Вы находитесь на странице: 1из 132

Haematology FOURTH EDITION

AN ILLUSTRATED COLOUR TEXT

tahir99-VRG & vip.persianss.ir


Content Strategist: Jeremy Bowes
Content Development Specialist: Fiona Conn
Project Manager: Srividhya Vidhya Shankar
Design Direction: Greg Harris
Illustration Manager: Jennifer Rose

tahir99-VRG & vip.persianss.ir


Haematology FOURTH EDITION

AN ILLUSTRATED COLOUR TEXT

Martin R Howard
MBChB MD FRCP FRCPath
Consultant Haematologist
York Teaching Hospital NHS Foundation Trust
Clinical Senior Lecturer
Hull York Medical School
R G
York, UK

- V
Peter J Hamilton 9 . i r
MA BM BCh FRCP FRCPath (retired)
i r 9 & s s
Formerly Consultant Haematologist
a h i a n
Royal Victoria Infirmary
Lecturer in Medicine
t r s
University of Newcastle-upon-Tyne
p e
Newcastle-upon-Tyne, UK .
Illustrated by Robert Britton and Antbits Ltd. vip

EDINBURGH  LONDON  NEW YORK  OXFORD  PHILADELPHIA  ST LOUIS  SYDNEY  TORONTO  2013

tahir99-VRG & vip.persianss.ir


iv

© 2013 Elsevier Ltd. All rights reserved.

No part of this publication may be reproduced or transmitted in any form or by any means,
electronic or mechanical, including photocopying, recording, or any information storage and retrieval
system, without permission in writing from the publisher. Details on how to seek permission,
further information about the Publisher’s permissions policies and our arrangements with
organizations such as the Copyright Clearance Center and the Copyright Licensing Agency, can be
found at our website: www.elsevier.com/permissions.

This book and the individual contributions contained in it are protected under copyright by the
Publisher (other than as may be noted herein).

First edition 1997


Second edition 2002
Third edition 2008
Fourth edition 2013

ISBN 978-0-7020-5139-5
ebook ISBN 978-0-7020-5415-0

British Library Cataloguing in Publication Data


A catalogue record for this book is available from the British Library R G
Library of Congress Cataloging in Publication Data
- V
9
A catalog record for this book is available from the Library of Congress

. i r
i r 9 & s s
Notices
a h i a n
t
Knowledge and best practice in this field are constantly changing. As new research and

r s
experience broaden our understanding, changes in research methods, professional practices, or
medical treatment may become necessary.

e
Practitioners and researchers must always rely on their own experience and knowledge in

p
.
evaluating and using any information, methods, compounds, or experiments described herein. In

vip
using such information or methods they should be mindful of their own safety and the safety of
others, including parties for whom they have a professional responsibility.
With respect to any drug or pharmaceutical products identified, readers are advised to check
the most current information provided (i) on procedures featured or (ii) by the manufacturer of
each product to be administered, to verify the recommended dose or formula, the method and
duration of administration, and contraindications. It is the responsibility of practitioners, relying
on their own experience and knowledge of their patients, to make diagnoses, to determine
dosages and the best treatment for each individual patient, and to take all appropriate safety
precautions.
To the fullest extent of the law, neither the Publisher nor the authors, contributors, or editors,
assume any liability for any injury and/or damage to persons or property as a matter of products
liability, negligence or otherwise, or from any use or operation of any methods, products,
instructions, or ideas contained in the material herein.

The
publisher’s
policy is to use
paper manufactured
from sustainable forests

Printed in China

tahir99-VRG & vip.persianss.ir


Preface to the fourth edition
The principles that guided the first This edition has been considerably enthusiastic student to test their
edition of this book sixteen years ago updated to acknowledge recent learning. These are intended to be
still apply. In the era of whole genome developments in the understanding and enjoyed. Although it can seem
sequencing and targeted therapies, the treatment of benign and malignant complicated, haematology remains a
proper clinical management of patients diseases of the blood. The addition of a fascinating specialty which well repays
with blood disorders relies very heavily section addressing ‘Haematology in the attempts at understanding.
on traditional skills: history taking, Elderly’ reflects the increasing age of
clinical examination and careful the population. A number of case MRH
selection of laboratory investigations. histories have been added to allow the

tahir99-VRG & vip.persianss.ir


Preface to the first edition
Blood is a ‘hot, temperate, red humour that the format, with double page We have stressed recent advances in
whose office is to nourish the whole spreads and extensive colour technology in the laboratory and newer
body to give it strength and colour, illustration, will allow information treatment strategies on the ward.
being dispersed by the veins through to be absorbed painlessly and with However, if this book has a ‘message’ it
every part of it,’ wrote Richard Burton enjoyment! The text is designed to be is that best practice and management
in 1628. Studying the red humour can suitable for medical students, junior of blood diseases still relies heavily on
be hard work. Complex nomenclature hospital doctors, general practitioners, traditional skills – history taking,
and classifications make haematology biomedical scientists, and nurses clinical examination, and careful
seem tedious and unintelligible to the with a special interest in haematology. selection of laboratory investigations.
uninitiated. The object of this book is to Those taking higher medical
give a basic grounding in the biology examinations should find it a MRH, PJH
and diseases of the blood. We hope useful revision aid.

tahir99-VRG & vip.persianss.ir


 v

Acknowledgements
We are grateful to the following Saunders (Newcastle-upon-Tyne); Mr M Laboratories at York Teaching Hospital
colleagues for their advice and help Cullen, Mrs H Dickinson, Dr D Norfolk, and The Royal Victoria Infirmary,
with illustrations: Dr A Anderson, Dr L Mrs S Ricketts, Dr A Scarsbrook Newcastle-upon-Tyne; the Medical
Bond, Dr A Clarke, Dr I N Reid, Dr R. (Leeds); Prof D Grimwade, Dr E Letsky, Illustration Department at York
Mannion, Dr A Turnbull, Dr H Dr J Marsh (London); Prof E Preston Teaching Hospital. Thanks to all
Wilkinson (York); Dr A Hall, Dr A (Sheffield); Dr P Bolton-Maggs, Mrs H involved at Elsevier.
Lennard, Dr M Reid, Dr P W G Jones (SHOT); Haematology

tahir99-VRG & vip.persianss.ir


2 1 ANATOMY AND PHYSIOLOGY

1 The bone marrow


In early fetal life, blood is produced in
the mesoderm of the yolk sac. During 100 Yolk sac Bone
the second to seventh months the liver marrow Vertebrae
and spleen take over. Only in the last 2 and pelvis
Liver
months of fetal development does the

Blood production (%)


bone marrow become the predominant
site of blood formation. During child- Sternum
hood, marrow in the more peripheral 50
bones becomes gradually replaced by
fat, so that in adult life over 70% is Rib
Spleen
located in the pelvis, vertebrae and
sternum (Fig 1.1). This explains the sites Femur
Tibia
used for bone marrow sampling (see
0
p. 106). 0 1 2 3 4 5 6 7 8 9 10 20 30 40 50 60 70
Prenatal (months) Postnatal (years)
Birth
The structure of the Fig 1.1  Sites of blood production in the fetus and after birth.
bone marrow
A trephine biopsy allows a two-
dimensional view of the bone marrow
down the light microscope (Fig 1.2). Haematopoietic cells of
varying lineage and maturity are packed between fat spaces
and bony trabeculae. Ultrastructural studies reveal clusters of
haematopoietic cells surrounding vascular sinuses which
allow eventual discharge of mature cells into the blood. Dif-
ferent lineages are compartmentalised; for example, the most
immature myeloid precursors lie deep in the marrow paren-
chyma while more mature forms migrate towards the sinus
wall. Lymphocytes tend to surround small radial arteries
while erythrocytes form islands around the sinus walls.
Blood precursor cells in the marrow exist in close proximity
to stromal cells. Stromal cells are those cells which do not
Fig 1.2  Normal bone marrow. Light microscopy of bone marrow
mature into the three main types of peripheral blood cells – trephine biopsy.
thus they include macrophages, fat cells, endothelial cells and
reticulum cells.
Immature blood cells are attached to these stromal cells by
multiple cellular adhesion molecules (e.g. fibronectin and col- detectable precursor cell creates granulocytes, erythrocytes,
lagen). Adhesive molecules have specific receptors on stromal monocytes and megakaryocytes and is thus called CFUGEMM.
and haematopoietic cells. As blood cells mature, the receptors HSCs may also be identified and separated from more com-
down-regulate and the cells become less adherent and com- mitted progenitors by the use of flow cytometry as they have
mence the journey through the sinus wall and into the a characteristic immunophenotype.
bloodstream. HSCs have the capacity for self-renewal as well as differen-
tiation and the system allows enormous amplification. A life-
time of human haematopoiesis with the generation of
Haematopoiesis: the stem cell hierarchy
incalculable numbers of mature cells may rely on only a few
Haematopoiesis means the formation of blood. A number of thousand stem cells present at birth. These cells depend on
transcription factors (e.g. GATA-1, MLL) are critical both for their micoenvironment, the ‘niche’, for regulation of self-
stem cell formation and function and lineage-specific renewal and differentiation. Both haematopoietic and stromal
differentiation. stem cells have the capacity to produce cells associated with
The first adult haematopoietic stem cells (HSCs) are gener- other tissues such as bone, liver, lung and muscle. This
ated in the aorto-gonad-mesonephros (AGM) region of the concept of ‘plasticity’ has therapeutic implications as stem
embryo. The classical hierarchy diagram (Fig 1.3) where all cells are used to repair a variety of damaged tissues.
cells arise in orderly fashion from a HSC is helpful but simpli-
fied; in reality, HSCs are groups of cells with diverse potentials
depending on transcription factors and the local microenvi-
Regulators of haematopoiesis
ronment. HSCs are not detectable by microscopic techniques Control of haematopoiesis is mediated via regulatory mole-
but their existence can be inferred from cell cultures. Culture cules (or ‘growth factors’ – Table 1.1). These are generally
of these early cells on agar generates groups of more mature glycoproteins produced by stroma and differentiated blood
and thus recognisable progenitor cells known as colony- cells. They may act on more than one cell lineage and fre-
forming units (CFUs). For myeloid development the earliest quently show additive and synergistic interactions with each

