Animal studies have shown H3 C H3 C
68% recovery of the drug in the urine following intra- O H 2N O H2N
peritoneal administration, 43% urinary recovery follow- Oseltamivir Active metabolite
ing nasal administration, and only 3% urinary recovery
following oral administration. Human data gave similar ω-Oxidation
results to those obtained in animal models. Human effi-
cacy studies with nasal drops or sprays demonstrated that HO HO
the drug was effective when administered before exposure
and after exposure to influenza A or B virus. When given O H O O H
before viral inoculation, the drug reduced viral shedding, HN COOC2H5 HN COOC2H5
H3 C H3C
infection, and symptoms. When administered beginning
O H 2N O H2N
at either 26 or 32 hours after inoculation, there was a
reduction in shedding, viral titer, and fever. Presently the FIGURE 38.6 Metabolism of oseltamivir by hydrolysis and
drug is available as a dry powder for oral inhalation by ω-oxidation.
adults and adolescents who have been symptomatic for
no more than 2 days. Zanamivir is able to more rapidly
resolve influenza symptoms and improve recovery (from PERAMIVIR In 2009, the experimental NA inhibitor peramivir
7 days with placebo to 4 days with treatment). Additional (Fig. 38.5, Table 38.2) received emergency FDA approval
studies have suggested the prophylactic benefit of zanami- for use in treatment of certain hospitalized patients with
vir when administered to family members after one mem- known or suspected H1N1 influenza infections. This rep-
ber of the family developed flu-like symptoms. As a result, resents the first time in which an experimental drug in
the manufacturer has submitted an application for the phase III clinical trials has received an emergency use
use of the drug for the prevention of influenza A and B. approval. The drug is only available by electronic request
from the U.S. Strategic National Stockpile. The use of per-
OSELTAMIVIR X-ray crystallographic studies further dem- amivir is restricted to severe H1N1 influenza cases where
onstrated that additional binding sites exist between other antiviral agents (including NA inhibitors) have
NA and substrate involving the C-5 acetamido carbonyl failed (38). For example, a patient infected with an H1N1
of DANA and an arginine (Arg 152), the C-2 carboxyl strain of influenza that is resistant to oseltamivir and who is
and arginines as 118, 292, and 371, and the potential for unable to inhale zanamivir would be a candidate for pera-
hydrophobic binding to substituents at C-6 (with glutamic mivir treatment. Peramivir is structurally distinct from osel-
acid, alanine, arginine, and isoleucine). Structure–activity tamivir and zanamivir in that it contains a cyclopentane
relationship studies showed that maximum binding ring structure that replaces the sialic acid core and because
occurred to NA when C-6 (based on DANA numbering) it does not contain a glycerol side chain, which is thought
was substituted with the 3-pentyloxy side chain such as to reduce the oral bioavailability of oseltamivir and zanami-
the one found in oseltamivir (Fig. 38.5). In addition, vir. The structure of peramivir was proposed using the tech-
esterification with ethanol gave rise to a compound that nique of structure-based design. In this approach, in silico
is orally effective. Oseltamivir phosphate was approved docking of proposed NA inhibitors suggested that perami-
as the first orally administered NA inhibitor used against vir bound most efficiently to the four known binding pock-
influenza A and B (Table 38.2). The drug is indicated for ets in the viral NA catalytic site. Peramivir is administered
the treatment of uncomplicated acute illness due to influ- intravenously as a single dose (0.5 to 8 mg/kg) or as two
enza infection. Recently, the manufacturer has submitted daily doses (4 mg/kg). It has a serum half-life of 7.7 to
a request for approval of the drug for prevention of influ- 20 hours, and 90% of the drug is excreted unchanged in
enza A and B for use in adults, adolescents, and children the urine. Because peramivir is still in phase III clinical tri-
1 year of age and older. The drug is effective in treat- als, efficacy data have not yet been released.
ing influenza if administered within 2 days after onset of
symptoms. The recommended dose is 75 mg twice daily Entry (Fusion) Inhibitors
for 5 days. The prophylactic dose is 75 mg taken once Enfuvirtide (39,40)
daily for 7 days. Oseltamivir is readily absorbed from the
N-acetyl-Tyr-Thr-Ser-Leu-Ile-His-Ser-Leu-Ile-Glu-Glu-Ser-Gln-Asp-
GI tract following oral administration. It is a prodrug Gln-Gln-Glu-Lys-Asp-Glu-Gln-Glu-Leu-Leu-Glu-Leu-Asp-Lys-Try-Ala-
that is extensively metabolized in the liver undergoing Ser-Leu-Try-Asp-Try-Phe-NH2
ester hydrolysis to the active carboxylic acid (Fig. 38.6).
Two oxidative metabolites have also been isolated, with Enfuvirtide
the major oxidation product being the ω-carboxylic acid
(37). Side effects with oseltamivir are minor, consisting Entry inhibitors, also known as fusion inhibitors, are
of nausea and vomiting that occur primarily in the first a new class of drugs for the treatment of HIV infec-
2 days of therapy. tion, and enfuvirtide is the first compound of this
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1283

family to be approved for clinical use. Enfuvirtide is an (∼23%) and is a substrate for P-glycoprotein, possibly
oligopeptide consisting of 36 amino acids. It is a syn- accounting for the poor bioavailability. The drug is highly
thetic peptide that mimics an HR2 fragment of gp41, bound to plasma proteins (∼76%) and is metabolized to
blocking the formation of a six-helix bundle struc- inactive metabolites via CYP3A4. The major metabolite
ture that is critical in the fusion of the HIV-1 virion results from N-dealkylation.
to a CD4-positive T lymphocyte. Specifically, it binds
to the tryptophan-rich region of the gp41 protein.
Agents Interfering with Viral Nucleic Acid
Enfuvirtide is used in combination with other antiret-
Replication (Table 38.3)
rovirals and works against a variety of HIV-1 variants,
but it is not active against HIV-2. Resistance to enfu- Acyclic Nucleoside Analogs (Fig. 38.7)
virtide can develop when the virus produces changes ACYCLOVIR
in a 10–amino acid domain between residues 36 to Mechanism of Action Acyclovir is a synthetic analog of
45 in the gp41 HIV surface glycoprotein. The drug is deoxyguanosine in which the carbohydrate moiety is acy-
administered twice daily as a subcutaneous injection clic (Fig. 38.7) (41). Because of this difference in structure
and has a complex absorption pattern. Enfuvirtide is as compared to other antiviral compounds (idoxuridine,
highly bound to plasma protein (∼92%) and is prone vidarabine, and trifluridine, see below), acyclovir pos-
to proteolytic metabolism. Adverse reactions include sesses a unique mechanism of antiviral activity. The mode
pain, erythema, and pruritus at the site of injection of action of acyclovir consists of three consecutive mecha-
and insomnia. nisms (42). The first of these mechanisms involves conver-
sion of the drug to active acyclovir monophosphate within
F F cells by viral thymidine kinase (Fig. 38.8). This phosphory-
lation reaction occurs faster within cells infected by her-
H H H pesvirus than in normal cells because acyclovir is a poor
N
H3C H
N
H3C substrate for the normal cell thymidine kinase. Acyclovir
O N CH3 CH3
H is further converted to di- and triphosphates by a normal
H N H N
N N cellular enzyme called guanosine monophosphate kinase.
N N
H3C H3C In the second mechanism, viral DNA polymerase is com-
petitively inhibited by acyclovir triphosphate with a lower
Maraviroc Maraviroc metabolite half maximal inhibitory concentration (IC50) concentra-
tion than that for cellular DNA polymerase. Acyclovir
Maraviroc triphosphate is incorporated into the viral DNA chain dur-
CCR5 is a protein produced by humans that is a member ing DNA synthesis. Because acyclovir triphosphate lacks
of the chemokine coreceptor family and is located in the the 3’-hydroxyl group of a cyclic sugar, it terminates fur-
cell membrane of specific human cells (i.e., T cells, mac- ther elongation of the DNA chain. The third mechanism
rophages, dendritic cells, microglia). Although there are a depends on preferential uptake of acyclovir by herpes-
number of natural substrates for this receptor protein, HIV infected cells as compared to uninfected cells, resulting
can use this G protein–coupled receptor in part as a core- in a higher concentration of acyclovir triphosphate and
ceptor for entry into its targeted cells. In addition, indi- leading to a high therapeutic index between herpes-
viduals who have a mutation in the CCR5 gene (CCR5-δ32) infected cells to normal cells. Acyclovir is active against
have a deletion of a portion of the CCR5 gene, which in certain herpes virus infections. These viruses induce virus-
turn leads to reduced or absent expression of CCR5 in specific thymidine kinase and DNA polymerase, which are
heterozygous or homozygous genotypes, respectively, with inhibited by acyclovir. Thus, acyclovir significantly reduces
no apparent deleterious consequences. Individuals homo- DNA synthesis in virus-infected cells without significantly
zygous with respect to CCR5-δ32 have a high degree of disturbing the active replication of uninfected cells.
resistance to HIV infection, whereas heterozygotes have
a reduced rate of disease progression. As a result, CCR5 Clinical Application Acyclovir has potent activity against
antagonists represent a promising new class of cell entry several DNA viruses including HSV-1, the common cause
inhibitor drugs under development for the treatment of of labial herpes (cold sore), and HSV-2, the common
HIV-1 infection (fusion inhibitor). The CCR5 antagonist cause of genital herpes (Table 38.3) (43). Varicella-zoster
maraviroc has potent anti-HIV activity in vitro and is safe virus (VZV) and some isolates of EBV are affected to a
and well-tolerated at multiple doses up to 300 mg twice daily lesser extent by acyclovir. However, CMV is less sensitive
in healthy volunteers. Maraviroc was recently approved by to acyclovir, which has no activity against vaccinia virus,
the FDA (Selzentry; Pfizer). Maraviroc is an orally adminis- adenovirus, and parainfluenza infections. An ointment
tered drug available as 150- and 300-mg film-coated tablets. containing 5% acyclovir has been used in a regimen of
The current FDA-approved dosage of maraviroc is 300 mg five times a day for up to 14 days for the treatment of
twice daily, and it must be used in combination with other herpetic keratitis and primary and recurrent infections
antiretroviral medications. Maraviroc is actively absorbed of herpes genitalis. Mild pain, transient burning, sting-
following oral administration but has a low bioavailability ing, pruritus, rash, and vulvitis have been noted. The
Kaduse.com
1284 PART III / PHARMACODYNAMIC AGENTS

TABLE 38.3 Antiviral Agents Interfering with Viral Nucleic Acid Replication
Generic Name Common Trade Spectrum of Activity Dosage Form (mg/unit)
Name Name

Acyclic nucleoside analogs (Fig. 38.7)

Acyclovir Acyclo-G Zovirax HSV-1; HSV-2; VZV; 5% Oint, Inj (5mg/ml), Cap
EBV (200), Tab (400, 800), Susp
(200/5 mL)

Valacyclovir Valtrex HSV-1; VZV; CMV Tab (500)

Cidofovir HPMPC Vistide CMV; HSV-1; HSV-2; Inj (75/mL)


VZV; CMV; EBV

Famciclovir FCV Famvir HSV; VZV; EBV; Tab (125, 250, 500)
chronic HBV

Ganciclovir DHPG Cytovene CMV retinitis Inj (50/mL), Cap (250, 500),
Vitrasert Insert (4.5/insert)

Adefovir dipivoxil Hepsera Hepatitis B Tab (10)

Conventional nucleoside analogs (Fig. 38.11)

Idoxuridine 5-IUDR Herplex HSV keratitis 0.1% Sol, 0.5% Oint


Stoxil

Ribavirin Virazole RSV; influenza A and B; Aerosol (20/mL)


HIV-1; parainfluenza

Trifluorothymidine TFT, FT3 Viroptic HSV-1 1% Sol

Vidarabine Ara-A Vira A HSV-1; HSV-2 3% Oint (monohydrate)

Cytarabine Ara-C Cytosar Herpes zoster Inj (10, 20, 50, 100/mL)

Non-nucleoside analogs (Fig. 38.13)

Boceprevir Victrelis HCV Tab (200)

Fomivirsen CMV retinitis

Foscarnet PFA Foscavir CMV retinitis; HSV Inj (24/mL)

Telaprevir Incivek HCV Tab (375)

CMV, cytomegalovirus; EBV, Epstein-Barr virus; HBV, hepatitis B virus; HSV, herpes simplex virus; VZV, varicella-zoster virus.