tahir99-VRG & vip.persianss.ir


The bone marrow 3

other. Their actions are multiple, includ-


ing the promotion of proliferation, dif- Self-renewal
Pluripotent
ferentiation and maturation, as well as LT-HSC stem cells
changing functional activity. Prolifera-
tive regulators alter the behaviour of
cells by interacting with specific recep-
tors on the cell surface (Fig 1.4).
Receptors for haematopoietic regula- ST-HSC
tors have been molecularly cloned and
many are related in structure (haemat-
Multipotent
opoietic receptor superfamily). The
CLP progenitors
combination of regulator and mem- CMP
brane receptor leads to a structural
change in the receptor and the trigger-
ing of a complex sequence of biochemi-
cal events (signal transduction). The end MEP GMP
result is the generation of intracellular
regulators in the cell cytoplasm which
have the capacity to activate genes,
which in turn encode proteins essential BFU-E CFU-Meg CFU-GM Pre B Pre-T
in cell activation.
Under normal circumstances regula- Committed
tors circulate in the plasma at virtually precursors
unidentifiable levels. The activities of
CFU-E CFU-G CFU-M
many factors are likely to be localised
and transient so that systemic levels are
of limited significance. For instance, in Mature
the marrow, regulators acting at the ear- cells
liest stages of haematopoiesis (e.g. c-kit Red Megakaryocyte Neutrophil Monocyte B-lymphocyte T-lymphocyte
ligand) are released from stromal cells cell
in close proximity to haematopoietic
precursor cells.
Platelets
The colony-stimulating factors (CSFs)
were originally defined by their ability Fig 1.3  The stem cell hierarchy. LT-HSC, long-term haematopoietic stem cell; ST-HSC, short-term
haematopoietic stem cell; CMP, common myeloid progenitor; CLP, common lymphoid progenitor; MEP,
to stimulate blood progenitor cells
megakaryocyte/erythroid progenitor; GMP, granulocyte/macrophage progenitor; CFU, colony-forming
while the interleukins (ILs) were defined unit; BFU, burst-forming unit; G, granulocyte; E, erythroid; M, monocyte; Meg, megakaryocyte.
by their effects on mature lymphocytes.
Subsequent discoveries have rendered Table 1.1  Key actions of some haematopoietic regulators
this dual nomenclature unhelpful – thus Growth factor Key actions
IL-3 is a key stem cell growth factor. The Interleukin-1 Mediates acute phase responses; cofactor for other growth factors
term cytokine incorporates all growth Interleukin-2 Growth factor for activated T-lymphocytes
factors. Interleukin-3 Supports early haematopoiesis by promoting growth of stem cells
c-kit ligand (stem cell factor) Interacts with other factors to stimulate pluripotent stem cells
Erythropoietin Lineage-specific growth factor promoting production of red cells
GM-CSF Growth factor promoting production of neutrophils, monocytes, macrophages,
eosinophils, red cells and megakaryocytes
G-CSF Lineage-specific growth factor promoting production of neutrophils
M-CSF Lineage-specific growth factor promoting monocyte and macrophage production
Thrombopoietin (Mpl ligand) Lineage-specific growth factor promoting platelet production

CSF, colony-stimulating factor; G, granulocyte; M, macrophage.


Haematopoietic Specific
regulator membrane
receptor
The bone marrow
■ The bone marrow is the site of blood formation (haematopoiesis)
after birth.
■ The cells recognisable in the blood are ultimately all derived from
2nd haematopoietic stem cells (HSCs) which reside in a bone marrow
messengers Gene Cell proliferation
activation Differentiation ‘niche’.
Maturation ■ Immature blood cells in the marrow are attached to stromal cells
Change in functional by multiple cellular adhesion molecules. Maturing blood cells are
Proteins activity
eventually released through vascular sinus walls into the
bloodstream.
■ Controlof haematopoiesis is mediated via transcription factors and
Fig 1.4  Schematic view of action of regulator on haematopoietic haematopoietic regulators.
cell. Second messengers include protein kinase C and calcium ions.

tahir99-VRG & vip.persianss.ir


4 1 ANATOMY AND PHYSIOLOGY

2 Red cells
The mature red cells of the blood transport the respiratory
gases, oxygen and carbon dioxide (CO2). Oxygen is carried
from the lungs to the tissues, where it is exchanged for CO2.
Red cells are equipped to perform this function for 120 days
during which they make a 300 mile journey around the
microcirculation.
Prior to discharge from marrow sinuses into the peripheral
blood, red cells shed their nuclei. This gives the advantages
of reduced weight and transformation into a biconcave disc
with increased deformability compared with the more rigid
spheroidal nucleated precursor (Fig 2.1).
The blood volume comprises the mass of red cells and the
plasma. Plasma volume is regulated by stretch receptors in
the heart and kidney which influence secretion of antidiuretic
hormone (ADH) and aldosterone. Erythropoiesis is regulated
chiefly by the growth factor erythropoietin.

Erythropoietin
Unlike other growth factors, erythropoietin is mainly synthe-
sised by the peritubular endothelial cells of the kidney. Pro-
duction is triggered by tissue hypoxia (lack of oxygen). Cells
can sense hypoxia via mediators such as the transcription
factor HIF (hypoxia-inducible factor). HIF activates genes vital
in the adaptive response to hypoxia including the erythropoi-
Fig 2.1  Scanning electron microscope picture of mature red cells
showing clearly the characteristic biconcave shape. (Copyright Dennis
etin gene. Erythropoietin molecules bind to specific mem- Kunkel Microscopy Inc.)
brane receptors on primitive erythroid cells in the bone
marrow and induce maturation. The increase in red cells
released into the blood stops when normal oxygen transport
is restored – this feedback circuit is illustrated in Figure 2.2.
Stem Erythroid
cells precursors
Structure
Bone marrow
The mature red cell is around 7.8 µm across and 1.7 µm thick.
Its biconcave shape allows maximum flexibility and an
umbrella shape is adopted to traverse the smallest capillaries Erythropoietin Red cell mass
which have diameters of only 5 µm. The ability of red cells
to recover from the recurrent stresses of the turbulent circula-
tion hinges on the design of the membrane. Kidney
The red cell membrane is composed of a collapsible lattice
Erythropoietin Oxygen
of specialised proteins (the ‘cytoskeleton’) and an outer lipid production sensor
bilayer (Fig 2.3). The protein skeleton is responsible for main-
taining red cell shape while the lipid bilayer provides a hydro-
phobic skin. The main skeletal proteins are spectrin, actin, Fig 2.2  Feedback circuit in production of erythropoietin.
proteins 4.1 and 4.2, and ankyrin. Spectrin is the most abun-
dant and consists of alpha and beta chains wound around
each other. Spectrin heterodimers can align at the ends to Glycophorin
form tetramers (i.e. four chains). Spectrin tetramers are joined
together by actin in association with protein 4.1. This flexible
skeleton is attached to the rest of the membrane by ankyrin,
which interacts with protein 4.2 to link the spectrin beta chain
Band Lipid bilayer
to the cytoplasmic end of the transmembrane protein band 3
3. The lipid bilayer consists mainly of a mixture of phospholi-
pids and cholesterol. Cholesterol molecules are inserted
between phospholipid molecules in such a way that they
stiffen the membrane while still allowing a degree of fluidity
between the bilayers. Cytoplasm
Defects of both the red cell membrane proteins and Protein 4.1 Ankyrin Spectrin Actin
lipids may lead to changes in red cell shape and premature Protein 4.2
destruction. Fig 2.3  The red cell membrane.
Red cells 5

in mature cells. The normal adult hae-


Metabolism Ageing and death
moglobin molecule (HbA) contains four
Red cells require an energy source to polypeptide chains (‘globin’ chains): the Beyond 100 days red cells start to show
maintain their structure and also a two alpha chains and two beta chains features of ageing including a declining
mechanism for detoxification of oxi- are often notated as α2β2. Combined rate of glycolysis, reduced levels of ATP
dants. Energy is provided by the with each of the polypeptide chains is a and membrane lipid, and a loss of flex-
Embden–Meyerhof pathway, a sequence ‘haem’ molecule which contains ferrous ibility. The terminal event is unclear but
of biochemical reactions in which iron (Fe2+) and protoporphyrin (Fig 2.4). effete cells are removed from the circula-
glucose is metabolised to lactate with The iron combines reversibly with tion by the macrophages of the liver and
the generation of two molecules of ade- oxygen and thus haem forms the spleen.
nosine triphosphate (ATP). ATP main- oxygen-carrying part of the molecule. Most of the catabolised haemoglobin,
tains the osmotic pressure of the cell by Other globin chains are formed by the particularly the iron, is reused (see also
driving sodium and calcium pumps in fetus and the change from fetal to adult p. 24). The protoporphyrin of haem is
the membrane. It also provides energy haemoglobin occurs in the first 3–6 metabolised to the yellow pigment
for the cytoskeletal changes needed for months of life. However, the subunits bilirubin which is bound to albumin in
recovery of cell shape. The Embden– designated γ and δ persist into later life the plasma. Bilirubin is conjugated in
Meyerhof pathway does not require and small amounts of fetal haemoglobin the liver to a water-soluble diglucuro-
oxygen as a substrate but a small amount (HbF; α2γ2) and HbA2 (α2δ2) are found in nide that is converted to stercobilin and
of oxidative glycolysis occurs by the adults. stercobilinogen and excreted in the
hexose monophosphate shunt (pentose Haemoglobin is more than an inert faeces. Some stercobilin and sterco-
phosphate pathway) in which glucose-6- carrier molecule. The individual globin bilinogen are reabsorbed from the intes-
phosphate is metabolised to generate chains interact with each other to facili- tine and excreted in the urine as urobilin
nicotinamide adenine dinucleotide tate the offloading of oxygen at lower and urobilinogen.
phosphate (NADPH). The hexose oxygen saturations. The metabolite
monophosphate shunt plays a vital role 2,3-diphosphoglyceride (2,3-DPG) gen-
in oxygen detoxification and when oxi- erated in a side-arm of the Embden–
dised substrates accumulate in the cell Meyerhof pathway has an important
it increases activity several fold. Inher- role in the process, which results in a
ited deficiencies of red cell enzymes in sigmoid-shaped oxygen dissociation Saturation (%)
either the Embden–Meyerhof pathway curve (Fig 2.5). In anatomical terms hae- 100
(e.g. pyruvate kinase) or the hexose moglobin has a high affinity for oxygen
monophosphate shunt (e.g. glucose-6- in the lungs and a much lower affinity
phosphate dehydrogenase) can lead to in the tissues. The oxygen dissociation
shortened red cell survival and haemo- curve moves to the left when oxygen
lytic anaemia (see p. 29). affinity increases; this occurs when H+
ion concentration is reduced or haemo- 50
globin F (which cannot bind 2,3-DPG)
raised. The curve moves to the right
Haemoglobin and
when oxygen affinity decreases; for
oxygen transport
instance when 2,3-DPG concentration
The key function of red cells, to carry rises or the abnormal sickle haemo-
0
oxygen to the tissues and return CO2 globin (HbS) is present. The P50 level 3.6 13
from the tissues to the lungs, depends is defined as the partial pressure of Oxygen tension (kPa)
on the specialised protein haemoglobin oxygen at which haemoglobin is half Fig 2.5  The oxygen dissociation curve. The
which is present in large amounts saturated. P50 is 3.6 kPa in normal red cells.