FDA has approved topical and intravenous (IV) acyclovir Pharmacokinetics Pharmacokinetic studies show that
preparations for initial herpes genitalis and HSV-1 and IV dose administration of acyclovir 2.5 mg/kg results in
HSV-2 infections in immunocompromised patients (44). peak plasma concentrations of 3.4 to 6.8 mg/mL (46,47).
In these individuals, early use of acyclovir shortens the The bioavailability of acyclovir is 15% to 30%, and it is
duration of viral shedding and lesion pain. Oral doses metabolized to 9-carboxymethoxymethylguanine, which
of 200 mg of acyclovir, taken five times a day for 5 to 10 is inactive. Plasma protein binding averages 15%, and
days, have not proven successful because of the low bio- approximately 70% of acyclovir is excreted unchanged in
availability of current preparations. Oral doses of 800 mg the urine by both glomerular filtration and tubular secre-
of the drug given five times daily for 7 to 10 days have tion. The half-life of the drug is approximately 3 hours
been approved by the FDA for treatment of herpes zoster in patients with normal renal function. In an individual
infection. This treatment shortens the duration of viral with renal diseases, the half-life of the drug is prolonged.
shedding in chickenpox and shingles. The IV injection of Therefore, acyclovir dosage adjustment is necessary for
the drug (10 mg/kg three times daily for 10 to 12 days) patients with renal impairment. Acyclovir easily pen-
has been approved for the treatment of herpes simplex etrates the lung, brain, muscle, spleen, uterus, vaginal
encephalitis (45). Excessive and high doses of acyclovir mucosa, intestine, liver, and kidney.
have, however, caused viruses to develop resistance to
the drug. This resistance results from reduction of virus- VALACYCLOVIR Valacyclovir hydrochloride is an amino
encoded thymidine kinase, which does not effectively acid ester prodrug of acyclovir, which exhibits antiviral
activate the drug. activity only after metabolism in the intestine or liver to
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1285

O O NH2
N HN N N N N
HN

H2N N N H2N N N H2N N N O N


HOCH2 (CH3)2CH CH C OCH2 HOCH2 CH2 O
O O O H C O CH2 P OH
NH2 O
CH2OH OH

Acyclovir Valacyclovir 6-Deoxyacyclovir Cidofovir

O NH2
N N N N
HN N
O
H2N N N H2N N N N O
N
H3C C OCH2 HOCH2 O P O O
O
O O
O
O OH
C CH3 O
O
Famciclovir Ganciclovir Adefovir dipivoxil

FIGURE 38.7 Agents that interfere with viral nucleic acid replication: acyclic nucleosides.

acyclovir followed by conversion to the triphosphate as by viral nucleoside kinases. It produces antiviral effects
shown in Figure 38.8 (48). Structurally, it differs from through interfering with DNA synthesis and inhibiting
acyclovir by the presence of the amino acid valine attached viral replication.
to the 5′-hydroxyl group of the nucleoside. Valacyclovir’s
benefit comes from an increased GI absorption resulting Clinical Application Cidofovir is active against herpes
in higher plasma concentrations of acyclovir, which is viruses including HSV-1 and HSV-2, VZV, CMV, and EBV.
normally poorly absorbed from the GI tract. As with acy- It is effective against acyclovir-resistant strains of HSV and
clovir, valacyclovir is active against HSV-1, VZV, and CMV ganciclovir-resistant strains of CMV. Cidofovir is a long-
because of its affinity for the viral form of the enzyme thy- acting drug for the treatment of CMV retinitis in AIDS
midine kinase. Oral valacyclovir is used for the treatment patients given as IV infusion or intravitreal injection. It is
of acute, localized herpes zoster (shingles) in immuno- not a curative drug, and its benefit over foscarnet or gan-
competent patients and may be given without meals. It is ciclovir is yet to be determined. The major adverse effect
also used for the initial and recurrent episodes of genital is nephrotoxicity, which appears to result in renal tubular
herpes infections (Table 38.3). The adverse effects are damage. Concomitant administration of cidofovir with
similar to acyclovir and include nausea, headache, vomit- probenecid is contraindicated because of increased risk
ing, constipation, and anorexia. The binding of valacy- of nephrotoxicity. Topical cidofovir (0.2%) is as effec-
clovir to human plasma proteins ranges from 13.5% to tive as trifluridine (1%) in reducing HSV-1 shedding and
17.9%. The plasma elimination half-life of acyclovir is healing time in rabbits with dendritic keratitis. Cidofovir
2.5 to 3.3 hours. The bioavailability of valacyclovir hydro- is administered IV, topically, and by ocular implant (Table
chloride is 54% compared to approximately 20% for oral 38.3). Peak plasma concentration of 3.1 to 23.6 mg/mL
acyclovir. It is as effective as acyclovir in decreasing the is achieved with doses of 1.0 to 10.0 mg/kg, respectively.
duration of pain associated with posttherapeutic neural- The terminal plasma half-life is 2.6 hours, and 90% of the
gia and episodes of genital lesion healing. The related drug is excreted in the urine. It has a variable bioavail-
analog 6-deoxyacyclovir (Fig. 38.8) is a prodrug form of ability (2% to 26%).
acyclovir that is activated through metabolism by xan-
thine oxidase. This drug, which has improved solubility FAMCICLOVIR
characteristics when compared to acyclovir, is used for Mechanism of Action Famciclovir is a synthetic purine
the treatment of VZV infection. nucleoside analog related to guanine (50,51). It is the
diacetyl 6-deoxy ester of penciclovir, which is structur-
CIDOFOVIR ally related to ganciclovir. Its pharmacologic and micro-
Mechanism of Action Cidofovir (1-[(S)-3-hydroxy-2- biologic activities are similar to acyclovir. Famciclovir is
(phosphonomethoxy)propyl]cytosine) is a synthetic acy- a prodrug of penciclovir (Fig. 38.9), which is formed in
clic purine nucleotide analog of cytosine (Fig. 38.7) (49). vivo by hydrolysis of the acetyl groups and oxidation at
It is a phosphorylated nucleotide that is additionally the 6-position by mixed function oxidases. Penciclovir
phosphorylated by host cell enzymes to its active intra- and its metabolite penciclovir triphosphate possess
cellular metabolite, cidofovir diphosphate. This reaction antiviral activity resulting from inhibition of viral DNA
occurs without initial virus-dependent phosphorylation polymerase.
Kaduse.com
1286 PART III / PHARMACODYNAMIC AGENTS

O O
N N N N N N
HN HN

H2N N N H2N N N H2N N N H2N N N

CH C OCH2 HOCH2 H3C C OCH2 HOCH2


NH2 O O O O

Valacyclovir 6-Deoxyacyclovir O OH
C CH3
O
O
Famciclovir Penciclovir
HN N
O
H2N N N
HN N
HOCH2
O H2N N N
HOCH2
O
Acyclo-G (acyclovir)

OH
O O Ganciclovir
HN N N
HN FIGURE 38.9 Metabolic activation of famciclovir.
H2N N N N
H2N N
O
O (HO)2 P OCH2
HO C
O
O is ganciclovir triphosphate, which is an inhibitor of viral
rather than cellular DNA polymerase. The phosphoryla-
9-Carboxymethoxy Acylo-G monophosphate
tion of ganciclovir does not require a virus-specific thymi-
methylguanine
dine kinase for its activity against CMV. The mechanism of
action is similar to that of acyclovir; however, ganciclovir
O
is more toxic to human cells than is acyclovir. Ganciclovir
N
has greater activity than acyclovir against CMV and EBV
HN
infection in immunocompromised patients. It is also
N
H2N N active against HSV infection and in some mutants resis-
O
H O P O CH2
tant to acyclovir. In AIDS patients, ganciclovir stopped
O progressive hemorragic retinitis and symptomatic pneu-
OH
3 monitis related to CMV infection.
Acyclo-G triphosphate
Clinical Application Ganciclovir is absorbed and phos-
FIGURE 38.8 Metabolic reactions of acyclovir and valacyclovir. phorylated by infection-induced kinases of HSV and VZV
infections. Common side effects are leukopenia, neutro-
Clinical Application Famciclovir is active against recur- penia, and thrombocytopenia. Ganciclovir with zidovu-
rent HSV (genital herpes and cold sores), VZV, and EBV dine causes severe hematologic toxicity. Ganciclovir is
but less active against CMV (Table 38.3). It is used in the available only as an IV infusion because oral bioavailabil-
treatment of recurrent localized herpes zoster and geni- ity is poor (Table 38.3). It is given in doses of 5 mg/kg
tal herpes in immunocompetent adults and is also prom- twice daily for 14 to 21 days. When ganciclovir is given
ising for the treatment of chronic HBV reinfection after by IV administration, concentrations of the drug in the
liver transplantation. CSF and the brain vary from 25% to 70% of the plasma
concentration. After minimal metabolism, ganciclovir is
Pharmacokinetics Famciclovir can be given with or excreted in the urine. In adults with normal renal func-
without food. The most common adverse effects are tion, the serum half-life of the drug is approximately
headache and GI disturbances. Concomitant use of fam- 3 hours. Ganciclovir has been approved by the FDA for
ciclovir with probenecid results in increased plasma con- the treatment of CMV retinitis in immunocompromised
centrations of penciclovir. The recommended dose of and AIDS patients.
famciclovir is 500 mg every 8 hours for 7 days. The abso-
lute bioavailability of famciclovir is 77%, and area under ADEFOVIR DIPIVOXIL (53)
plasma concentration–time curve (AUC) is 86 mcg/mL. Mechanism of Action Adefovir dipivoxil is an orally
Famciclovir with digoxin increased plasma concentration active prodrug indicated for the treatment of chronic
of digoxin to 19% as compared to digoxin given alone. HBV (Fig. 38.7). The drug is hydrolyzed by extracellular
esterases to produce adefovir, which in turn is phosphor-
GANCICLOVIR ylated by adenylate kinase to adefovir phosphate, which
Mechanism of Action Ganciclovir sodium is an acyclic inhibits HBV DNA polymerase. Incorporation of adefo-
deoxyguanosine analog of acyclovir (52). Its active form vir into viral DNA also leads to DNA chain termination.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1287

NH2 O
Esterase N R
N HN
Adefovir dipivoxil Idoxuridine (R = I, X = OH)
N N O X O N
Fluorodeoxyuridine (R = F, X = OH)
O P O O
Bromodeoxyuridine (R = Br, X = OH)
Adefovir O
5'-Aminoidoxuridine (R = I, X = NH2)
OH

Adenylate kinase O O
CF3
NH2 H2N N HN
N N
N HO N HO O N
O O
N N O O
O P O P O
O O OH OH OH

Adefovir phosphate Ribavirin Trifluorothymidine


(TFT, F3T)
FIGURE 38.10 Activation of the prodrug adefovir dipivoxyl by
NH2 NH2
esterase and adenylate kinase.
N N N

N N O N
HO HO
As shown in Figure 38.10, adefovir dipivoxil is activated HO HO
O O
in two steps involving an esterase that exposes a free
phosphate group (adefovir), followed by addition of a OH OH
second phosphate by adenylate kinase to form adefovir
Vidarabine (ara-A) Cytarabine
phosphate.
(ara-C)