Fig 2.4  The essential elements


Globin
of the haemoglobin molecule.
Red cells
chain
In reality each globin chain has a ■ Erythropoiesis (the formation of red
a1 b1 complex helical structure. The α cells) is regulated by the growth factor
chain has 141 amino acids and the erythropoietin.
β chain 146. The haem molecule
■ Mature red cells have a biconcave disc
consists of four pyrrole rings
shape and no nucleus.
arranged around a ferrous ion.
■ The red cell membrane consists of a
Haem
molecule lattice of specialised proteins and an
b2 a2 outer lipid bilayer.
■ Red cells derive energy principally from
the metabolism of glucose to lactate
(Embden–Meyerhof pathway).
■ Red cells contain a specialised protein,
haemoglobin, which allows carriage of
oxygen to the tissues and return of CO2
from the tissues to the lungs.
6 1 ANATOMY AND PHYSIOLOGY

3 Neutrophils, eosinophils, basophils


and monocytes
The term ‘white cells’ or ‘leucocytes’ refers to the nucleated actin–myosin assembly in the cell membrane, the latter medi-
cells of the blood – the neutrophils, lymphocytes, monocytes, ating the movement necessary for locomotion and phagocy-
eosinophils and basophils. All these cells play a role in defend- tosis. Once the cell is at the target site the foreign antigen or
ing the host against infection and other insults. Neutrophils, particle is recognised via cell surface receptors and engulfed
monocytes, eosinophils and basophils are phagocytes. They within a phagocytic vacuole. There are various methods of
engulf and destroy foreign material and damaged cells. The killing; key mechanisms are the generation of nitric oxide and
term ‘granulocytes’ may be used to particularly describe neu- antimicrobial proteins, and oxidative metabolism in which
trophils, eosinophils and basophils. antimicrobial oxidants are formed (the ‘respiratory burst’).
Cytokines such as G-CSF and GM-CSF (see p. 3) not only
increase neutrophil production but also promote chemotaxis
Neutrophils
and phagocytosis.
The blood neutrophil (Fig 3.1a) is the end-product of an In clinical practice an increase in neutrophils in the blood
orchestrated sequence of differentiation in the myeloid cells (‘neutrophil leucocytosis’ or ‘neutrophilia’) is a common
of the bone marrow. The mature cell has a multi-lobed
nucleus and four different types of granules in the cytoplasm.
Neutrophils have a limited lifespan of around 5–6 days in the
blood. Approximately half the cells are included in a normal
blood count (the circulating pool), the remainder being in the
‘marginal pool’. The essential function of all these cells is to
enter the tissues and combat infection. This requires both
migration to the site of infection or tissue injury (chemotaxis)
and the destruction of foreign material (phagocytosis). Normal
chemotaxis is dependent on the release of chemotactic factors
generated by bacteria and leucocytes already present at the
infection site. Neutrophils may migrate intravascularly as they
navigate healthy tissues to reach the site of tissue injury.
Neutrophil mobility is imbued both by the presence
of adhesion molecules on the cell surface and by an
(c)

(a) (d)

(b) (e)

Fig 3.1  Leucocytes in the blood. (a) Neutrophils; (b) neutrophil with phagocytosed bacteria; (c) eosinophil; (d) basophil; (e) monocyte.
Neutrophils, eosinophils, basophils and monocytes 7

accompaniment to infection and tissue Table 3.1  Causes of a neutrophil recognised by their abundant dark
injury (Table 3.1). The strain on the neu- leucocytosis purple cytoplasmic granules (Fig 3.1d).
trophil compartment often leads to ■ Physiological (e.g. pregnancy) The granules contain mediators of acute
younger ‘band forms’ being discharged ■ Bacterial infections inflammation, including heparin and
from the marrow into the bloodstream ■ Inflammatory diseases (e.g. vasculitis, inflammatory histamine. Basophils and their tissue
bowel disease)
and the appearance of toxic changes, equivalent, mast cells, have receptors for
■ Trauma/surgery
including coarsened granulation and ■ Malignancy
the Fc portion of IgE. They play a central
vacuolation. Occasionally, phagocytosed ■ Acute haemorrhage role in immediate hypersensitivity reac-
bacteria are visible (Fig 3.1b). ■ Severe metabolic disorders (e.g. diabetic tions. Basophilia is usually associated
Reduced neutrophils in the blood ketoacidosis) with myeloproliferative disorders (e.g.
■ Myeloproliferative diseases (e.g. chronic myeloid
(neutropenia) is seen in a wide range chronic myeloid leukaemia). However, it
leukaemia)
of inherited and acquired disorders. ■ Iatrogenic (e.g. treatment with growth factors, may be reactive to a range of systemic
Serious infection is not seen regularly corticosteroids) diseases including inflammatory bowel
until the count falls below 0.5 × 109/L. disease and hypothyroidism. It some-
Neutropenia may be an isolated abnor- times occurs during the recovery phase
mality or associated with a pancytope- Table 3.2  Causes of an isolated
from acute infection.
nia. Some causes of an isolated neutropenia1
neutropenia are listed in Table 3.2. In ■ Drugs2 Monocytes
general, neutropenia may be caused by ■ Idiopathic/benign/constitutional
underproduction from the marrow (e.g. ■ Congenital (Kostmann’s syndrome) Monocytes circulate in the blood
leukaemia), reduced neutrophil lifespan ■ Cyclical neutropenia before entering the tissues where they
(e.g. immune neutropenia), or pooling
■ Autoimmune (sometimes with a connective tissue undergo transformation into macro-
disorder)
of neutrophils in a large spleen. It is phages. Monocyte colony-stimulating
■ Infections (e.g. viral, typhoid, tuberculosis)
important to remember that drugs may factor (M-CSF) is vital for monocyte
be responsible. The term chronic benign
1
Most bone marrow diseases (e.g. leukaemia, aplastic anaemia) and macrophage development and acti-
cause a pancytopenia.
neutropenia is generally used in patients 2
Some drugs are well-documented causes (e.g. penicillin,
vation. The ‘mononuclear phagocyte’
who have an isolated moderate neutro- co-trimoxazole, carbimazole, phenothiazines) but in practice any system consisting of monocytes and
penia with no clear aetiology and a
agent the patient is taking should be viewed with suspicion. macrophages is a potentially confusing
benign course. There may be an associ- concept as macrophages subserve differ-
ated monocytosis. There is some ethnic The most common causes of eosi- ent functions and adopt discrete nomen-
variation in neutrophil counts with nophilia in the Western world are allergic clature in different tissues (e.g. osteoclasts
black people having a lower normal ref- disorders such as asthma, eczema and hay in bone, Kuppfer cells in liver). Macro-
erence range than white people. In the fever. In developing countries, parasitic phages are phagocytic cells but unlike
rare genetic disorder cyclical neutrope- infections are frequently implicated. neutrophils are able to survive the
nia, the neutrophil count falls every Other relatively common aetiologies are phagocytic event. They also act as acces-
15–35 days and recurrent infections drug hypersensitivity, malignancy, various sory cells in the immune response by
occur. skin diseases and connective tissue disor- presenting antigens to T-lymphocytes
In addition to quantitative abnormali- ders. Hypereosinophilic syndrome is (see p. 8) and secreting a wide range of
ties, neutrophils can be functionally characterised by a marked sustained eosi- cytokines involved in inflammation,
abnormal. There are several rare inher- nophilia and associated tissue damage. immunity and haematopoiesis.
ited diseases characterised by impaired The disorder is very variable with several Blood monocytes typically have a
neutrophil adherence, chemotaxis or subtypes. The myeloproliferative variant kidney-shaped nucleus (Fig 3.1e). A
bactericidal activity. In chronic granulo- is associated with a FIP1L1-PDGFRA monocytosis in the blood occurs in
matous disease, neutrophils are able to fusion gene and often responds to imat- chronic bacterial infections such as
phagocytose but not kill catalase-positive inib (see p. 45). tuberculosis and may accompany a wide
microorganisms. Inheritance is auto- range of infective, inflammatory and
malignant disorders. Monocytopenia is
somal or X-linked and patients suffer Basophils less frequently noted but can be severe
recurrent purulent infections and asso-
ciated granuloma formation. Diagnosis Basophils are the least numerous of in patients receiving corticosteroid
is made in the nitroblue tetrazolium test the blood leucocytes. They are easily treatment.
where the patient’s neutrophils fail to
reduce the dye.

Neutrophils, eosinophils,
Eosinophils basophils and monocytes
Eosinophils (Fig 3.1c) are characterised ■ The white cells of the blood (leucocytes) play a key role in defending the host against
by their two-lobed nucleus and red- infection and other insults.
orange staining granules. They have sig- ■ Neutrophils, monocytes, eosinophils and basophils are phagocytes.
nificant proinflammatory and cytotoxic ■ These phagocytic cells may perform other functions; monocytes act as accessory cells
activity and play a role in the pathogen- presenting antigens to T-lymphocytes.
esis of various allergic, parasitic and neo- ■ Each cell has a characteristic morphological appearance in the blood film.
plastic disorders. Interleukin 5 is a key
■ Changes in leucocyte numbers (e.g. neutrophil leucocytosis) are common accompaniments
mediator of eosinophil differentiation
of various disease states.
and activation.
8 1 ANATOMY AND PHYSIOLOGY

4 Lymphocytes
Lymphocytes are found in large numbers in blood, lymph
(the clear fluid of the lymphatic vessels) and in lymphoid
organs such as the thymus, lymph nodes and spleen. They
are essential for immunity. B-lymphocytes produce antibody
against a specific antigen (humoral immunity) while
T-lymphocytes are the cells of the cell-mediated response.
Primary lymphoid organs (bone marrow, thymus) are the
sites of lymphoid development. In the secondary lymphoid
organs (lymph nodes, spleen), mature lymphocytes meet anti-
gens and the immune response is triggered.
Most mature lymphocytes appear under the light micro-
scope as cells with round nuclei and a thin rim of agranular
cytoplasm (Fig 4.1). Although B- and T-cells are not distin-
guishable by their morphology, there are major differences in
their mode of maturation and function. Fig 4.1  Mature lymphocytes in the blood.

Antigen (Ag) B-cell Plasma cell


T-lymphocytes Differentiation
T-cells make up 75% of the lymphocytes of the blood and Ag processed into
peptides and combined
form the basis of cell-mediated immunity. They are less auton- with MHC molecules
Growth
omous than their B-cell companions, needing the cooperation factors
of antigen-presenting cells expressing self-histocompatibility Class II MHC TCR
CD4+
molecules (human leucocyte antigens (HLA)) for the recogni- T helper cell
tion of the antigen by the T-cell receptor (TCR) (Fig 4.2).
Recognition
T-cells originate in the marrow but many are destroyed in Ag presenting cell
subsequent processing by the thymus, the objective being to
select the minority of cells which will recognise self-HLA but Class I MHC
not react with self-tissue antigens. The maturation sequence CD8+
is characterised by changing patterns of cell surface molecules Cytotoxic T-cell
(Fig 4.3). Mature T-cells are divisible into two basic types. Cell lysis
Ag infected cell
About two-thirds of blood T-cells are ‘helper’ cells expressing
the surface marker CD4, while the remainder express CD8 Fig 4.2  Interaction of T-lymphocyte and antigen-presenting cells.
The T-cell receptor complex (TCR) recognises the combination of processed
and are mostly of ‘cytotoxic’ type.
antigen and major histocompatibility complex (MHC) molecule and the
It appears that helper cells recognise the combination of immune response is initiated.
antigen and self-HLA class II molecules on the antigen-
presenting cell, and cytotoxic cells bind with antigen in con-
B-lymphocytes
junction with HLA class I molecules on the target cell (Fig
4.2). TCR genes, like Ig genes, are subject to rearrangement B-lymphocytes are responsible for humoral immunity. Fol-
of germ-line DNA. Following triggering of T-cells by specific lowing an appropriate antigenic stimulus they transform into
antigen reacting with the TCR, the clonal proliferation of plasma cells and secrete antibody specific to that antigen.
activated T-cells is sustained by the secretion of cytokines. B-cells are derived from the stem cells of the bone marrow.
Interleukin-2 is the main T-cell growth factor. Unlike T-cells it is not clear whether they are subject to further

Stem PC Plasma Large Small


Pre-B Early Mature Activated Stem cortical cortical Medullary Virgin
cell cell cell Pre-T cell
B-cell cell thymocyte thymocyte thymocyte T-cell

Antigen independent Antigen dependent Bone Thymus Blood,


marrow lymph node,
spleen
First Ig rearrangement (VDJ) Second Ig rearrangement (isotope switch) Rearrangement of TCR genes

Expression of selected CD markers Expression of selected CD markers


Tdt Tdt
cyt m CD7
Surface Ig Cyt CD3
CD10 CD2
CD19 CD1
CD20 CD4+8 / CD4 / CD8

PC : plasmacytoid Ig : immunoglobulin TCR : T-cell receptor


(a) (b)
Fig 4.3  Maturation of B- and T-lymphocytes. (a) B-lymphocyte maturation in the bone marrow. (b) T-lymphocyte development in the bone marrow
and thymus.
Lymphocytes 9

processing at a site outside the marrow IgA is found in secretions, while IgE In the blood film, NK cells appear as
in humans. The various stages of B-cell plays a role in delayed hypersensitivity large lymphocytes with abundant cyto-
maturation are illustrated in Figure 4.3. reactions. plasmic granules.
Each cell can be defined by its expres- The genes encoding the heavy and
sion of membrane and cytoplasmic light chains of immunoglobulin are rear-
Changes in disease
antigens in addition to the stage of ranged from their germ-line configura-
immunoglobulin gene rearrangement. tion during early B-cell maturation. The An increase in lymphocytes in the blood
Within the lymphoid tissues, such as variable (V), diverse (D), joining (J) and (lymphocytosis) is generally a reaction
the lymph nodes and spleen, mature constant (C) region exons undergo a to infection or is part of a malignancy.
unactivated or virgin B-cells can be complex sequence of DNA splicing, dele- A polyclonal T-cell lymphocytosis is a
stimulated by antigen to undergo a mor- tions and juxtapositions. The rationale of common response to viral infection,
phological transformation into immu- this frenetic activity prior to transcrip- particularly in childhood. Lymphocytes
noblasts and, ultimately, plasma cells. tion is to allow the totality of B-cells to may be morphologically abnormal with
Stimulation of a single B-cell by produce an enormously diverse popula- variable changes including increased
antigen combining with its cell surface tion of immunoglobulins (antibodies) size and cytoplasmic basophilia. These
immunoglobulin variable region leads targeting a vast number of potential anti- heterogeneous atypical lymphocytes are
to a sequence of proliferation and gens. Lymphocytes that can react against seen in numerous viral infections but
differentiation resulting in a clone of self-molecules are usually functionally they are a particular feature of infectious
immunoglobulin-secreting plasma cells. inactivated or deleted so that the adap- mononucleosis (see p. 97).
This adaptive immune response is tive immune system normally only A number of lymphoid malignancies
antigen-specific and is facilitated by targets foreign antigens (natural immu- are associated with lymphocytosis
helper T-cells and cytokine-secreting nological tolerance). (Table 4.1). In acute lymphoblastic leu-
macrophages. Memories of particular kaemia and ‘spill-over’ of non-Hodgkin’s
antigens are immortalised by ‘memory’ lymphoma cells into the blood, the
Natural killer (NK) cells
B-cells, allowing a prompt response malignant lymphocytes are usually mor-
to reinfection. The immunoglobulins NK cells are a subset of lymphocytes phologically distinctive and confusion
secreted by lymphocytes and plasma which share many of the characteristics with a reactive lymphocytosis rarely
cells are heterogeneous proteins, each of cytotoxic T-cells. However, NK cells occurs. In chronic lymphocytic leukae-
designed to interact with a specific do not rearrange or express TCR genes. mia (CLL), the lymphocytes often appear
antigen in the defence of the body They particularly kill target cells that unremarkable although the presence of
against infection (Fig 4.4). There are five poorly express class I MHC and are less disrupted forms, termed ‘smear cells’, is
subclasses of immunoglobulin (Ig), able to signal viral infection to cytotoxic characteristic.
dependent on the type of heavy chain T-cells. NK cells express two classes of Lymphocyte counts are often tran-
(IgG, IgA, IgM, IgD and IgE), with some receptors which either activate or inhibit siently low after surgery and trauma. A
further division of subclasses (e.g. their killing role. Activating receptors more chronic lymphopenia is a feature
IgG1–4). IgM is generally produced as the bind to a variety of ligands on the target of ongoing cytotoxic drug treatment and
initial response to infection, followed by cell whereas the inhibitory receptors late HIV infection when CD4 counts fall
a more prolonged production of IgG. generally bind to HLA class I molecules. to low levels.

Heavy Table 4.1  Common causes of a lymphocytosis


chains
Infections Acute viral infections (e.g. pertussis, infectious mononucleosis,
VH VH rubella)
Chronic infections (e.g. tuberculosis, toxoplasmosis)
Fab CH CH
VL VL Malignancy Chronic lymphocytic leukaemia and variants
Light Non-Hodgkin’s lymphoma (minority)
CL CL chain Acute lymphoblastic leukaemia

Hinge
region
CH CH
Fc Heavy
chain Lymphocytes
CH CH ■ Lymphocytes are essential for normal immunity.
■ B-lymphocytes respond to an appropriate antigen by transforming
into plasma cells and secreting specific antibody (humoral
immunity).
V = variable (antibody binding) region
C = constant region ■ T-lymphocytes cooperate with antigen-presenting cells in the
recognition of antigen; recognition triggers a clonal proliferation of
Fig 4.4  Basic immunoglobulin structure. The ‘Fab’ portion is involved activated T-cells (cell-mediated immunity).
in antigen binding and the ‘Fc’ portion attaches to macrophages or
lymphocytes expressing the relevant ‘Fc’ receptor. ■ The genes encoding immunoglobulin chains and the T-cell
receptor are subject to rearrangement of germ-line DNA.
■ Various
disease states lead to an increase in blood lymphocyte
numbers (lymphocytosis): in those over 50 years, chronic
lymphocytic leukaemia is a common cause.
10 1 ANATOMY AND PHYSIOLOGY

5 The spleen
Although the spleen has been known Marginal
of since ancient times, its function zone
Capsule
has remained obscure until relatively Lymphatic
recently. Hippocrates thought it was the nodule
source of ‘black bile’. Galen suggested it Splenic cords
Periarterial in red pulp
might be a filter, in view of its spongy lymphatic
consistency. Our current understanding sheath Direct connection
between artery
of the spleen is dependent on a detailed and sinus
appreciation of its vascular supply and
Central
the organisation of its main component artery
parts: the lymphoid white pulp, the
Lymphatic Trabecular
blood-containing red pulp and the inter- vein
nodule
vening marginal zone.
Splenic
sinuses in
red pulp
Structure
The spleen is derived from condensa-
Fig 5.1  Structure of the spleen. The white pulp is composed of the periarteriolar lymphatic sheath
and lymphatic nodules. The red pulp contains the splenic cords and sinuses and is separated from the
tion of the mesoderm in the dorsal white pulp by the marginal zone. See text for full discussion.
mesogastrium of the embryo. It plays a
modest haematopoietic role in the
middle part of fetal life, but in the adult carbohydrate antigens. The red pulp is response to these organisms. Phago-
haematopoiesis is usually only seen in composed of two alternating structures: cytic cells in the spleen also remove red
pathological states. An average adult the splenic sinuses and the splenic cords cells coated with IgG antibody.
spleen weighs about 150 g and it has to (the ‘cords of Billroth’). The cords are a The second mechanism at work is the
become enlarged to at least three times reticular meshwork packed with macro- removal of red cells which are not suf-
its normal size before becoming palpa- phages and antibody-secreting plasma ficiently deformable to pass through
ble on clinical examination (p. 17). cells. The sinuses are broad channels the sinus wall. Pathological states where
The splenic artery penetrates the thick lined with fusiform endothelial cells. red cells lose deformability and are
capsule which invests the organ (Fig Most of the central arterioles open into destroyed prematurely in the spleen
5.1). Branches of the splenic artery are the marginal zone. As alluded to already, include sickle cell anaemia, hereditary
surrounded by a highly organised aggre- circulating T-lymphocytes move into spherocytosis and malaria.
gate of lymphoid tissue which is termed the periarteriolar lymphatic sheath and Finally, the spleen can remove debris
the ‘white pulp’ (Fig 5.2). Intimate to the B-lymphocytes migrate to the follicles. or organisms from within cells. Howell–
central arteriole is the ‘periarteriolar Other blood cells move slowly through Jolly bodies (fragments of nucleus) and
lymphatic sheath’ – an area mainly the complex meshwork of the red pulp, malarial parasites are removed when
populated by T-lymphocytes. Among and cells which are sufficiently deforma- most of the cell passes through the
these T-lymphocytes are non-phagocytic, ble and compliant squeeze between the inter-endothelial slit with the intracel-
antigen-presenting cells known as ‘inter- endothelial cells in the sinus wall into lular particle abandoned on the cord
digitating cells’. Spaced at intervals in the lumen of the sinus and back into side.
the periarteriolar lymphatic sheath are the circulation. The organisation of the The spleen has the capacity to mount
lymphoid follicles (‘Malpighian bodies’). spleen into the different compartments is complex innate and adaptive immune
In an inactive state these follicles are under the control of various cytokines responses. Both types of response occur
composed of recirculating B-lymphocytes and adhesion molecules. in the marginal zone, rich in macro-
intertwined with cytoplasmic processes phages and marginal zone B-cells, while
of follicular dendritic cells. The latter the white pulp is limited to adaptive
Function
cells may play a role in long-term anti- immunity.
body production. When contact with The spleen has two key functions. It
antigen stimulates B-cell activation, a removes older red cells, blood-borne
Abnormal splenic states
germinal centre of rapidly dividing cells microorganisms and cellular debris
forms in the follicle. This is a key area from the blood. It also plays a vital role Asplenism and hyposplenism
in the normal B-lymphocyte prolifera- in the body’s response to bacterial and Surgical removal of the spleen (splenec-
tive response and development of B-cell fungal infections. tomy) may be indicated in a variety of
memory (see p. 8 for discussion of It clears unwanted red cells and parti- haematological disorders and following
lymphocytes). cles from the blood in three ways. trauma. The spleen may also be absent
The periarteriolar lymphatic sheath Firstly, they can be removed by phago- as a congenital anomaly, often associ-
and B-lymphocyte follicles are separated cytes. Bacteria, particularly encapsulated ated with transpositions or malforma-
from the red pulp by a ‘marginal zone’ organisms that are not opsonised by tions of the great vessels and viscera
constituted mainly of non-circulating antibodies and complement, are cleared (‘asplenia syndrome’). Reduced splenic
B-cells. The marginal zone also contains from the circulation. The spleen is prob- function can result from splenic atrophy
specialised macrophages able to take up ably the site of the initial immune in disorders such as sickle cell anaemia,
The spleen 11