Pharmacokinetics Adefovir is poorly absorbed orally, but FIGURE 38.11 Agents that interfere with viral nucleic acid repli-
the bioavailability of adefovir dipivoxil reaches approx- cation: conventional nucleosides.
imately 59%. The drug is absorbed to an equal extent
with or without the presence of food, and it is excreted
unchanged by the kidney. Adefovir dipivoxil joins inter- genital HSV or for cutaneous herpes zoster infection.
feron and lamivudine in the treatment of chronic HBV. Idoxuridine in dimethylsulfoxide (DMSO), however,
It can be used singly or in combination with lamivudine. has been used in mucocutaneous HSV infection of the
Early clinical studies indicate benefit of the use of adefo- mouth and nose. Because DMSO facilitates drug absorp-
vir dipivoxil to treat lamivudine-resistant HBV with a low tion and also has some therapeutic effect, a 40% solution
level of resistant virus developing to monotherapy with of idoxuridine in DMSO is more effective than idox-
adefovir dipivoxil. uridine used without this vehicle. Therefore, the FDA
approved idoxuridine only for topical treatment of her-
Conventional Nucleoside Analogs (Fig. 38.11) pes simplex keratitis, and it is more effective in epithelial
IDOXURIDINE than in stromal infections. It is less effective for recurrent
Mechanism of Action Idoxuridine is a nucleoside ana- herpes keratitis, probably because of the development of
log of thymidine containing a halogenated pyrimidine drug-resistant virus strains.
(54). It acts as an antiviral agent against DNA viruses by
interfering with their replication based through com- Side Effects Adverse reactions of idoxuridine include
petitive inhibition. Idoxuridine is first phosphorylated by such local reactions as pain, pruritus, edema, burning,
the host cell virus-encoded enzyme thymidine kinase to and hypersensitivity. Systemic administration of idox-
an active triphosphate form. The phosphorylated drug uridine by IV injection may be given in an emergency
inhibits cellular DNA polymerase to a lesser extent than but leads to bone marrow toxicities such as leukopenia,
HSV DNA polymerase, which is necessary for the synthe- thrombocytopenia, and anemia. It may also induce sto-
sis of viral DNA. The triphosphate form of the drug is matitis, nausea, vomiting, abnormalities of liver func-
then incorporated during viral nucleic acid synthesis by a tions, and alopecia. Idoxuridine has a plasma half-life of
false pairing system that replaces thymidine. When tran- 30 minutes and is rapidly metabolized in the blood to
scription occurs, faulty viral proteins are formed, result- idoxuracil and uracil.
ing in defective viral particles (55). Other halogenated uridine derivatives can also be used
as antiviral agents (Fig. 38.11). Fluorodeoxyuridine has in
Clinical Application Idoxuridine is available as ophthal- vitro antiviral activity but is not used in clinical practice.
mic drops (0.1%) and ointment (0.5%) for the treat- Bromodeoxyuridine has been used in subacute scleros-
ment of HSV keratoconjunctivitis (Table 38.3). Because ing panencephalitis, a deadly virus-induced CNS disease.
of its poor solubility, the drug is ineffective in labial or This agent appears to interfere with DNA synthesis in the
Kaduse.com
1288 PART III / PHARMACODYNAMIC AGENTS

same way as idoxuridine. The 5′-amino analog of idox- and is a potent, specific inhibitor of replication of HSV-1
uridine (5-iodo-5′-amino-2,′5′-dideoxyuridine) is a bet- in vitro. Its mechanism of action is similar to that of idox-
ter antiviral agent than idoxuridine and is less toxic. It uridine. Like other antiherpes drugs, it is first phosphory-
is metabolized in herpesvirus-infected cells only by thy- lated by thymidine kinase to mono-, di-, and triphosphate
midine kinase to di- and triphosphoramidates. These forms, which are then incorporated into viral DNA in
metabolites inhibit HSV-specific late RNA transcription, place of thymidine to stop the formation of late virus
causing reduction of less infective abnormal viral pro- mRNA and subsequent synthesis of the virion proteins.
teins. Although all of these analogs of idoxuridine have
been studied for various clinical applications, none are Clinical Application Because of its greater solubility in
presently marketed in the United States. water, it is active against HSV-1 and HSV-2. It is also useful
in treating infections caused by human CMV and VZV
RIBAVIRIN infections. The advantage of use of this agent over idox-
Mechanism of Action Ribavirin, a guanosine analog, has uridine is its high topical efficacy in the cure of primary
broad-spectrum antiviral activity against both DNA and keratoconjunctivitis and recurrent epithelial keratitis. It
RNA viruses (Fig. 38.11) (56,57). It is phosphorylated by is also useful for difficult cases of herpetic iritis and estab-
adenosine kinase to the triphosphate, resulting in inhibi- lished stromal keratitis.
tion of viral-specific RNA polymerase, disrupting messen-
ger RNA and nucleic acid synthesis. Pharmacokinetics Trifluorothymidine is available as a
1% ophthalmic solution, which is effective in dendritic
Clinical Application Ribavirin is highly active against ulcers (Table 38.3). Generally, a 1% eye solution of triflu-
influenza A and B and the parainfluenza group of orothymidine is well tolerated. Cross-hypersensitivity and
viruses, genital herpes, herpes zoster, measles, and acute cross-toxicity between trifluorothymidine, idoxuridine,
hepatitis types A, B, and C. Aerosolized ribavirin has been and vidarabine are rare. The most frequent side effects
approved by the FDA for the treatment of lower respira- are temporary burning, stinging, localized edema, and
tory tract infections (bronchiolitis and pneumonia) and bone marrow toxicity. It is less toxic but more expen-
serious RSV infections, but it can cause cardiopulmonary sive than idoxuridine. Trifluorothymidine, given intra-
and immunologic disorders in children. Ribavirin inhib- venously, shows a plasma half-life of 18 minutes and is
its in vitro replication of HIV-1; clinically, ribavirin was excreted in urine unchanged or as the inactive metabo-
shown to delay the onset of full-blown AIDS in patients lite 5-carboxyuracil.
with early symptoms of HIV infection. Some viruses are
less susceptible, for example, poliovirus, herpes viruses VIDARABINE
excluding varicella, vaccinia, mumps, reovirus, and rota- Mechanism of Action Vidarabine is an adenosine nucle-
virus. A randomized double-blind study of aerosolized oside obtained from cultures of Streptomyces antibioticus
ribavirin treatment of infants with RSV infections indi- (Fig. 38.11) (60). Cellular enzymes convert vidarabine
cated significant improvement in the severity of infection to mono-, di-, and triphosphate derivatives that interfere
with a decrease in viral shedding (58). with viral nucleic acid replication, specifically inhibiting
the early steps in DNA synthesis. This agent was used orig-
Pharmacokinetics Oral or IV forms of ribavirin are use- inally as an antineoplastic drug. Its antiviral effect is, in
ful in the prevention and treatment of Lassa fever. The some cases, superior to that of idoxuridine or cytarabine.
oral bioavailability is approximately 45%, and serum
half-life is 9 hours. Peak plasma level after 1 hour is Clinical Application Vidarabine is used mainly in human
1 to 3 mg/mL. IV administration of the drug has higher HSV-1 and HSV-2 encephalitis, decreasing the mortality
peak plasma levels. Aerosol preparation delivery of drug rate from 70% to 30%. Whitley et al. (61) reported that
(0.8 mg/kg/hour) produced drug levels in respiratory early vidarabine therapy is helpful in controlling com-
secretions of 50 to 200 mg/mL (Table 38.3). The clinical plications of localized or disseminated herpes zoster in
benefits of this agent have yet to be confirmed. Its few immunocompromised patients. Vidarabine is also use-
side effects are generally limited to GI disturbances, such ful in neonatal herpes labialis or genitalis, vaccinia virus,
as nausea, vomiting, and diarrhea. The drug is contra- adenovirus, RNA viruses, papovavirus, CMV, and small-
indicated in asthma patients because of deterioration of pox virus infections. Given the efficacy of vidarabine in
pulmonary function. Viral strains susceptible to ribavirin certain viral infections, the FDA approved a 3% ointment
have not been found to develop drug resistance, as is the for the treatment of herpes simplex keratoconjunctivitis
case with other antiviral agents, such as acyclovir, idoxuri- and recurrent epithelial keratitis and a 2% IV injection
dine, and bromovinyldeoxyuridine. for the treatment of herpes simplex encephalitis and her-
pes zoster infections (Table 38.3). A topical ophthalmic
TRIFLUOROTHYMIDINE preparation of vidarabine is useful in herpes simplex ker-
Mechanism of Action Trifluorothymidine is a fluori- atitis but shows little promise in herpes simplex labialis
nated pyridine nucleoside structurally related to idoxuri- or genitalis. The monophosphate esters of vidarabine are
dine (Fig. 38.11) (59). It has been approved by the FDA more water-soluble and can be used in smaller volumes
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1289

and even intramuscularly. These esters are under clini- (shingles) infection. It is also used to treat herpetic
cal investigation for the treatment of HBV, systemic and keratitis and viral infections resistant to idoxuridine. The
cutaneous herpes simplex, and herpes zoster virus infec- drug is usually used topically, but it has been given by
tions in immunocompromised patients. IV injection to individuals with serious herpes infection
(64). Cytarabine is deaminated rapidly in the body to an
Pharmacokinetics Vidarabine is deaminated rapidly by inactive compound, arabinosyluracil, which is excreted
adenosine deaminase, which is present in serum and in the urine. The half-life of the drug in plasma is 3 to
red blood cells. The enzyme converts vidarabine to its 5 hours. The toxic effects of cytarabine are chiefly on
principal metabolite, arabinosyl hypoxanthine (ara-HX), bone marrow, the GI tract, and the kidney. The drug is
which has weak antiviral activity (Fig. 38.12) (62). The not given in the early months of pregnancy because of its
half-life of vidarabine is approximately 1 hour, whereas teratogenic and carcinogenic effects in animals.
ara-HX has a half-life of 3.5 hours. The drug is detected
mostly in the kidney, liver, and spleen because 50% of it Nonnucleoside Analogs (Fig. 38.13)
is recovered in the urine as ara-HX. Levels of vidarabine FOMIVIRSEN SODIUM Fomivirsen was the first antisense oli-
in CSF are 50% of those in the plasma. The most com- gonucleotide agent approved as an alternative medicine
mon side effects of vidarabine are GI disturbances such for patients with CMV retinitis for whom other agents
as anorexia, nausea, vomiting, and diarrhea. CNS side did not work. Fomivirsen is a 21-mer phosphorothio-
effects include tremors, dizziness, pain syndromes, and ate oligodeoxynucleotide (Fig. 38.13). Fomivirsen was
seizures. Bone marrow suppression is reported at higher used to treat CMV, which causes opportunistic retinitis
doses. Because vidarabine is reported to be mutagenic, in patients with AIDS. Such patients respond to fomi-
carcinogenic, and teratogenic in animal studies, its use virsen but not to other treatment for CMV retinitis, which
in pregnant women is to be avoided. Allopurinol and leads to blindness (65). It works by inhibiting synthesis
theophylline may interfere with the metabolism of vida- of proteins responsible for the regulation of viral gene
rabine at higher doses because of the xanthine oxidase expression that is involved in infection of CMV retinitis.
metabolism of vidarabine. Therefore, this agent should Fomivirsen has recently been withdrawn from the market
be avoided or given with caution to patients receiving by the drug manufacturer Novartis and is thus no longer
these medications concurrently. Also, adjustment of the available.
dose is necessary in patients with renal insufficiency.
FOSCARNET SODIUM Foscarnet is a trisodium phosphofor-
CYTARABINE mate hexahydrate that inhibits DNA polymerase of her-
Mechanism of Action Cytarabine (1-β′-arabinofu- pes viruses including CMV and retroviral RT (Fig. 38.13)
ranosylcytosine) is a pyrimidine nucleoside related to (66). It is not phosphorylated into an active form by viral
idoxuridine (Fig. 38.11) (63). It is used primarily as an host cell enzymes. Therefore, it has the advantage of not
anticancer rather than an antiviral agent (see Chapter 37). requiring an activation step before attacking the target
Cytarabine acts by blocking the utilization of deoxycytidine, viral enzyme. Foscarnet sodium was approved by the FDA
thereby inhibiting the replication of viral DNA. The drug
is first converted to mono-, di-, and triphosphates, which
interfere with DNA synthesis by inhibiting both DNA poly- 5'-GCG TTT GCT CTT CTT CTT GCG-3'
merase and the reductase that promotes the conversion of
Fomivirsen
cytidine diphosphate into its deoxy derivatives. H3C CH3
H H O
Clinical Application Cytarabine is used to treat Burkitt H
N NH2
lymphoma and both myeloid and lymphatic leukemias. O N
H H
N N O O
Its antiviral use is in the treatment of herpes zoster O P COO
(Na )3 O
O
O