Fig 5.2  Light microscopy of the spleen clearly showing the Fig 5.3  The blood film in hyposplenism. A Howell–Jolly body is seen
distribution of red and white pulp. within a red cell. There are target cells and acanthocytes.

adult coeliac disease and essential Table 5.1  Causes of hyposplenism Table 5.2  Management recommendations
thrombocythaemia (Table 5.1). ■ Congenital absence of spleen in the asplenic patient
Hyposplenism leads to characteristic ■ Splenectomy Immunisation1 Pneumococcus, Haemophilus
changes in the blood film (Fig 5.3). ■ Sickle cell anaemia influenzae type B, group C
Changes in red cell appearance include ■ Coeliac disease meningococcus, influenza
■ Essential thrombocythaemia Antibiotic Oral phenoxymethylpenicillin
the presence of Howell–Jolly bodies,
■ Dermatitis herpetiformis prophylaxis2 or erythromycin
Pappenheimer (siderotic) granules and ■ Inflammatory bowel disease
Prompt treatment Patients need systemic
target cells. Other less regular red cell ■ Amyloidosis
of infection antibiotics and urgent
features are lipid-rich acanthocytes and ■ Advanced age
admission to hospital
circulating nucleated cells. There is Medicalert disc or Detailing asplenic state and
often a moderate rise in the lymphocyte, card medical contacts
monocyte and platelet count. Approxi- Avoid travel to
mately one-third of circulating platelets Hypersplenism high-risk malarial

are pooled in the normal spleen. The Hypersplenism is usually defined as a areas

increase in platelets post-splenectomy is depression of one or more of the cell 1


Where possible at least 2 weeks prior to splenectomy.
frequently impressive (greater than counts in the blood which can be wholly Reimmunisation is usually required, the timing determined by
measurement of specific antibody levels.
1000 × 109/L) but the count usually falls attributed to splenic enlargement. Other 2
The duration of antibiotic prophylaxis is controversial but should
to a lower level in the longer term. criteria such as the presence of a normal generally be ‘lifelong’.

Quantitation of splenic function is not bone marrow, or correction of cytopenia


straightforward. Methods include the by splenectomy may be appended.
measurement of the percentage of Although the definition only requires an
isolated anaemia, leucopenia or throm- ■ Hypervolaemia consequent upon a
pitted erythrocytes using interference
phase microscopy, various immunologi- bocytopenia, there is frequently a mod- disproportionately expanded plasma
cal parameters and scintigraphy. erate pancytopenia. volume filling the vascular space of
The clinical significance of an absent Splenomegaly is not always associ- the enlarged spleen and the
spleen is the associated increased risk of ated with hypersplenism, and hyper- splanchnic bed.
splenism can occur irrespective of the ■ Intrasplenic pooling of red cells which
life-threatening infection. The risk is
greatest in children under 5 years of age degree of splenic enlargement. Thus, it is increased from the normal 5–15%
and where there is a serious underlying may be seen in the modest splenomeg- to 40% in moderate splenomegaly.
medical disorder such as Hodgkin’s lym- aly of liver cirrhosis. This is accompanied by pooling of
phoma or thalassaemia. Most infections The pancytopenia of hypersplenism is neutrophils and platelets.
probably induced by three contributory ■ Premature destruction of circulating
occur within 2 years of splenectomy but
fulminating infection can strike at any mechanisms: blood cells.
stage. In most cases infection is with
encapsulated bacteria, notably Strepto-
coccus pneumoniae, Haemophilus influen-
The spleen
zae and Neisseria meningitidis. In
temperate regions more than half of ■ The spleen is organised into three main components: the white pulp, the red pulp and the
serious infections are caused by the intervening marginal zone.
pneumococcus, with high mortality. ■ The spleen acts as a filter, removing unwanted red cells and particles from the blood.
Splenectomised patients have an ■ The spleen can mount complex adaptive immune responses.
increased susceptibility to severe malaria.
■ An absent or poorly functioning spleen leads to characteristic blood changes and an
Prophylaxis against such infections is increased risk of overwhelming infection, including fulminating malaria.
the best approach and recommenda-
■ An enlarged spleen (splenomegaly) may cause ‘hypersplenism’ with reduced cell counts in
tions for the management of asplenic the blood.
patients are shown in Table 5.2.
12 1 ANATOMY AND PHYSIOLOGY

6 Haemostasis
Blood clotting is a critical defence mech- 10 days. They have no nucleus and no refers to the mechanism directly leading
anism which, in conjunction with capacity for DNA biosynthesis but do to the conversion of the soluble plasma
inflammatory and general repair have a complex infrastructure. Pores in protein fibrinogen to the insoluble rigid
responses, helps protect the integrity of the trilaminar platelet membrane polymer fibrin. The formation of the
the vascular system after injury. The connect with an open canalicular system stable haemostatic plug composed of
complex sequence of events described in allowing transport of agonists in and enmeshed fibrin and platelets is the cul-
detail below is activated within seconds discharge of secretions out. The mem- mination of a complex biochemical
of tissue damage. It is easiest to divide brane receptors for agonists include: cascade involving circulating coagulation
the description of normal haemostasis factors. This system allows extreme
■ the glycoprotein (GP) Ia/IIa complex
into a platelet component, with forma- amplification with a robust thrombus
(α2β1 integrin) and glycoprotein (GP)
tion of a loose platelet plug at the site of arising from the initial stimulus of tissue
VI which are receptors for collagen
injury, and a coagulation component, injury. Most activated coagulation factors
■ the GPIb/IX/V complex, a receptor
where there is generation of a more are proteolytic enzymes (serine pro-
for vessel wall von Willebrand factor
robust fibrin scaffold (thrombus) teases) which in the presence of cofac-
(vWF) and thrombin
around the platelets. This approach tors cleave other factors in an ordered
■ the GPIIb/IIIa complex (αIIbβ3
facilitates understanding but in practice sequence. Thus, prothrombin (factor II),
integrin), which is an agonist-induced
the two mechanisms are inextricably factor VII, factor IX and factor X are
receptor for fibrinogen and vWF
linked. proenzymes which are converted to
(vWF is discussed in more detail on
their active enzyme form (denoted by
p. 74).
the letter ‘a’) by cleavage of one or two
The role of platelets In the platelet cytoplasm are organelles peptide bonds. Factors V and VIII are
Following damage to a blood vessel including alpha granules (containing procofactors which are converted to the
there is immediate vasoconstriction to fibrinogen, vWF, thrombospondin and active cofactors (Va and VIIIa) also by
slow blood flow and reduce the risk of other proteins) and dense granules cleavage of peptide bonds. The blood
exsanguination. The break in the (containing small molecules such as clotting proenzymes prothrombin and
endothelial cell barrier leads to the ADP and calcium). factors VII, IX and X require vitamin K
recruitment of platelets from the circula- Platelet activation follows stimulation for their activation (see pp. 76, 77).
tion to form an occlusive plug. Platelets by agonists such as ADP and thrombox- The coagulation cascade, leading to
interact both with the vessel suben- ane A2 interacting with surface recep- the generation of thrombin and the for-
dothelial matrix (platelet ‘adhesion’) and tors, or by direct contact with the vessel mation of a fibrin thrombus, is classi-
with each other (platelet ‘aggregation’) wall subendothelial matrix. Platelets cally divided into two parts: the intrinsic
(Fig 6.1). The first step in this process, convert from a compact disc to a sphere, and extrinsic pathways (Table 6.1).
adhesion, does not require platelet met- surface receptors become activated, and In the intrinsic pathway factor XII is
abolic activity. It does, however, lead to cytoplasmic granules secrete their con- activated by exposed collagen and other
the ‘activation’ of platelets. tents. The net effect is the mediation and negatively charged components of the
Platelets are small disc-shaped parti- reinforcement of aggregation and adhe- subendothelium. Activation of factor XII
cles produced in megakaryocyte cyto- sion, and the promotion of further acti- leads to the sequential activation of
plasm which have a lifespan of around vation. Other circulating platelets adhere factors XI, IX, VIII (as cofactor), X and
to the initial layer and a loose platelet prothrombin. In the extrinsic pathway
Translocation Tethering of plug is formed. tissue factor complexes with factor VII
αIIbβ3 platelets In addition to the formation of a phys- with sequential activation of factors VII,
α granule ical barrier at the site of injury, platelets X and prothrombin. Both intrinsic and
α2β1
GPIb/IX/V Dense granule have a procoagulant action. The coagu- extrinsic pathways terminate in the final
Collagen
lation sequence described below com- common pathway where activated factor
pletes much more rapidly in the X, in association with the cofactor factor
vWF Primary adhesion
presence of platelets. Following activa- Va in the presence of phospholipid and
and activation
tion, platelets rearrange their membrane calcium, converts prothrombin into
phospholipids and shed vesicles from thrombin. Thrombin in turn converts
αIIbβ3 GPVI
their surface. The platelet surface and fibrinogen to fibrin by splitting the fibri-
α2β1
vesicles reveal binding sites for coagula- nopeptides A and B from the centre
tion proteins leading to the creation of domain to form fibrin monomers. These
Fibrinogen Aggregation
coagulation complexes (e.g. the ‘pro- monomers combine spontaneously into
thrombinase complex’) which accelerate dimers which assemble to form the
formation of factor Xa and thrombin. fibrin polymer. Factor XIII crosslinks
the fibrin polymer to consolidate the
ADP thrombus. The conventional division
Thromboxane Coagulation
A2 into two pathways is useful in the inter-
Although often loosely used to encom- pretation of in vitro laboratory tests of
Fig 6.1  Primary platelet adhesion, activation pass all aspects of clot formation, the haemostasis. The prothrombin time
and aggregation. vWF, von Willebrand factor. term ‘coagulation’ more specifically (PT) is a simple measure of the function
Haemostasis 13