NH2 O
Foscarnet sodium Boceprevir
N N HN N

N N N N
H H O
HO HO H H
HO Adenosine HO N N
O O O N
deamination H
O O
N N
N O
OH OH H
O CH3
N
Vidarabine Arabinofuranosylhypoxanthine
(ara-A) (ara-HX)
Telaprevir
FIGURE 38.12 Metabolism of vidarabine by adenosine
deaminase. FIGURE 38.13
Kaduse.com
Nonnucleoside inhibitors of viral replication.
1290 PART III / PHARMACODYNAMIC AGENTS

for the treatment of CMV retinitis in AIDS patients. In Methisazone (70)


combination with ganciclovir, the results have been prom- Methisazone interferes with the translation of mRNA
ising, even in progressive disease with ganciclovir-resistant message at the ribosome, preventing the synthesis of
strains. Foscarnet sodium is also effective in the treatment protein synthesis. Ultimately, it produces a defect in
of mucocutaneous diseases caused by acyclovir-resistant protein incorporation into the virus. Although viral
strains of HSV and VZV in AIDS patients. The drug is DNA increases and host cells are damaged, an infec-
administered intravenously (60 mg/kg) three times a day tious virus is not produced. Methisazone is active
for initial therapy and at a dose of 90 to 120 mg/kg daily against poxviruses, including variola and vaccinia (71).
for maintenance therapy (Table 38.3). The plasma-half life Some RNA viruses, such as rhinoviruses, echoviruses,
is 3 to 6 hours. Foscarnet sodium penetrates into the CSF reoviruses, influenza, parainfluenza, and polioviruses
and the eye. The drug is neurotoxic, and common adverse are also inhibited. Therapeutically, methisazone is
effects include phlebitis, anemia, nausea, vomiting, and given in 1.5- to 3.0-g doses, twice daily by mouth. It has
seizures. Foscarnet sodium carries risk of severe hypocal- also been used as a prophylactic agent against small-
cemia, especially with concurrent use of IV pentamidine. pox. Historically, methisazone was one of the first anti-
Foscarnet sodium used with zidovudine has an additive viral compounds used in clinical practice. It is orally
effect against CMV and acts synergistically against HIV. absorbed, with nausea and vomiting as the principal side
effects. The drug is also used in vaccinia gangrenosa
BOCEPREVIR AND TELAPREVIR Boceprevir (SCH 503034, and disseminated vaccinia infections. This drug is not
Victrelis) (67) and telaprevir (VX-950, Incivek) (68) available in the United States, but it has been used in
represent significant advances in the treatment of HCV Europe for some time. Several analogs of methisazone
infection (Fig. 38.13) (Table 38.3). HCV is an enveloped, (R = H, R = ethyl) possess activity against variola, neu-
single-stranded sense RNA virus that encodes a polyprot- rovaccinia, smallpox in mice, Rous sarcoma virus, and
ein precursor of about 3,000 amino acids. The essential vaccinia generalisata.
HCV enzyme NS3-4A protease is responsible for cleaving
this HCV polyprotein at four sites to generate four pep- Antiretroviral (Anti-HIV) Agents Including Protease
tides called NS4A, NS4B, NS5A, and NS5B. Boceprevir Inhibitors (72,73)
and telaprevir act as potent inhibitors of the HCV NS3-4A While there can be no permanent cure of AIDS with-
protease, and this inhibition prevents the processing of out prevention or elimination of HIV infection, AIDS
the HCV polyprotein and prevents the virus from replicat- patients can prolong their life if the disease is diagnosed
ing. Both compounds have recently been approved by the early and treatment is promptly initiated. Initial HIV
FDA and can be used in combination with pegylated inter- treatment requires specific drugs that inhibit RT and
feron and ribavirin (69). For untreated HCV infection, tel- HIV protease. In advanced HIV infection, AIDS is com-
aprevir is administered for the first 12 weeks of therapy at plicated by other organisms that proliferate in immuno-
750 mg three times per day in combination with pegylated compromised hosts, known as opportunistic infections.
interferon α-2a 180 μg/week, and ribavirin 1.0 to 1.2 g/ Such patients are treated symptomatically with a vari-
day according to body weight. After 12 weeks, telaprevir ety of drugs depending on the opportunistic infections
is discontinued, and pegylated interferon α-2a and riba- (74–76). Anti-HIV agents have side effects, but patients
virin are continued until HCV RNA is undetectable in can be managed by a careful monitoring of the drugs.
serum. Boceprevir must be administered using a different Opportunistic diseases include infections by parasites,
regimen. For untreated HCV infection, pegylated inter- bacteria, fungi, and viruses. Neoplasms including Kaposi
feron α-2b and ribavirin are given alone for 4 weeks, and sarcoma and Burkitt lymphoma also commonly occur.
then boceprevir (800 mg three times a day) is given in Anti-HIV agents are classified according to the mode
combination with pegylated interferon α-2b (1.5 μg/kg/ of action. The drugs inhibiting RT interfere with repli-
week) and ribavirin (0.8 to 1.4 g/day according to body cation of HIV and stop synthesis of infective viral par-
weight) for 24 weeks. Total treatment duration is 28 weeks ticles. They are further classified into nucleoside and
if patients test negative for HCV RNA but may need to be nonnucleoside RT inhibitors. The drugs inhibiting HIV
continued for an additional 20 weeks to clear the infec- protease inactivate RT activity and block release of viral
tion if HCV RNA is still detected after 28 weeks. particles from the infected cells. The chemistry, pharma-
cokinetics, side effects, toxicity, and drug interactions of
Agents Affecting Translation by the Ribosome RT inhibitors and protease inhibitors are discussed in the
following sections.
H
N
N NH2 Nucleoside Reverse Transcriptase Inhibitors (77,78)
S The synthesis of viral DNA under the direction of RT
O
N
requires availability of purines and pyrimidine nucleo-
R sides and nucleotides. Therefore, it is not surprising that
a variety of chemical modifications of natural nucleo-
Methisazone ( R = CH3) sides have been investigated. Two such modifications
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1291

have resulted in active drugs. Removal of the ribosyl was first synthesized by Horwitz (1964) (90), biologic
3′-hydroxyl group of the deoxynucleosides has given rise activity was reported by Ostertag et al. (1974) (91), and
to dideoxyadenosine (didanosine is the prodrug for this in 1986, Yarchoan et al. (92) demonstrated application
derivative) (79,80), dideoxycytidine (81–83), and didehy- of ZDV in clinical trials of AIDS and related diseases.
drodideoxythymidine (84). Replacement of the 3′-deoxy ZDV is recommended in the control of the disease in
with an azido group has given 3′-azidothymidine (85–88) asymptomatic patients in whom absolute CD4+ lympho-
and 3′-azidouridine (no longer used as a drug) (Fig. 38.14 cyte counts are less than 200/mm3. It prolongs the life
and Table 38.4) (89). All of these drugs have similar of patients affected with Pneumocystis jirovecii pneumonia
mechanisms of action in that their incorporation into and improves the condition of patients with advanced
the viral DNA will ultimately lead to chain-terminating ARC by reducing the severity and frequency of opportu-
blockade due to the lack of a 3′-hydroxyl needed for the nistic infections. Substantial benefits are obtained when
DNA propagation. the drug is given after the CD4+ cell counts fall below
500/mm3. Therefore, ZDV is used in early and advanced
ZIDOVUDINE symptomatic treatment of AIDS or ARC patients. ZDV
Mechanism of Action Zidovudine (ZDV) is an analog of with other RT inhibitors or in combination with prote-
thymidine in which the azido group is substituted at the ase inhibitors is more beneficial when resistance to ZDV
3-carbon atom of the dideoxyribose moiety (Fig. 38.14). occurs.
It is active against RNA tumor viruses (retroviruses) HIV attacks susceptible cells and interacts mainly with
that are the causative agents of AIDS and T-cell leuke- CD4+ cell surface proteins of helper T cells. As discussed
mia. Retroviruses, by virtue of RT, direct the synthesis of earlier, the viral glycoprotein gp120 forms a complex
a provirus (DNA copy of a viral RNA genome). Proviral with CD4 receptor on host cells and enters the cells by
DNA integrates into the normal cell DNA, leading to the endocytosis. The sequence of events is shown in Figure
HIV infection. ZDV is converted to 5′-mono-, di-, and tri- 38.2. Ultimately, the immune system of the host is altered
phosphates by the cellular thymidine kinase. These phos- and AIDS symptoms appear. AIDS patients have symp-
phates are then incorporated into proviral DNA, because toms such as high fever, weight loss, lymphadenopathy,
RT uses ZDV-triphosphate as a substrate. This process chronic diarrhea, myalgias, fatigue, and night sweats.
prevents normal 5′,3′-phosphodiester bonding, result- ZDV is given in such conditions. However, the drug is
ing in termination of DNA chain elongation due to the toxic to the bone marrow and causes macrocytic anemia,
presence of an azido group in ZDV. The multiplication of neutropenia, and granulocytopenia. Other adverse reac-
HIV is halted by selective inhibition of RT and thus viral tions include headache, insomnia, nausea, vomiting, sei-
DNA polymerase by ZDV-triphosphate at the required zures, myalgias, and confusion.
dose concentration. ZDV is a potent inhibitor of HIV-1
but also inhibits HIV-2 and EBV. Pharmacokinetics ZDV is available in 100-mg capsules
for oral administration. For asymptomatic adults, the
Clinical Application ZDV is used in AIDS and AIDS- initial recommended dosage is 1,200 mg daily (200 mg
related complex (ARC) to control opportunistic infec- every 4 hours) reducing to 600 mg daily (100 mg every
tions by raising absolute CD4+ lymphocyte counts. ZDV 4 hours) for patients with advanced disease (Table 38.4).

O O
O NH2
CH3 CH3
HN N HN
HN N
HO O N HO N HO HO O N
N O N
O O O O

N3
Didanosine(ddI) Zalcitabine Stavudine
Zidovudine(AZT) (2',3'-Dideoxy-2',3'-didehydro-
(2',3'-Dideoxycytidine, ddC)
thymidine, D4T)

NH2 HN NH2 NH2


N N F
N N N N
O
HO O N H2N N N N N O HO O N
O O
O HO O P O
O
O
S CH3 S
O O

O
Lamivudine Abacavir (ABC) Tenofovir disoproxil Emtricitabine
(2'-Deoxy-3'-thiacytidine, 3TC)

FIGURE 38.14 Nucleoside reverse transcriptase inhibitors (NRTIs).


Kaduse.com
1292 PART III / PHARMACODYNAMIC AGENTS

TABLE 38.4 HIV Reverse Transcriptase Inhibitors


Generic Name Common Name Trade Name Dosage Form (mg/unit)

Nucleoside reverse transcriptase inhibitors

Zidovudine AZT Retrovir Tab (300), Cap (100), Syrup (50/5 mL), Inj (10/mL)
ZDV

Didanosine ddI Videx Tab (25, 50, 100, 150, 200)

Dideoxyadenosine ddA Powder for oral Sol (100, 167, 250)

Zalcitabine ddC Hivid Tab (0.375)

Stavudine D4T Zerit Cap (15, 20, 30, 40)


Powder for oral Sol (1/mL)

Lamivudine 3TC Epivir Tab (150), Sol (10/mL)


Epivir HBV Tab (100), Sol (5/mL)

Abacavir ABC Ziagen Tab (300), Sol (20/mL)

Tenofovir disoproxil Viread Tab (300)

Emtricitabine Emtriva Cap (200)

Nonnucleoside reverse transcriptase inhibitors

Nevirapine Viramune Tab (200)

Delavirdine Rescriptor Tab (100)

Efavirenz Sustiva Cap (50, 100, 200)

Cap, capsule; Inj, injection; Sol, solution; Tab, tablet.

The maintenance dose is 600 mg daily in symptomatic As such this agent causes chain termination due to the
patients. ZDV is sensitive to heat and light because of its absence of a 3′-hydroxyl group. ddI inhibits HIV RT and
azide group and should be stored in colored bottles at exerts a virustatic effect on the retroviruses. Combined
15° to 25°C. ZDV is well absorbed through the GI tract. with ZDV, antiretroviral activity of ddI is increased.
It concentrates in the body tissues and fluids, includ-
ing CSF. The bioavailability of the drug was found to be Pharmacokinetics ddI has a plasma half-life of 1.5 hours
approximately 65%. Its half-life is approximately 1 hour. and is given in 200-mg dose twice daily. Oral bioavailabil-
IV doses of 2.5 mg/kg or oral doses of 5 mg/kg yielded ity of the drug is approximately 25% at doses of 7 mg/kg
peak plasma concentrations of 5 mmol/L. Plasma pro- or less. It significantly decreases p24 antigen levels and
tein binding was approximately 30%. Most of the drug increases CD4+ cell counts. In some cases, viral resistance
is converted to its inactive glucuronide metabolite and to ddI has been known to occur after treatment for
is excreted unchanged through urine. ZDV also crosses 1 year. It is given in advanced HIV infection, ZDV intoler-
the blood–brain barrier. Pentamidine, dapsone, ampho- ance, or significant clinical/immunologic deterioration.
tericin B, flucytosine, and doxorubicin may increase the The major side effects of ddI are painful peripheral neu-
toxic effects of ZDV. Probenecid prolongs the plasma ropathy and pancreatitis. Some of the minor side effects
half-life of the drug. include abdominal pain, nausea, and vomiting. The use
of products, such as pentamidine, sulfonamides, and
DIDANOSINE cimetidine should be avoided with ddI.
Mechanism of Action Didanosine (ddI) is a purine
dideoxynucleoside analog of inosine. Chemically, it is ZALCITABINE Zalcitabine (ddC) is a useful alternate to ZDV
2′,3′-dideoxyinosine and differs from inosine by having and is a synthetic pyrimidine nucleoside analog. It differs
hydrogen atoms in place of 2′- and 3′-hydroxyl groups on from 2′-deoxycytidine in that the 3′-hydroxyl group of the
the ribose ring (Fig. 38.14). ddI is a prodrug that is bioac- 2′-deoxyribose moiety is replaced with a hydrogen atom
tivated by metabolism to dideoxyadenosine triphosphate. (Fig. 38.14). It is given in combination with ZDV when
Dideoxyadenosine triphosphate is a competitive inhibi- CD4+ cell counts fall below 300 cells/mm3. Monotherapy
tor of viral RT and is incorporated into the developing with ddC was more active than ZDV. Its oral bioavailabil-
viral DNA in place of deoxyladenosine triphosphate. ity is 87%, and plasma half-life is approximately 1 hour.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1293

It has side effects such as stomatitis, rash, fever, malaise, progression caused by HIV infection. The combinations
arthritis, and arthralgia. In low doses (0.005 mg/kg of 3TC with ddI, ddC, or D4T also are used for advanced
every 4 hours), ddC produced sustained decrease in p24 HIV infection. Such combinations have the ability to
antigen level and increase in CD4+ cell counts. The CSF delay resistance to ZDV and restore ZDV sensitivity in
fluid/plasma ratio of ddC is 0.2. AIDS patients. Recently, oral therapy in lower doses of
After oral administration, bioavailability of ddC was 3TC (Table 38.4) has been approved by the FDA for
less than 80%, which was further reduced with food. The treatment of chronic HBV. Peripheral neuropathy and
mean maximum plasma concentration of the drug was GI disturbances are the major side effects of 3TC. The
also reduced from 25.2 to 15.5 ng/mL when the drug was minor side effects are nausea, vomiting, and diarrhea.
taken with food. Dideoxyuridine is the major metabolite
in urine and feces. The drug demonstrated penetration ABACAVIR SULFATE Abacavir was approved in 1998 as a
through blood–brain barrier. The major toxicity of ddC nucleoside reverse transcriptase inhibitor to be used in
is peripheral neuropathy, in which case it should be dis- combination with other drugs for the treatment of HIV
continued. Pancreatitis occurs in some cases when given and AIDS. The drug is extensively metabolized via step-
alone or in combination with ZDV. wise phosphorylation to 5′-mono-, di-, and triphosphate.
Abacavir is well absorb (>75%) and penetrates the CNS
STAVUDINE Stavudine (D4T) is a pyrimidine nucleo- and can be taken without regard to meals. The drug does
side analog that has significant activity against HIV-1 not show any clinically significant drug–drug interactions
after intracellular conversion of the drug to a D4T- but has been reported to produce life-threatening hyper-
triphosphate. It differs in structure from thymidine by sensitivity reactions. The major use of abacavir appears
the replacement of the 3′-hydroxyl group with a hydro- to be in combination with other nucleoside reverse tran-
gen atom and a double bond in the 2′ and 3′ positions scriptase inhibitors. A fixed combination product has
on the deoxyribose ring (Fig. 38.14). It decreased p24 recently been approved by the FDA under the trade name
antigen and raised CD4+ cell counts. D4T is beneficial of Trizivir and consists of 300 mg of abacavir, 150 mg of
for patients in whom CD4+ cell counts do not decrease 3TC, and 300 mg of ZDV. The combination has been
below 300 cells/mm3 with ZDV and ddI. It was shown to shown to be superior to other combinations in reducing
be more effective than ZDV or ddC in delaying the pro- viral load as well as showing improvement in CD4+ cell
gression of HIV infection. It is recommended for patients count. The most common adverse effects reported with
with advanced HIV infection. abacavir include headache, nausea, vomiting, malaise,
and diarrhea.
Pharmacokinetics D4T is rapidly absorbed, and abso-
lute bioavailability in adults is 85% at an oral dose of TENOFOVIR DISOPROXIL (94)
4 mg/kg. A peak plasma concentration in a dose-depen- Mechanism of Action Tenofovir is a prodrug with a bio-
dent manner occurs within an hour. It can be taken with availability of 25%, which is improved in the presence of
food. The apparent volume of distribution after oral food (35%). The drug is approved for the treatment of
dose is 66 L. The plasma half-life of D4T is approximately HIV infections in adult patients. Tenofovir diphosphate
1.5 hours, and the intracellular half-life of D4T-triphosphate is an HIV reverse transcriptase inhibitor. The drug is
is 3.5 hours. It is less toxic to bone marrow but causes hydrolyzed via plasma esterase to tenofovir, which is then
peripheral neuropathic toxicity. The side effects include phosphorylated to the active tenofovir diphosphate, as
pain, tingling, and numbness in the hands and feet. shown in Figure 38.15. The diphosphate competes with

LAMIVUDINE Lamivudine (3TC) is an analog of ddC in


NH2 NH2
which 3′-methylene group is replaced with a sulfur (S) O
N N
atom in the ribose ring (Fig. 38.14) (93). It exerts viru- N
O
N
O
static effect against retroviruses by competitively inhibit- N N O N N O
Esterase O P O H
ing HIV RT after intracellular conversion of the drug to O P O
O O H
its active 5′-3TC-triphosphate form. It is usually given with CH3 CH3
O O
Cellular
other antiretroviral agents, such as ZDV or D4T. 3TC in a kinase
600 mg/day dose reduced HIV cells by 75%, and in com- O
bination with ZDV, the reduction in viral load was 94%. Tenofovir disoproxil Tenofovir
NH2
3TC is rapidly absorbed through the GI tract. Its bioavail-
N
ability is approximately 86% after oral administration N
of 2 mg/kg twice daily; peak serum 3TC concentration N N O O
was approximately 2 mg/mL. 3TC binding to human O P O P OH
O O
plasma was approximately 36%. In vivo, it is converted to CH3
trans sulfoxide metabolite, and a majority of the drug is
Tenofovir diphosphate
eliminated unchanged in urine. The FDA approved 3TC
in combination with ZDV for the treatment of disease FIGURE 38.15
Kaduse.com
Metabolic activation of tenofovir disoproxil.
1294 PART III / PHARMACODYNAMIC AGENTS

deoxyadenosine triphosphate for incorporation into viral activity in decreasing the viral load and increasing CD4+
DNA, and when incorporated, tenofovir diphosphate cell count. These drugs are primarily designed and syn-
results in premature termination of DNA growth and thesized by protein structure-based drug design meth-
inhibition of DNA polymerase. Tenofovir disoproxil is odologies. Their use as monotherapy may be limited
indicated for treatment-experienced patients with HIV-1. because of rapid onset of resistance and hypersensitivity
The drug appears to also be effective in treatment-naive reactions. However, interaction of nonnucleoside drugs
patients, but initial approval is for treatment-experienced with other protease inhibitors, such as saquinavir, indi-
patients. The drug is administered as one tablet once navir, and ritonavir, is being investigated. In addition,
daily (300 mg). It is recommended that the drug be interaction of these drugs with clarithromycin, ketocon-
combined with other reverse transcriptase inhibitors or azole, rifabutin, and rifampin is under study.
HIV protease inhibitors, which results in additive or syn-
ergistic activity. At present, two fixed combinations exist NEVIRAPINE
that contain tenofovir: tenofovir disoproxil fumarate Mechanism of Action Nevirapine and its analogs exhibit
plus emtricitabine (Truvada) and a fixed triple combina- antiretroviral effects against AZT-resistant HIV strains
tion of tenofovir, emtricitabine, and efavirenz (Atripla). (96). Nevirapine in combination with ZDV and ddI
Other fixed-dose products may be approved in the future produced approximately 18% higher CD4+ cell counts
for treatment of HIV. and a decrease in viral load compared with ZDV and
ddI. Nevirapine is recommended with nucleosides for
EMTRICITABINE (95) Emtricitabine is an orally active nucle- HIV-1–infected patients who have experienced clinical or
oside RT inhibitor (Fig. 38.14). It is metabolized in vivo immunologic deterioration. The significant side effects
to the 5′-triphosphate, which in turn competes with the of nevirapine are liver dysfunction and skin rashes.
normal substrate (deoxycytidine-5′-triphosphate) for Nevirapine is a dipyridodiazepinone derivative, which
incorporation into DNA. In addition, incorporation binds directly to RT. Thus, it blocks RNA- and DNA-
of emtricitabine into viral DNA inhibits further chain dependent polymerase activities by causing a disrup-
elongation, resulting in chain termination. The drug is tion of the RT catalytic site. HIV-2 RT and human DNA
administered orally once daily. The (–)-enantiomer is polymerases are not inhibited by nevirapine. The 50%
the most active form of the drug, although the (+)-isomer inhibitory concentration ranged from 10 to 100 nmol/L
is also active. Emtricitabine is not bound to plasma pro- against HIV-1.
tein, and approximately 86% is excreted unchanged in
the urine. The only metabolites identified consist of the Pharmacokinetics Nevirapine is rapidly absorbed after
3′-sulfoxide and the 2′-O-glucuronide. Emtricitabine is oral administration, and its bioavailability is approxi-
reported to be more active than 3TC with a low level mately 95%. Peak plasma nevirapine concentrations of
of resistance developing when used in combination 2 ± 0.4 mg/mL (7.5 mmol/L) are obtained in 4 hours
therapy with efavirenz and ddI. As indicated earlier, the after a single 200-mg dose (Table 38.4). Following
drug is also available in fix-dosed combinations with multiple doses, nevirapine concentrations appear to
tenofovir. increase linearly in the dose range of 200 to 400 mg/
day. Nevirapine is about 60% bound to plasma proteins
Nonnucleoside Reverse Transcriptase Inhibitors in the plasma concentration range of 1 to 10 mg/mL.
The FDA has recently approved several nonnucleosides It readily crossed the placenta and was found in breast
that inhibit RT activity (Fig. 38.16 and Table 38.4). They milk. Nevirapine is metabolized as glucuronide conju-
are used with nucleoside drugs to obtain synergistic gates of hydroxylated metabolites, which are excreted
in urine. In vivo, ketoconazole did not produce any
significant inhibitory effect on nevirapine metabolism.
O
H3C O CH(CH3)2 The plasma concentrations of nevirapine were elevated
HN H HN
H3C S N O or reduced in patients receiving cimetidine or rifabutin,
N O C N N respectively.
N N N N
H
DELAVIRDINE Delavirdine, a bisheteroarylpiperazine deriv-
Nevirapine Delavirdine ative, is a potent nonnucleoside RT inhibitor with activity
specific for HIV-1 (97). The FDA has approved this drug
F3C C C
Cl in combination with other anti-HIV agents (Table 38.4).
O
In phase I/II trials, it demonstrated sustained improve-
N
H
O ments in CD4+ cell counts, p24 antigen levels, and RNA
viral load. Promising results were obtained when the
drug was used in two- or three-drug combination with
Efavirenz
nucleoside drugs. Combination of delavirdine with ddI,
FIGURE 38.16 Nonnucleoside reverse transcriptase inhibitors ddC, or ZDV demonstrated additive or synergistic effect.
However, delavirdine with ZDV was more beneficial in
(NNRTI).
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1295

early HIV infection. Combinations of nevirapine and TABLE 38.5 HIV Protease Inhibitors
delavirdine have an antagonistic effect on HIV-1 RT inhi-
bition. Delavirdine directly inhibits RT and DNA-directed Generic Name Trade Name Dosage Form (mg/unit)
DNA polymerase activities of HIV-1 after the formation of
Saquinavir Invirase Cap (200)
the enzyme–substrate complexes, thereby causing chain
termination effects. Fortovase Cap (200)