of the extrinsic pathway and the activated partial thrombo- and thrombin. Its activity is greatly increased by interaction
plastin time (APTT) monitors the intrinsic pathway (p. 20). with heparin in the microvasculature and on the surface of
However, the physiological pathways at work in vivo are not endothelial cells.
so simply defined (see Fig 6.2). It seems that the intrinsic ■ Proteins C and S. Protein C is a vitamin K-dependent

pathway is rarely relevant to coagulation in vivo – patients plasma protein which inactivates the cofactors Va and
with hereditary deficiency of factor XII have a prolonged VIIIa and stimulates fibrinolysis. Protein C is converted to
APTT but no bleeding disorder. The crucial protein in the its active enzymic form by interaction with thrombin.
initiation of blood coagulation is tissue factor, an integral Protein S acts as a cofactor for protein C.
membrane protein expressed on non-vascular cells. When a ■ Tissue factor pathway inhibitor (TFPI). TFPI

blood vessel is damaged, circulating factor VII comes into inactivates factor Xa and then the TFPI/factor Xa complex
contact with tissue factor. The tissue factor/factor VIIa complex inhibits factor VIIa within the VIIa/tissue factor complex.
activates not only factor X (the extrinsic pathway) but also
factor IX.
Fibrinolysis
Regulation of coagulation Once damaged endothelium is repaired the fibrin thrombus
Blood coagulation is modulated by three major inhibitory must be removed to restore normal blood flow. Thrombus
systems: removal is facilitated by a fibrin-splitting serine protease,
plasmin. The fibrinolytic system is shown schematically in
■ Anti-thrombin. This is the most important inhibitor of
Figure 6.3. Release of tissue plasminogen activator (t-PA) from
the terminal proteins of the cascade, particularly factor Xa
endothelial cells leads to conversion of the proenzyme plas-
minogen into plasmin. t-PA is most active when bound to
fibrin, thus maximising its action at the site of the thrombus.
Plasmin has the capacity to digest fibrin in addition to fibrino-
Table 6.1  The classic coagulation cascade gen and a number of other proteins. Digestion of a cross-
Intrinsic pathway linked thrombus by plasmin leads to the formation of
Factor XIIa + Kallikrein → XIa → IXa → Xa Final common pathway
‘degradation products’ which themselves act as anticoagu-
Extrinsic pathway Factor Xa → Thrombin → Fibrin
lants. Fibrinolysis is under strict control; circulating plasmin
Factor VIIa – Tissue factor → Xa
is inactivated by the protease inhibitor α2-antiplasmin.

Plasminogen activators
Plasma
Tissue plasminogen activator (t-PA)
Monocyte Urokinase
Factor XIIa
Kallikrein
TF
Streptokinase Fibrin(ogen)

FVIIa FVII
T

Plasminogen Plasmin
FXI FXIa FVIIa TF

Fibrin(ogen) degradation
FIX FIXa products
T Fig 6.3  The fibrinolytic system. Note that, unlike the other activators
FVIII FVIIIa FIXa FVIIIa listed, streptokinase is an exogenous activator derived from β-haemolytic
streptococci.

FX FXa

T
FV FVa FXa FVa
Haemostasis
PT T ■ The clotting of blood is a critical defence mechanism protecting
the integrity of the vascular system after injury.
■ Platelets
form an occlusive plug at the site of tissue injury. They also
Fibrinogen Fibrin
have procoagulant action.
■ The term ‘coagulation’ describes the process by which fibrinogen is
Clot formation converted to the insoluble rigid polymer fibrin; the final thrombus
is formed of enmeshed fibrin and platelets.
Fig 6.2  Physiological pathways of blood coagulation. Green arrows
indicate the action of enzymes on substrates; red arrows indicate the ■ The term ‘coagulation cascade’ describes the sequential activation
conversion of a protein from one functional state to another after the of coagulation factors; in vivo the major initiator of coagulation is
cleavage of one or more peptide bonds. F, factor; TF, tissue factor; T, tissue factor.
thrombin; PT, prothrombin. Reprinted with permission from Furie B, Furie BC ■ Fibringeneration is regulated by naturally occurring anticoagulants
2004 Role of platelet P-selectin and microparticle PSGL-1 in thrombus and fibrin is ultimately removed by the ‘fibrinolytic system’.
formation. Trends in Molecular Medicine 10(4):171–178.
14 2 THE HAEMATOLOGY PATIENT

7 History taking
Abnormalities of the blood are associated with a wide range  
of symptoms and these are discussed in detail under diagnos-
tic headings in subsequent sections. The intention of this
section is to give an overview of history taking in patients with
blood disorders. Despite the advent of sophisticated labora-
tory equipment to test blood, a thorough history remains
fundamental to accurate diagnosis. In practice the history may
precede and then follow the knowledge of a laboratory test
abnormality. Whatever the order of events, only by consider-
ing symptoms, physical signs and laboratory results in con-
junction can the correct conclusion be reached and the patient
be managed in the appropriate psychosocial setting (Fig 7.1).

History of the presenting complaint


Patients may be asymptomatic and have an unpredictable
abnormality detected on a routine blood count. Other patients
present to the doctor with complaints dependent on the
nature of the change in the blood. Some will have several
blood abnormalities and present with a large number of
symptoms. Despite this complexity it is possible to highlight

some common groups of symptoms (Table 7.1).   
Fig 7.1  The history, clinical examination and laboratory investigations
Symptoms attributable to anaemia (low are all essential in the diagnosis of a disorder of the blood.
haemoglobin concentration)
Patients with anaemia have a reduced supply of oxygen to the
tissues. Symptoms include fatigue, weakness, dyspnoea, pal- Table 7.1  Common haematological abnormalities and associated
pitations, headaches, tinnitus and chest pain (due to exacerba- symptoms
Nature of abnormality1 Commonly associated symptoms
tion of angina). The symptoms are affected not only by the
Anaemia Fatigue, weakness, dyspnoea, palpitations,
severity of the anaemia but by its speed of onset. Anaemia
headache, dizziness, tinnitus
which develops rapidly is usually less well tolerated and
Leucopenia (particularly neutropenia) Unusually severe or recurrent infections
patients are more debilitated.
Thrombocytopenia Easy bruising, excessive bleeding after
trauma, spontaneous bleeding from
Symptoms attributable to a low white cell mucous membranes
count (leucopenia) Defective coagulation (e.g. key factor Excessive bleeding after trauma,
It is usually a reduction in neutrophils (neutropenia) which deficiency) spontaneous bleeds into joints and
muscles
causes clinical problems. Patients are susceptible to infections,
Infiltration by malignancy (e.g. ‘Lumps’ caused by lymphadenopathy,
the risk rising sharply at neutrophil counts below 0.5 × 109/L.
leukaemia, lymphoma) pain, neurological symptoms
Serious blood diseases such as acute leukaemia can present
as life-threatening infections or as apparently trivial infections 1
The haematological abnormalities have many possible causes but will always tend to lead to the
symptoms shown.
(e.g. a sore throat) which are unusually refractory to normal
treatment. Perineal sepsis can be a particular problem.
and muscles. Lifelong symptoms suggest an inherited abnor-
Symptoms attributable to a low platelet mality while recent onset is consistent with an acquired
count (thrombocytopenia) aetiology.
Thrombocytopenia leads to a haemorrhagic tendency and
common presentations include epistaxes (nose bleeds), bleed- Symptoms attributable to infiltration
ing from gums, menorrhagia (heavy periods) and excessive by malignancy
bleeding after trauma or surgery. Patients may also complain Malignant disorders of the blood such as leukaemias and
of easy bruising or a petechial rash. Spontaneous bleeding is lymphomas have the capacity to invade tissues. Patients may
usually restricted to platelet counts below 20 × 109/L. In disor- complain of lumps in the neck, axillae or groin caused by
ders of platelet function similar symptoms may occur even lymphadenopathy or of abdominal pain or distension caused
when the count is normal. by splenomegaly. Involvement of the nervous system may
manifest as headache, pain in dermatomal distribution or loss
Symptoms attributable to abnormal coagulation of function.
Patients with a defect in the coagulation cascade (e.g. low The severity, quality and temporal characteristics of pain
factor VIII level in haemophilia A) bleed easily after surgery may or may not be helpful in identifying an underlying blood
and trauma but the pattern of spontaneous haemorrhage is disorder. The pain of the vaso-occlusive crisis of sickle cell
normally different to that seen in platelet disorders. The com- anaemia is often distinctive whereas the chronic low back pain
monest complaints are of bleeding into joints (haemarthroses) of myeloma is all too easily dismissed.
History taking 15