Ritonavir Norvir Cap (200)


Pharmacokinetics Delavirdine is rapidly absorbed by
oral administration, and peak plasma concentration is Indinavir Crixivan Cap (200, 400)
obtained in 1 hour. The single-dose bioavailability of
Nelfinavir Viracept Tab (250), Powder (50/g)
delavirdine tablets relative to oral solution was approxi-
mate 85%. The 50% inhibitory concentration for dela- Amprenavir Agenerase Cap (50, 150)
virdine against RT activity was 6.0 nmol/L. Delavirdine Sol (15/mL)
is extensively bound to plasma protein (∼98%). Lopinavir/ritonavir Kaletra Cap (133.3/33.3)
Delavirdine is metabolized to its inactive N-desisopropyl Sol (80/20 per mL)
metabolite in liver, and the pharmacokinetics is non-
Atazanavir Reyataz Cap (150, 200)
linear. This reaction is catalyzed by CYP3A4, and dela-
virdine appears to be an inhibitor of this enzyme, Fosamprenavir Lexiva Tab (700)
increasing the risk for drug–drug interactions with
Tipranavir Aptivus Cap (250)
other drugs that are substrates for this cytochrome iso-
form. Skin rashes are the major side effect of delavir- Darunavir Prezista Tab (75, 150, 300, 400 or 600)
dine therapy. Cross-resistance between delavirdine and
Cap, capsule; Sol, solution; Tab, tablet.
protease inhibitors, such as indinavir, nelfinavir, ritona-
vir, and saquinavir, is unlikely because they act at differ-
ent target enzymes.
HIV Protease Inhibitors (Table 38.5) (99)
EFAVIRENZ Efavirenz is a new nonnucleoside RT inhibi- Mechanism of Action
tor that was approved by the FDA (Table 38.4) as a The HIV genome contains various structural genes,
potent inhibitor of wild-type and resistant mutant HIV-1, such as the gag and gag-pol genes, that are translated into
which is inhibited up to 95% with efavirenz concentra- precursor polyproteins that form immature viral par-
tions of 1.5 mmol/L (98). In combination with indi- ticles. These precursor protein molecules are processed
navir, a mean reduction in HIV-RNA of 1.68 log and (cleaved) by an essential viral pol-encoded aspartic pro-
an increase in CD4+ cell counts of 96 cells/mm3 were teinase, HIV protease, to form the desired structural pro-
reported. Coadministration of efavirenz with indinavir teins of the mature viral particle that are necessary for
reduced indinavir concentration (AUC) by approxi- virus replication and survival. For example, the structural
mately 35%. proteins p7, p9, p17, and p24, which play important roles
in infectivity of HIV, are products of the pol gene. HIV
Pharmacokinetics Efavirenz is administered once a day protease also activates RT and plays an important role
and can be used as a substitute for indinavir in combina- in the release of infectious viral particles. Thus, an area
tion therapy with standard drugs, such as ZDV and 3TC of considerable interest has been the development of
(also as a fixed combination of tenofovir, emtricitabine, drugs that act as inhibitors of HIV protease. Such inhibi-
and efavirenz [Atripla]). Because it is given once a day, tors act on HIV protease and prevent posttranslational
this cuts down the number of pills that an AIDS patient processing and budding of immature viral particles from
has to swallow. In the current cocktail therapy of AIDS the infected cells. This group of drugs represents a major
patients, efavirenz is a good option for reducing the breakthrough in treatment of HIV when used in combi-
many side effects of cocktail therapy. It is administered nation with RT inhibitors, and their development is one
to both adults and children and may be less expensive of the most significant advances in medicinal chemistry.
than indinavir. The side effects of efavirenz include diz- Functional HIV protease exists as a dimer in which
ziness, insomnia, impaired concentration, abnormal each monomer contains one of two conserved aspartate
dreams, and drowsiness. The most common adverse residues at the active site. There are several HIV protease
effect is a skin rash. Other side effects are diarrhea, cleavage sites on the protein precursors, but the enzyme
headache, and dizziness. Efavirenz is recommended to prefers to perform the hydrolysis of the peptide bond on
be taken at bedtime with or without food. Avoiding driv- the amino terminal side of a proline (100). As shown in
ing or operating machinery and intake of high-fat meals Figure 38.17, the amino acid R groups flanking the scis-
are recommended. It should always be taken in combi- sile bond are designated P1, P2, and so on, on the amino
nation with at least one other anti-HIV agent. Efavirenz terminus, and P1′, P2′, and so on, on the carboxyl termi-
is contraindicated with midazolam, triazolam, or ergot nus of the cleavage site (Fig. 38.17A). The corresponding
derivatives. pockets on the enzyme that are responsible for binding
Kaduse.com
1296 PART III / PHARMACODYNAMIC AGENTS

Scissile bond +
Asp25 O Asp25 O
S3 S1 S2'
O O
H H
P3 O P1 O P2' O H H
O H O N O N
H H
N N N O O
N N N N H H
H H H H N N
O P2 O P1' O P3' N N
O O
S2 S1 ' S 3' OH
O
H H
A. Nomenclature of amino acid side chains (P) and bind- OH
ing pockets (S) surrounding a protease cleavage site. O
Asp25 O
Asp25 O

Asp25 Asp25 B. Role pf twp Asp25 residues in formation of the


O O
hydrolytic transition state.
O O
S2
H H
S1'
O
O O OH
H H H
N N N COOH
N N
H H
S4 O O O

S3 S2 '
S1

C. Coordination of the Asp25 residues by the transition state


analog pepstatin.

FIGURE 38.17 Principles of the design of transition-state analog inhibitors of HIV protease.

the P groups are termed S1, S2, S1′, S2′, and so on. In the most useful of these are selective for the viral enzyme
active site, a pair of aspartates (the Asp25 on each of the HIV-1 protease. Structurally, these agents are either pep-
two subunits of the protein) work together to complete tidomimetic or nonpeptide compounds. Their effective-
the reaction (Fig. 38.17B). One coordinates the water ness is related to their ability to inhibit the processing of
that performs the hydrolysis, while the other coordinates the gag-pol precursor polyprotein to p24, p55, and p160.
the carboxyl group on the P1′ amino acid. Rationally Consequently, the infectivity of HIV-1 is diminished.
designed drugs that inhibit HIV protease are designed Although, some compounds exhibit in vitro and in vivo
as transition-state mimics that align at the active site of antiviral activities, optimization of their pharmacokinetic
HIV-1 protease, as defined by three-dimensional crystal- and pharmacodynamic properties has presented major
lographic analysis of the protein structure. Figure 38.17C problems. In view of the great demand for successful
shows the aspartic protease inhibitor pepstatin bound in anti-AIDS drugs, the FDA has approved the protease
the HIV protease active site. Pepstatin is an inhibitor of inhibitors described in the following sections using the
all aspartic proteases, but can be used to illustrate the accelerated approval process. The structures of cur-
mechanism by which transition-state analogs inhibit HIV rently marketed HIV protease inhibitors are shown inn
protease (101). Pepstatin contains an unnatural amino Figure 38.20.
acid core known as statine (Fig. 38.18), and the hydroxyl-
bearing sp3 carbon of this core mimics the tetrahedral
transition state that occurs during hydrolysis of the pep-
tide bond. Other transition-state cores have been used in R1
H
O
the design of highly potent aspartic protease inhibitors, N
N
N
including the hydroxyethylene and hydroxyethylamine H H
O R2 OH O
cores (Fig. 38.18). The crystal structures of several HIV
Native peptide Statine
protease inhibitors bound to wild-type and mutant HIV
proteases have been solved by x-ray crystallography. The
structure of the HIV protease inhibitor lopinavir bound R1 O R1 R2
to wild-type HIV protease is depicted in Figure 38.19. N N N
H H H
Note that the hydroxyl group of the transition-state core OH R2 OH O
is hydrogen bonded to the two Asp25 carboxylate moi-
Hydroxyethylene Hydroxyethylamine
eties at a distance of about 3 Å. A number of oligopep-
tide-like analogs have been synthesized that differentially FIGURE 38.18 Transition-state mimics used in the design of HIV
inhibit viral and mammalian aspartic proteases, and the protease inhibitors.
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1297

..ccoomm
u
u s
s e
e
K Ka
a d
d
ww..
wwww
FIGURE 38.19 Model of the HIV protease inhibitor lopinavir bound to wild-type HIV protease.

m
m
Potential Drug Interactions not slow progression of disease, CD4+ cell counts were

e
e . o
o
Protease inhibitors have a high potential for drug inter-

. cc
actions, stemming from the fact that they are substrates
for and inhibitors of the CYP3A4 enzyme system. As a
increased in patients infected with HIV in the United
States and European countries. Triple therapy with saqui-
navir, ZDV, and ddC has been more effective than double

dd us
s
result, concurrent use of protease inhibitors with other

u
drugs metabolized by CYP3A4 may be contraindicated,
therapy with saquinavir plus ZDV or ddC. Thus, combi-
nation therapy slowed disease progression and mortality.

aa
and in some cases, the resulting drug interactions can The IC50 concentration of saquinavir in both acutely

KK
be life-threatening. The most potent CYP450 inhibitor and chronically infected cells was 1 to 30 nmol/L. In

ww..
in this class is ritonavir (used to advantage in combina-
tion with lopinavir and other HIV protease inhibitors),
combination with ZDV, ddC, or ddI, the activity of saqui-
navir was increased without increased cytotoxicity. The

ww
followed by indinavir, nelfinavir, and amprenavir (mod-

ww
erate inhibitors) and saquinavir (the least potent inhibi-
tor). Drug interactions have been reported with bepridil,
dihydroergotamine, and a number of benzodiazepines.
Marked increases in the activity of amiodarone, lidocaine
resistance of HIV isolates to saquinavir was observed due
to substitution mutations in the HIV protease at amino
acid positions 48 (glycine to valine) and 90 (leucine to
methionine).