Systemic enquiry Table 7.2  Possible haematological side-effects of drugs


Haematological abnormality Drugs1
A thorough systemic enquiry is essential as blood abnormali- Marrow aplasia Chloramphenicol (idiosyncratic)
ties are more often caused by a general systemic disorder than Cytotoxics (dose-related)
by a specific blood disease. It can be difficult to establish Haemolytic anaemia Cephalosporins
whether the primary problem is in the bone marrow or if the Penicillins
blood is ‘reacting’ to pathology elsewhere. One example is a Leucopenia/agranulocytosis Phenothiazines
Sulphonamides
high platelet count (thrombocytosis). This may be caused by
Thrombocytopenia Quinine
the bone marrow disorder essential thrombocythaemia but
Thiazide diuretics
equally can be secondary to infection, inflammation or malig-
nancy (‘reactive thrombocytosis’). Only by excluding a non- 1
Many drugs have been implicated in all these abnormalities – the examples shown are some of the
more common offenders.
haematological aetiology can the diagnosis of essential
thrombocythaemia be confidently made. On occasion the hae-
Table 7.3  Some inherited blood disorders
matological diagnosis prompts a return to a particular part of
Red cell disorders
the systemic enquiry. Thus the finding of unexplained iron
Disorders of the membrane Hereditary spherocytosis and elliptocytosis
deficiency necessitates an exhaustive enquiry for symptoms
Disorders of haemoglobin Thalassaemias and sickle syndromes
of gastrointestinal disease associated with chronic blood loss.
Disorders of metabolism Glucose-6-phosphate dehydrogenase and pyruvate
kinase deficiencies
Past medical history Coagulation disorders
Factor deficiency Haemophilia A and B
It is important to elicit a history of diseases which may have Combined factor and platelet von Willebrand disease
caused a haematological abnormality or which may affect the abnormality
management of a primary blood disorder such as leukaemia. Platelet abnormality Bernard–Soulier syndrome (rare)
Where there is a known abnormality in the blood count it is White cell disorders Rare functional disorders (e.g. chronic granulomatous
helpful to establish whether previous counts have been per- disease)
formed. Where past results are available they will clarify The mode of inheritance of these disorders is discussed in the relevant sections.
whether the problem is of recent onset or longstanding. For
patients presenting with easy bruising or bleeding, previous and nurses. Often such diseases are incurable and expert man-
surgical exposure is of particular interest. The lack of exces- agement of symptoms has to be complemented by an under-
sive bleeding after surgery suggests that the bleeding tendency standing of the patient’s need to sort out affairs and
is either of limited significance or of more recent onset. communicate the news to family and friends. In working
adults the onset of diseases like leukaemia, with frequent clinic
visits and hospitalisation, can lead to unemployment and
Drug history
marital and financial difficulties. In children chronic blood
Drugs can cause haematological problems – some commoner disorders such as haemophilia and haemoglobinopathies may
examples are listed in Table 7.2. A careful drug history (wher- cause time lost from school and create stresses for the whole
ever possible verified by checking tablets) may suggest a likely family. Good practice of clinical haematology requires consid-
offending agent. If the problem is of sufficient severity to eration of the far-reaching effects of the diagnosis and neces-
cause concern the drug should ideally be discontinued and sary treatment on the patient.
the blood count monitored to check resolution. It is as rele-
vant to obtain a history of allergy in haematology as in other
areas of medicine. Indeed, patients with haematological malig-
Miscellaneous
nancies are often given an unusually large number of chemo- Alcohol misuse can cause blood changes, the most common
therapeutic and antimicrobial agents and possible reactions being macrocytosis (enlarged red cells). A positive history will
have to be vigilantly documented to avoid repeat exposure. prevent unnecessary investigation for other causes. Smoking is
a cause of moderate polycythaemia (elevated haematocrit/
haemoglobin level) and appears to be associated with an
Family history
increased incidence of acute myeloid leukaemia. Travel to
As can be seen from Table 7.3, a number of blood diseases are tropical areas raises the possibility of malaria and other tropi-
inherited. A knowledge of the mode of inheritance is useful in cal diseases which can affect the blood.
diagnosis and essential in counselling the patient and family.
A simple question as to the presence of ‘anaemia’ or a ‘bleeding
problem’ in other family members can prevent unnecessary History taking
investigation and delay in diagnosis.
■ In the diagnosis of blood disorders, the history is complementary to
the clinical examination and laboratory testing.
Social history ■ Blood abnormalities such as anaemia, leucopenia and
thrombocytopenia lead to predictable groups of symptoms.
With the growing reliance on technology for diagnosis and
■ Blood abnormalities may be caused by systemic diseases, familial
treatment it can be surprisingly easy to forget that a blood
disorders and drugs. A thorough systemic enquiry, past medical
disorder is affecting a ‘real person’. An understanding of the
history, drug history and family history should be elicited.
patient’s normal lifestyle is particularly important where a
■ Serious and chronic blood diseases (e.g. leukaemia,
chronic or serious disease is diagnosed. Many people develop-
haemoglobinopathies, haemophilia) have major social implications
ing haematological malignancies are elderly and need support for children and adults; these should be explored not ignored.
in the community, including, perhaps, visits by social workers
16 2 THE HAEMATOLOGY PATIENT

8 Examining the patient


Abnormalities of the blood may arise as Table 8.1  Observation of the patient with
a result of a primary disorder of the a blood disorder. Some common signs
bone marrow (e.g. leukaemia) or from and their possible clinical relevance
a wide range of systemic disorders. A Clinical sign Possible haematological
thorough clinical examination is vital abnormality
Face
both to confirm a likely diagnosis and
Pallor Any anaemia
to exclude coexistent problems. There is Lemon tint Megaloblastic anaemia
not space here to detail all the elements Jaundice Haemolytic anaemia
of clinical examination; we have concen- Plethora Polycythaemia
trated on aspects of the examination Mouth
most relevant to patients with a primary Ulcers Neutropenia
Glossitis Megaloblastic anaemia
blood disorder.
Iron deficiency anaemia
Angular stomatitis Iron deficiency anaemia
Candida (‘thrush’) Immunosuppression
Look at the patient! Skin
Pallor Any anaemia
It is easy to examine a patient carefully Jaundice Haemolytic anaemia Fig 8.1  Massive cervical lymphadenopathy.
without properly observing them. A Excessive bruising Coagulation disorder,
deliberate inspection of the patient’s thrombocytopenia
face while taking the history may reveal Purpuric/petechial rash Thrombocytopenia
Table 8.2  Common causes of
Leg ulcers Sickle cell anaemia
vital clues even before the formal exami- lymphadenopathy
nation is commenced. Common exam- Localised
ples include the pallor of iron deficiency Local bacterial or viral infection
anaemia, the lemon tint of megaloblas- Lymphoma
universally performed but is crucial in a Metastatic malignancy
tic anaemia, the jaundice of a haemolytic
patient with haemophilia. Generalised
anaemia, and the plethora of polycythae- Systemic infection
mia. Before laying a hand on the patient, – bacterial (e.g. tuberculosis)
a careful inspection of the mouth and Examination of the – viral (e.g. Epstein–Barr, HIV)
skin may also point to particular blood lymph nodes Lymphoma
Other haematological malignancy (e.g. leukaemia)
abnormalities or disorders (Table 8.1).
Lymph nodes may be enlarged in Inflammatory disease (e.g. connective tissue disorder,
The patient’s ethnic origin can be of rel- sarcoid)
primary blood disorders and systemic
evance. Sickle cell anaemia is an unlikely Disseminated malignancy
diseases. Enlargement is referred to as
diagnosis in a patient with white skin
‘lymphadenopathy’ or just ‘adenopathy’.
while pernicious anaemia is equally
The differential diagnosis differs in gen-
unlikely in a patient with black skin.
eralised and localised forms of lym- cause (e.g. malignancy or infection);
Children with chronic blood disorders
phadenopathy (Table 8.2). In practice, formal ear, nose and throat examination
such as haemoglobinopathies are fre-
palpable lymphadenopathy is usually is often indicated.
quently thinner and shorter than their
limited to the cervical, axillary and The axillary nodes are best examined
healthy peers.
inguinal areas. with the patient supine and the arm
Enlargement of the cervical lymph supported by the side, the examiner
nodes is the most common cause of a using the right hand to gently palpate
General examination swelling in the neck and, if massive, may the left axilla and the left hand for the
Careful observation should be followed be easily visible (Fig 8.1). Following right axilla. Anatomically, the nodes are
by a methodical examination of the careful inspection of the neck, it is divided into medial, lateral, posterior,
major systems. The possible abnormali- easiest to examine the cervical nodes central and apical groups. Examination
ties in each system which may be seen from behind the seated patient, method- of inguinal nodes is most easily per-
in blood diseases are too numerous to ically palpating the anatomical areas formed while examining the abdomen.
detail here. They are referred to in the detailed in Figure 8.2. As for all lumps, Care must be taken not to confuse
relevant sections describing each disease. it is important to document not only the inguinal adenopathy with an irreducible
Although examination should be size and location of enlarged nodes, but femoral hernia. Enlarged abdominal
ordered, in a busy clinical practice it is also the shape, consistency and presence lymph nodes may cause an abnormal
often necessary to prioritise. Rectal of tenderness. Lymphadenopathy sec- fullness of the central abdomen on
examination is not routine in all patients ondary to infection is more often tender palpation.
with blood disorders but is definitely than that due to malignancy. Nodes On occasion it is difficult to be certain
indicated in unexplained iron deficiency involved by carcinoma are characteristi- that nodes are pathologically enlarged.
to exclude an otherwise asymptomatic cally stony hard while those involved by Interpretation must take account of the
rectal carcinoma; it is contraindicated in lymphoma are more ‘rubbery’. The pres- patient’s age and occupation. Large ton-
patients with suspected leukaemia and ence of cervical adenopathy should sillar glands are common in children,
neutropenia. Similarly, an exhaustive always prompt a thorough examination while people exposed to repeated minor
examination of the major joints is not of the head and neck to detect a local injuries of the hands and feet often have
Examining the patient 17

Table 8.3  Common causes of splenomegaly


Degree of Centimetres palpable Causes
Suboccipital enlargement below costal margin
Slight 0–4 Various acute and chronic infections
Sternomastoid muscle (e.g. septicaemia, tuberculosis)
Moderate 4–8 Haemolytic anaemia
Trapezius muscle Infectious mononucleosis
Upper deep cervical
(including tonsillar node) Portal hypertension
Posterior triangle Massive Greater than 8 Myelofibrosis
Supraclavicular Middle deep cervical Chronic myeloid leukaemia
Polycythaemia vera
Lower deep cervical Lymphoma
Malaria
Clavicle Leishmaniasis

Fig 8.2  Lymph nodes of the neck. Note: The division by size is clinically helpful but disorders associated with massive splenomegaly may
also cause lesser degrees of enlargement.

inspiration. If the spleen is not palpable


10th rib
using this technique, it is worth rolling
the patient slightly onto the right side
with the examiner’s left hand held
firmly behind the left lower ribs (Fig
8.4). This latter manoeuvre may lift
forward a slightly enlarged spleen and
make it palpable on deep inspiration.
The following features are typical of
an enlarged spleen:
■ It has a characteristic shape and
Fig 8.3  Schematic view of Fig 8.4  Examination of the spleen.
splenic enlargement. The sometimes a palpable notch on its
notch is frequently not palpable. upper edge.
The arrow shows the normal ■ You cannot get above it.
direction of enlargement. ■ It moves with respiration.
■ It is dull to percussion.
■ It cannot be felt bimanually or

balloted.
some lymphadenopathy in the draining and the patient is asked if they have any
areas. A period of observation can be abdominal tenderness. It is normal to In practice an enlarged spleen is most
helpful. If serious doubt persists then a palpate the whole abdomen and then likely to be misidentified as an enlarged
surgical biopsy is indicated. examine the major organs in turn. The left kidney. However, the kidney is not
spleen enlarges from below the tenth rib dull to percussion (it is covered by the
along a line heading for the umbilicus colon) and it can be felt bimanually and
Examination of
(Fig 8.3). Palpation for the spleen is com- balloted. It is worth listening with a
the spleen
menced in the right lower quadrant of stethoscope over an enlarged spleen as
The spleen is enlarged in many blood the abdomen, otherwise massive inflammation of the capsule may cause
disorders and in some systemic diseases enlargement can be missed. The hand is an audible ‘splenic rub’. The spleen is
(Table 8.3). The presence of a palpable moved in stages towards the tip of the usually uniformly enlarged and it is not
spleen and its characteristics often left tenth rib while the patient takes generally possible to identify the under-
narrows the differential diagnosis con- deep breaths. The edge of an enlarged lying disorder by palpation alone. The
siderably. Examination of the spleen is spleen connects with the tips of the degree of enlargement does, however,
frequently done badly. It is easy to miss index and middle fingers during deep give a diagnostic clue (see Table 8.3).
a slightly enlarged spleen which is just
palpable (‘tippable’) and it is also embar-
rassingly easy to miss a spleen which is
massively enlarged. However, neither of
Examining the patient
these mistakes is likely if the examina-
tion is conducted as below. ■ The clinical examination is an important part of the diagnosis of blood disorders.
The patient should be examined on a ■ It is helpful to carefully observe the patient prior to the formal examination of systems.
suitable examination couch or bed and ■ In routine clinical practice some aspects of examination are prioritised (e.g. rectal
should be encouraged to relax. The examination in unexplained iron deficiency).
whole abdomen is exposed. The exam-
■ Proper examination of the lymph nodes requires familiarity with the normal anatomical
iner sits or kneels to allow palpation groups and the causes of enlargement.
with a (warm) hand with the forearm
■ Examination of the spleen is frequently badly performed; with poor technique even
horizontal to the abdomen. First, the massive splenomegaly can be missed.
abdomen is inspected for a visible mass
18 2 THE HAEMATOLOGY PATIENT