(systemic), quinidine, the tricyclic antidepressants, and Pharmacokinetics The bioavailability of saquinavir in a
warfarin might be expected (102). Other interactions single 600-mg dose following a high-fat meal was shown
have been reported with rifampin, rifabutin, phenobar- to be about 4%. Approximately 30% of a 600-mg dose of
bital, phenytoin, dexamethasone, or carbamazepine. saquinavir reached the liver, where it showed first-pass
Because the protease inhibitors are themselves metabo- metabolism. The metabolites, various mono- and dihy-
lized by CYP450, their action may be altered by other droxylated compounds of which the major metabolites
agents that induce or inhibit this system. In the case of are the monohydroxylated products shown in Figure
rifabutin, which inhibits CYP3A4 in the gut, relative bio- 38.21, are not active. Approximately, 88% and 19% of a
600-mg oral dose was found in the feces and urine, respec-

m
m
availability of the protease inhibitors is increased, and the

o
o
dose of the protease inhibitor may need to be decreased. tively. The volume of distribution after IV administration

c
c
of a 12-mg dose of saquinavir was 700 L. The drug is 98%

sse
e ..
HIV Protease Inhibitors that are Used Clinically
bound to plasma proteins, and a very low concentration
of saquinavir was found in CSF. The steady-state AUC was

uu
(Fig. 38.20) 2.5 times higher than that observed after a single dose
SAQUINAVIR MESYLATE

K Kaadd
Clinical Application Saquinavir was the first prote-
of 600 mg in HIV-infected patients after a meal as com-
pared to multiple dosing. Saquinavir has a plasma half-life

..
ase inhibitor approved by the FDA in December 1995 of approximately 1.8 hours. Although saquinavir hard-gel
(103,104). It is a carboxamide derivative used in the capsule in combination with other antiretroviral drugs

wwww
treatment of advanced HIV infection in selected patients.
Saquinavir is used concomitantly in either ZDV-untreated
reduced the risk of disease progression or death, it has
limited bioavailability. To overcome this limitation, the

ww
patients or ddC-treated patients previously treated with
prolonged ZDV therapy. Although combined therapy did
Kaduse.com
FDA has approved saquinavir soft-gel capsules. Saquinavir
is well tolerated in combination with ZDV and/or ddC
1298 PART III / PHARMACODYNAMIC AGENTS

O O O
N O H H N OH OH
H C NH N N H
N N N N O N N
N N H H N
H H CH3 O OH
O S S C O
OH
O NH-C(CH3)3
H N
O NH2
H2SO4 H2SO4
H3C SO3H

Saquinavir Ritonavir Indinavir

O
Ca2 O P
S NH2 O NH2
O OH O
CH3 O H H
C NH O N N O N N
HO O S O S
N N O O O O
H H O O
OH
H
H3C SO3H

Nelfinavir Amprenavir Fosamprenavir calcium

N OH
H O
CH3 N
OH O O OH O S
H H H O
N N NH N N N O
O N CH3O N N CH3 O O N
H H H
CH3 O O O O
CF3
Lopinavir Atazanavir Tipranavir

NH2
OH
H
O O N N
S
O H2C O O
O

Darunavir

FIGURE 38.20 Structures of HIV protease inhibitors that are used clinically.

and has few side effects, but GI disturbances were com- solution indicated AUC values of 129.5 ± 47.1 and 129.0 ±
mon adverse effects. Saquinavir has a few mild side effects, 39.3 mg/hour/mL, respectively, when a 600-mg dose was
such as headache, rhinitis, nausea, and diarrhea. given under nonfasting conditions. Five ritonavir metab-
olites have been isolated from human urine and feces.
RITONAVIR Ritonavir is another HIV protease inhibitor The isopropylthiazole oxidation product was the major
that was approved by the FDA in March 1996 (Fig. 38.20) active metabolite (M2) (Fig. 38.22).
(105). Ritonavir is a peptidomimetic inhibitor of both As with saquinavir, ritonavir is metabolized by CYP3A4
the HIV-1 and HIV-2 proteases. A 50% reduction in viral and is an inhibitor of the CYP450 system. Ritonavir is
replication was obtained at a 3.8 to 153 nmol/L concen- contraindicated with several compounds, such as clar-
tration of ritonavir. ithromycin, desipramine, ethinyl estradiol, rifabutin, sul-
famethoxazole, and trimethoprim because of increased
Pharmacokinetics After a 600-mg dose of an oral solu- concentrations of these drugs in the plasma due to inhib-
tion, peak concentrations of ritonavir were obtained in ited oxidative metabolism. Ritonavir alone or in combi-
approximately 2 or 4 hours under fasting or nonfasting nation with 3TC, ZDV, saquinavir, or ddC increased CD4+
conditions, respectively. Under nonfasting conditions, cell counts and decreased HIV RNA particle levels. Cross-
peak ritonavir concentrations decreased 23%, and the resistance between ritonavir and RT inhibitors is unlikely
extent of absorption decreased 7% relative to fasting because of the different mode of action and enzyme
conditions. In two separate studies, the capsule and oral involved. Common adverse reactions, such as nausea,
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1299

isolates and clinical resistant virus to indinavir analogs, is


H C CH3
O 3 C 25 to 100 nmol/L in drug combination studies with ZDV
Saquinavir N O H CH H C NH CH2 and ddI. However, HIV has shown ritonavir resistance
H 2
N
N N OH in some patients. This resistance is due to mutation of
H
O H CH2H H OH the virus that is correlated with the expression of amino
H acid substitutions in the viral protease. Cross-resistance to
O NH2
indinavir is observed with other protease inhibitors but
M-7 not with the RT inhibitor. For this reason, indinavir is
beneficial with ZDV and other nucleoside drugs.

Pharmacokinetics Indinavir is rapidly absorbed in fast-


H C CH3 ing patients, and plasma peak concentration is observed
O 3 C
N O H H C NH CH3
H CH2 in about 1 hour. At a dose of 800 mg every 8 hours, peak
N
N N
H
plasma concentration is approximately 300 nmol/L. The
O H CH2H H OH
drug is approximately 60% bound to human plasma pro-
H
O NH2 teins. Indinavir is metabolized via oxidation and glucuro-
HO nide conjugation. These metabolites were recovered in
M-2
feces and urine, with about 20% of the drug excreted in the
urine. The half-life of indinavir is approximately 1.8 hours.
FIGURE 38.21 Major metabolic products from saquinavir. Because of indinavir’s metabolism, a number of drug
interactions are possible. Indinavir interacts with rifabu-
tin or ketoconazole, leading to increased or decreased
diarrhea, vomiting, anorexia, abdominal pain, and indinavir concentration, respectively, in the blood plasma.
neurologic disturbances, were reported with the use of Administration of drug combinations of indinavir with
ritonavir alone or in combination with other nucleoside antiviral nucleoside analogs, cimetidine, quinidine, tri-
analogs. Ritonavir is used for the treatment of advanced methoprim–sulfamethoxazole, fluconazole, or isoniazid
HIV infection including opportunistic infections. In resulted in an increased activity of indinavir. Indinavir is
combination with nucleoside drugs, ritonavir reduced contraindicated in patients taking triazolam or midazolam
the risk of mortality and clinical progression. because inhibition of metabolism of these drugs may result
in prolonged sedation, nephrolithiasis, asymptomatic
INDINAVIR SULFATE Indinavir, a pentanoic acid amide hyperbilirubinemia, and GI problems (anorexia, constipa-
derivative, was approved by the FDA in March 1996 tion, dyspepsia, and gastritis). The usual oral dose for indi-
(Fig. 38.20) (106). The 95% inhibitory concentration navir alone or in combination with other antiviral agents
against laboratory-adapted HIV variants, primary clinical is one 800-mg capsule every 8 hours. The drug is well
absorbed if given on an empty stomach or 1 hour before or
2 hours after a light meal with water. The dose is reduced to
H3C CH3
600 mg every 8 hours if given concurrently with ketocon-
O
H
CH2 azole. Indinavir activity is increased when combined with
N N RT inhibitors.
N N NH2
Ritonavir CH3
H
O H 2C OH
S
NELFINAVIR MESYLATE Nelfinavir is a peptidomimetic drug
M1
that is effective in HIV-1 and HIV-2 wild-type and ZDV-
resistant strains (Fig. 38.20). ED50 concentrations range
from 9 to 60 nmol/L (95% effective dose was 0.04 mg/mL)
H3C CH3
O
H
CH2 O (107). After IV administration, the elimination half-life
N N N
N
N O
of nelfinavir was about 1 hour. In combination with D4T,
HO H H nelfinavir reduced HIV viral load by about 98% after
CH3 O H2C OH S
S
4 weeks. It is well tolerated when used with azole anti-
M2 N fungals (ketoconazole, fluconazole, or itraconazole) or
macrolide antibiotics (erythromycin, clarithromycin, or
H3C CH3 azithromycin). However, it causes diarrhea and other
O CH2 O
H
H
N
side effects common to nonnucleoside drugs. Following
N N N O oral administration, nelfinavir peak levels in plasma
H H
CH3 O H2C OH S ranged from 0.34 mg/mL (10 mg/kg in the dog) to
N 1.7 mg/mL (50 mg/kg in the rat). Nelfinavir was slowly
M11
absorbed, and bioavailability was 47% in the dog. The
FIGURE 38.22 Major metabolic products from CYP3A4 metabo- drug appeared to be metabolized in the liver, and the
major excretory route was in feces.
lism of ritonavir.
Kaduse.com
1300 PART III / PHARMACODYNAMIC AGENTS

AMPRENAVIR Amprenavir is the fifth in a series of pro- agents, have been approved for use in adults and patients
tease inhibitors to be approved for marketing in the between the ages of 6 months and 12 years. This is the first
United States. While structurally unique from the previ- protease inhibitor to be indicated for the very young.
ous agents, its pharmacologic profile does not appear to
differ significantly from the previously marketed agents. ATAZANAVIR CALCIUM (109) Atazanavir is an antiretroviral
Early studies suggest that a different resistance profile agent approved for use in combination with other anti-
may exist and that the drug may be effective against some retroviral agents for the treatment of HIV infections
resistant strains of HIV. Side effects appear to be more (Fig. 38.20). It targets HIV-1 protease and reduces viral
common than with other protease inhibitors and include replication and thus virulence of HIV-1. Similar to saqui-
nausea, vomiting, paresthesia, depression, and rash. navir, ritonavir, indinavir, nelfinavir, amprenavir, and lopi-
Because amprenavir is a sulfonamide, there is some con- navir, the drug is used in combination with RT inhibitors
cern for cross-sensitivity with antibacterial sulfonamides. to produce excellent efficacy in AIDS patients. Atazanavir
Although this has not been reported, care should be is dosed orally once daily, thus reducing “pill burden,” and
taken if sensitivity to trimethoprim–sulfamethoxazole, seems to have a minimum impact on lipid parameters, but
used in Pneumocystis jirovecii pneumonia, is reported. does increase total bilirubin. The drug is well absorbed
when administered orally with food (bioavailability ∼68%).
Pharmacokinetics Amprenavir is rapidly absorbed follow- The drug is highly bound to plasma protein (86%) and is
ing oral administration and may be taken with or without metabolized by CYP3A isoenzyme. Atazanavir is a moder-
food. High-fat meals decrease the absorption of the drug ate inhibitor of CYP3A, and potential drug–drug interac-
and therefore should be avoided. The product is avail- tions are possible with CYP3A inhibitors and inducers.
able in capsule and liquid form. The recommended adult
and adolescent dose of 1,200 mg twice daily requires the TIPRANAVIR Tipranavir is a recently approved, nonpep-
patient to take eight capsules (150 mg) twice daily. The liq- tidic inhibitor of HIV protease (Fig. 38.20) (110). It is
uid preparation is recommended for children between 4 indicated for patients who have developed resistance to
and 12 years of age or for patients 13 to 16 years of age who other antiretroviral drugs, including other HIV protease
weigh less than 50 kg. The dose is 22.5 mg/kg twice daily or inhibitors. Although it is very potent, it also exhibits more
17 mg/kg three times a day. Since this preparation contains severe side effects than other HIV protease inhibitors,
the excipient propylene glycol, it is not recommended for including intracranial hemorrhage, hepatitis, and diabe-
children less than 4 years of age and certain other individu- tes mellitus. It is dosed at 500 mg twice daily in combina-
als who are unable to metabolize this alcohol. tion with 200 mg of ritonavir to suppress metabolism by
cytochrome P450 (111). There is evidence that tipranavir
FOSAMPRENAVIR CALCIUM (108) Fosamprenavir has been induces its own metabolism, but using twice-daily coad-
approved for the treatment of HIV in adults when used ministration of tipranavir and ritonavir at varying doses,
in combination with other anti-HIV drugs. It is a prodrug tipranavir levels were 24- to 70-fold higher than levels
that, upon hydrolysis by serum phosphatases, gives rise to achieved with administration of tipranavir alone.
amprenavir, which as indicated earlier is a peptidomimetic
transition-state inhibitor that targets HIV-1 protease and
DARUNAVIR Darunavir is a second-generation HIV protease
reduces viral replication and, thus, infectiousness of HIV-1.
inhibitor that can be used in treatment-naive and treatment-
It is commonly administered in combination with RT inhib-
experienced adult and pediatric patients (Fig. 38.20) (112).
itors to produce excellent efficacy in AIDS patients. The
It was designed to bind tightly and specifically to HIV pro-
drug is administered as two 700-mg tablets twice daily or,
tease and thereby overcome the problems associated with
in combination with ritonavir, can be given as two 700-mg
first-generation drugs, including toxicity, high dose, expen-
tablets once daily or one 700-mg tablet twice daily.
sive manufacture, and susceptibility to resistance. Like
Fosamprenavir is a slow-release version of amprenavir and,
other HIV protease inhibitors, darunavir is metabolized by
as a result, decreases the pills required for the patient and
cytochrome P450, and thus, the drug is dosed at 800 mg
lowers the “pill burden” in AIDS patients.
once daily in combination with 100 mg of ritonavir (113).
LOPINAVIR/RITONAVIR Recently, the FDA has approved the
HIV Integrase Inhibitors
release of lopinavir/ritonavir combination in patients who
have not responded to other regimens for treatment of RALTEGRAVIR
HIV. The product is available in a soft gelatin capsule con- O O
taining 133.3 mg of lopinavir and 33.3 mg of ritonavir, as N N
N N N
well as oral solutions containing 80 mg/mL of lopinavir H H
O N
and 20 mg/mL of ritonavir. The small amount of ritonavir H3C H3C OH F
is not expected to have antiretroviral activity, but rather the O

ritonavir is meant to increase the plasma concentrations of


lopinavir by inhibiting lopinavir’s metabolism by CYP3A4. Mechanism of Action Raltegravir (Isentress) is the first
These drugs, in combination with other antiretroviral (and thus far only) FDA-approved treatment for HIV that
Kaduse.com
CHAPTER 38 / ANTIVIRAL AGENTS AND PROTEASE INHIBITORS 1301

acts by inhibition of HIV integrase (114). A second HIV F

integrase inhibitor, elvitegravir, is currently in phase III


clinical trials. Like HIV protease, HIV integrase is essen-
tial for replication of the HIV virus and, as such, has
O NH
become a validated target for drug discovery. Each HIV N N
particle contains 40 to 100 integrase molecules the role N N
H
O O

of which is to facilitate the insertion of viral complemen- O N Me2- R Acid residue


H3C H3C O in integrase
O
tary DNA (cDNA) into the host cell genome. Following O
host cell infection, the viral RNA is converted to cDNA via Raltegravir
the action of RT. The preparation of cDNA for insertion
requires integrase-mediated trimming of the 3′-ends of the FIGURE 38.23 Chelation complex between raltegravir and inte-
viral DNA (3′-processing). The 3′-processing occurs within grase.
the cytoplasm, and the processed cDNA plus integrase
complex, which also contains viral and cellular protein, is antiretroviral therapy drugs (see next section), and in
referred to as the preintegration complex. This complex 2009, the FDA expanded approval for use in all patients.
is transported into the nucleus of the host cell where inte- It is taken twice daily, and doses of 200, 400, and 600 mg
gration into the host DNA occurs through the catalytic have been used.
action of integrase. The 3′-OH of the cDNA is joined to
the 5′-phosphate of the host chromosome with both ends Combination Drug Therapy (115,116)
of the cDNA being joined to the host DNA. This very com-
When several antiretroviral drugs, typically three or four,
plex integration of cDNA into the host genome involves
are taken in combination for the treatment of HIV, the
numerous viral and host proteins. Integrase itself is a 32-kd
approach is known as highly active antiretroviral therapy
protein with three structural domains: 1) amino-terminal
(HAART). Multiple HAART therapies have been under-
domain; 2) catalytic core domain (CCD); and 3) carboxy-
taken, and the National Institutes of Health recommends
terminal domain. All three domains are essential for inte-
that all infected patients should be treated in this man-
gration, whereas only the CCD is required for the reverse
ner (114). The synergistic antiviral effects of rimantadine
reaction referred to as disintegration. The amino-terminal
with ribavirin and tiazofurin against influenza B virus and
domain binds zinc, while the CCD binds manganese
ganciclovir with foscarnet against HSV-1 and HSV-2 are
(Mn2+) and magnesium (Mg2+). It has been proposed that
noteworthy. The synergistic action of either trifluorothy-
raltegravir acts by inhibiting cDNA integration via chela-
midine or acyclovir with leukocyte interferon has been
tion to the divalent cations in the CCD at the interface the
used in the topical treatment of human herpetic keratitis.
integrase-donor-acceptor complex (Fig. 38.23).
During the past decade, research into combination anti-
retroviral therapy for AIDS patients has made remarkable
Pharmacokinetics Raltegravir is readily absorbed fol-
progress. ZDV, the first approved drug for HIV-infected
lowing oral administration. The drug is highly bound
patients, produced bone marrow toxicity. To overcome
to plasma protein (∼83%). Food does not appear
toxic effects, combinations of ZDV with foscarnet, ddC, or
to affect the rate of absorption. The major route of
ddI have been used. Such combination therapy indicated
metabolism is glucuronidation mediated by uridine
improved efficacy and decreased side effects as compared
diphosphate-glucuronosyl-transferase.
to either drug used alone. The combination of ZDV with
O O α-interferon has been used to treat patients with AIDS-
N
N
N
N
N
related Kaposi sarcoma. This combination drug therapy
H
N
H
O delayed emergence of ZDV-resistant HIV strains.
F COOH O CH3 CH3 A combination of granulocyte-macrophage colony-
O O
OH stimulating factor with ZDV and α-interferon has been
HO successful in managing treatment-related cytopenia in
OH HIV-infected patients. The advantages of combination
therapy include therapeutic antiviral effect, decreased
Approximately 51% of the drug is recovered in the feces toxicity, and low incidence of drug-resistant infection.
unchanged, and 32% is recovered in the urine where In recent years, emergence of drug resistance has been
23% is the glucuronide. Glucuronidation inhibitors have demonstrated in patients receiving single antiviral agent
the potential for drug–drug interactions, although there therapy. Resistance to amantadine, acyclovir, ribavirin,
was no clinically significant effect when administered ganciclovir, ZDV, and other antiviral agents is noteworthy.
with atazanavir. Raltegravir is recommend for use in com- Combined antiretroviral drug therapy serves different
bination therapy and in patients who have demonstrated purposes. It prolongs the life of AIDS patients, removes
resistance to previous regimens. drug resistance, and/or reduces toxicity of drugs. With
Raltegravir was approved in 2008 for use in individu- these objectives, successful combinations of ZDV have
als whose infection has proven resistant to highly active been reported with ddC, ddI, 3TC, or D4T. Recently,
Kaduse.com
1302 PART III / PHARMACODYNAMIC AGENTS

combinations of nucleosides drugs (ZDV, ddC, ddI, 3TC) double-stranded duplexes 21 to 25 nucleotides in length
are used with protease inhibitors (saquinavir, indinavir, are created from a parent double-stranded RNA molecule
ritonavir) for delaying HIV infection. Combined nucleo- by an enzyme known as dicer. These short interfering RNAs
side drugs are known to delay progression of HIV infection. (siRNA) bind to specific mRNAs that feature a complimen-
Antiretroviral therapy includes nucleosides or non- tary sequence to form an RNA-induced silencing complex,
nucleoside RT inhibitors and protease inhibitors. These and thereby block the expression of predetermined spe-
drugs inhibit HIV replication at different stages of viral cific proteins at the posttranscriptional level. This bind-
infection. Nucleoside and nonnucleoside drugs inhibit RT ing also appears to direct the cell to cleave target mRNA/
by preventing RNA formation or viral protein synthesis. siRNA complexes. Many research groups are attempting
Nonnucleoside drugs inhibit RT by inactivating the cata- to develop RNAi therapies that induce the degradation of
lytic site of the enzyme. Protease inhibitors act after HIV target mRNA involved in inherited or acquired disorders.
provirus has integrated into the human genome. These This technology is especially well suited to treating viral
drugs inhibit protease, which is an enzyme responsible for infections, and numerous examples now illustrate that a
cleaving viral precursor polypeptides into effective virions. wide range of viruses can be inhibited with RNAi, both in
Thus, protease inhibitors combined with RT inhibitors act vitro and in vivo. Antiviral RNAi therapies can be tailored to
by a synergistic mechanism to interrupt HIV replication. the biochemical characteristics of each pathogen and can
Two-drug combinations, such as ZDV plus ddI or ddC, be made more specific through choice of delivery vehicle,
3TC plus ZDV, and D4T plus ddI, have been successful in route of administration, selection of gene targets, and reg-
raising CD4+ cell counts and decreasing HIV RNA viral ulation of RNAi induction. As mentioned earlier, success-
load. Triple-drug therapy consisting of ZDV, 3TC, and a ful antiviral therapeutics possess the ability to discriminate
protease inhibitor (indinavir, ritonavir, nelfinavir) has been virus from host. However, because viruses rely extensively
more effective than two-drug therapy consisting of two on host cell machinery for many functions and activities
nonnucleoside analog combinations. In addition, fewer involved in viral replication, they offer a very limited num-
opportunistic infections were noted when patients took the ber of therapeutic targets. Because RNAi specifically targets
three-drug combination. ZDV can also be combined with a short stretch of viral nucleic acids rather than a viral pro-
immunomodulators to increase immunologic response in tein, even a small viral genome can provide a large number
AIDS patients. ZDV has been combined with α-interferon of potential targets.
to obtain synergistic activity of the drug. An ideal approach Researchers are continuing to look for suitable siRNA
of combined antiretroviral drug therapy would be drugs treatments for viral infections such as HBV, HCV, and
acting at different stages of HIV cell replication. HIV. Despite extensive research, the development of an
siRNA-based therapy for viral infection faces significant
Investigational Antiviral Agents: Short Interfering barriers including poor siRNA stability, inefficient cellular
RNA (117) uptake, widespread biodistribution, activation of immune
The field of directed RNA interference (RNAi) has rapidly response, and occurrence of nonspecific (i.e., off-target)
developed into a highly promising approach for specifically effects (118). The most notable of these barriers is the
interrupting gene function in order to alleviate disease delivery of intact siRNA molecules to the interior of virus-
pathology. This technique was first described in 1998, and infected cells. A number of approaches have been tried,
the inventors of this technology, Andrew Fire and Craig C. including bioconjugation of siRNA with lipids, incorpora-
Mello, were awarded a Nobel Prize in 2006 for their work tion into nanoparticles, and in situ biosynthesis of siRNAs.
with RNAi. RNAi is a natural mechanism for silencing gene Although this strategy for antiviral development shows
expression that has been conserved through evolution in great promise, a reliable siRNA-based therapy for viral
eukaryotes ranging from plants to humans. In this process, infection will likely not be realized in the near future.

SCENARIO: OUTCOME AND ANALYSIS


Outcome Chemical Analysis
Douglas Slain, PharmD, BCPS Victoria Roche and S. William Zito
WM was started on the investigational zanamivir intravenous These three antiviral molecules all take advantage of the research
formulation. After about 5 days of therapy, the patient became conducted to map the binding site of the influenza virus activat-
clinically stable and was able to come off of mechanical ventila- ing enzyme neuraminidase. Neuraminidase facilitates the spread
tion. She ultimately recovered. Resistance testing did show that of shed viruses by cleaving sialic acid (N-acetylneuraminic acid)
she had an H1N1 strain that had the H274Y mutation. from the saccharide-coated viral particles, which promotes their
receptor-mediated absorption into unaffected cells.

Kaduse.com

Вам также может понравиться