9 Laboratory haematology I – Blood and


bone marrow
Diagnosis of most blood disorders is possible from a combi- HAEMATOLOGY YORK DISTRICT HOSPITAL TEL: (01904) 726802
nation of clinical history, clinical examination and relatively Surname
Other names
Hosp. No.
D.O.B.
Lab. No.
NHS No.

routine laboratory tests. Haematology laboratories are heavily Address Sex


Consultant/GP Source Accident & Emergency YDH Run 432
dependent on complex electronic machinery. The ubiquitous Clinical Chest pain
full blood count (FBC) is the archetypal haematological inves- Neutrophils 4.2 x109 /L ( 2.0 - 7.5 )
tigation and is performed by specialised automated cell Lymphocytes 2.3 x109 /L ( 1.0 - 4.5 )
Monocytes 0.5 x109 /L ( 0.2 - 1.2 )
counters. However, despite the accessibility of modern tech- Eosinophils 0.1 x109 /L ( 0.1 - 0.6 )
Basophils 0.0 x109 /L ( 0.0 - 0.2 )
nology, the more simple traditional techniques of blood and
Results validated and automatically authorised by computer
bone marrow film spreading, staining, and light microscopy
remain essential parts of the haematologist’s repertoire. HB WBC PLTS RBC MCV PCV MCH MCHC ESR
g/L x109/L x109/L x1012/L fl L/L pg g/dL mm/hr

133 7.1 246 4.46 89 0.397 29.8 33.5

The blood count


M 130 - 180 M 4.5 - 5.8 M 0.40 - 0.50 M 1 - 10
F 115 - 165 4.0 - 11.0 150 - 400 F 4.2 - 5.4 77 - 99 F 0.37 - 0.47 27 - 32 30 - 35 F 1 - 15

See handbook for paediatric ranges

Many of the diseases discussed in this book are first suggested NEDIFF, FBC
Sample type

Blood
Taken
04.09.09 13:32
Received
04.09.09 14:00
Date & Time printed
04/09.2009 14:58

by an abnormality in the blood count (often referred to as the


full blood count). The test is performed on a small specimen
Fig 9.1  Typical blood count report.
of anticoagulated venous blood; the normal anticoagulant is
ethylene diamine tetra-acetic acid (EDTA). A typical report is
illustrated in Figure 9.1. As can be seen, it contains a large
amount of numerical information pertaining to the three cell
lines in the peripheral blood: red cells (and haemoglobin),
white cells (with a differential count of each specific cell type)
and platelets.
When interpreting the report it is sensible to initially focus
on the haemoglobin (Hb) concentration, total white cell count
(WBC) and platelet count – most blood abnormalities of clini-
cal significance are associated with a derangement of at least
one of these values. Much of the remaining information
details the nature of the red cells and their degree of haemo-
globinisation, and the precise make-up of the white cell count. (a)
The former values are helpful in the diagnosis of anaemia,
and the latter in the diagnosis of a variety of diseases of white
cells (e.g. leukaemias) and reactions to systemic disease. To Fig 9.2  Blood film.
(a) Macroscopic;
understand the role of the automated blood count in clinical (b)
(b) light microscopy.
practice, and particularly its limitations, it is helpful to under-
stand how the numerical values are generated. light scattering method the cells deflect a beam of light (often
a laser beam) and a detector converts the scatter into pulses
Automated haematology counters proportional to cell size. For sophisticated measurements such
The two essential functions of the automated blood cell as the differential white cell count the two methods can be
counter are the measurement of Hb concentration in the used together with the addition of other modalities reliant on
blood and the counting and sizing of blood cells. biochemical reactions and light absorbance.
Most counters use a modification of the traditional cyan- Sophisticated though this technology is, automated cell
methaemoglobin method to measure Hb concentration. In counters are ultimately no substitute for the trained human
essence, blood is diluted in a solution where Hb is converted eye. Results outside the machine’s numerical normal range or
to cyanmethaemoglobin and then the Hb concentration the presence of unusual circulating cells (e.g. leukaemic cells)
derived from the light absorbance (optical density) of the should be flagged as being abnormal. This alerts the operator
resultant solution measured by a spectrophotometer. Auto- who will return to the original blood sample to make a film.
mated machines have at least two channels for cell counting.
In one, red cells and platelets may be counted and in the other
The blood film
red cells are lysed leaving white cells for analysis. Extra chan-
nels are often used for differential white cell and reticulocyte A blood film is simply made by smearing a drop of anticoagu-
counting. lated venous blood onto a glass slide with a glass spreader
There are two basic methods for cell counting and sizing: (Fig 9.2a). In larger laboratories film spreading can be auto-
electrical impedance and light scattering. The electrical imped- mated. Following drying, the film is fixed with methanol and
ance method relies on blood cells being very poor conductors stained. Routine stains are based on Romanowsky’s method
of electricity. Thus, when the cells are passed in a stream – commonly used variants are the May–Grünwald–Giemsa
through a narrow aperture across which an electrical current (MGG) stain and Wright’s stain. Constituent dyes include
is maintained, the individual cells create an increase in electri- methylene blue, azure B and eosin. Once stained, the blood
cal impedance of a size proportional to the cell volume. In the film should be systematically studied under the light
Laboratory haematology I – Blood and bone marrow 19

Table 9.1  Some morphological terms used in blood film reports


Red cells
Hypochromia Pale staining of cells
Polychromasia Grey-blue tint to cells (usually reticulocytes)
Anisocytosis Variation in cell size
Poikilocytosis Variation in cell shape
Macrocytosis/microcytosis Increase/decrease in cell size
Spherocyte Small spherical densely stained cell
Burr cell Crimpled cell membrane
Target cell Increased staining in middle of area of central
pallor – suggests increased surface area
Basophilic stippling Small basophilic inclusions in cytoplasm (RNA)
Howell–Jolly bodies Nuclear remnants in cytoplasm
Schistocyte Fragmented cell
White cells
Hypersegmented neutrophils Increased nuclear segmentation
Left-shifted neutrophils Reduced nuclear segmentation (a)
Toxic granulation Increased neutrophil cytoplasmic granularity Fig 9.3  Bone
Atypical lymphocytes Morphology variable; often seen in viral infections
marrow aspirate.
Blasts Leukaemic cells
(a) Macroscopic; (b)
Platelets (b) light microscopy.
Clumping Sticking together; can cause artefactually low count

Note: Causes of these morphological abnormalities are discussed in the disease sections.

microscope – the normal appearance of a film stained by the


MGG method is illustrated in Figure 9.2b. Alternative stains
are sometimes needed. Visualisation of reticulocytes requires
the use of a dye such as methylene blue on live unfixed cells
(‘supravital stain’). Malarial parasites are most easily seen fol-
lowing staining at a specific pH.
The first step in film examination is a decision as to whether
the film is of adequate quality. Either poor staining techniques
or prolonged storage of the specimen may make the film
worthless. Any comment on the film appearance is usually
appended to the blood count report. The nomenclature used
in film reporting can appear obscure; some more commonly (a)
used morphological terms are listed in Table 9.1. Microscopic
images of blood cells are now routinely photographed using Fig 9.4  Bone
marrow trephine
digital cameras. These images may increasingly be used to
specimen.
create ‘virtual slides’ or employed with cell recognition (a) Macroscopic; (b)
systems for automated morphological screening. (b) light microscopy.
Where the film is significantly abnormal, examination of
the bone marrow can give further diagnostic information. Trephine biopsy
The trephine biopsy (Fig 9.4) is sectioned and normally
stained by haematoxylin and eosin (H&E) and Giemsa
Bone marrow examination methods. Silver impregnation can be used to demonstrate
The clinical procedure for obtaining samples of bone marrow marrow fibrosis and Perl’s stain to highlight iron. The tre-
is described on page 106. From the favoured site, the posterior phine is less good than the aspirate for identifying morpho-
iliac crest, it is possible to obtain both a marrow aspirate logical abnormalities of individual cells but it is better for
sample and a marrow trephine biopsy. detecting abnormalities of marrow architecture and infiltra-
tion by solid malignancy. The two types of bone marrow
Aspirate sample are thus complementary.
The aspirate is simply sucked through the needle and spread
onto a glass slide; the marrow particles are normally easily
Laboratory haematology I
visible (Fig 9.3a). The marrow is fixed and stained as for a
– blood and bone marrow
blood film and additionally stained by Perl’s method to dem-
onstrate iron. Microscopy and reporting is systematic with ■ Many blood disorders are first suggested by an abnormality in the
reference to the overall cellularity, the appearance and number blood count – particularly in the haemoglobin concentration, total
white cell count or the platelet count.
of each normal cell line, possible infiltration by malignant
cells, and any other pathological features. The advantage of ■ Automated haematology counters measure haemoglobin
the aspirate specimen is that individual cells are well pre- concentration and count and size blood cells.
served and subtle morphological changes can be detected. The ■ Where the blood count is abnormal, examination of the blood film
major disadvantage is that the normal architecture of the often reveals morphological abnormalities undetectable by the
automated counter.
marrow is lost. In the investigation of haematological malig-
■ Significant blood abnormalities can be further investigated by
nancy (e.g. leukaemia) marrow aspirate samples are often also
examination of the bone marrow – aspirate and trephine biopsy
used for immunophenotyping and cytogenetic and molecular
specimens provide complementary information.
genetic testing.
20 2 THE HAEMATOLOGY PATIENT

10 Laboratory haematology II – Coagulation and


the acute phase response
Table 10.1  Common causes of abnormal first-line clotting tests
Simple tests of blood coagulation
Test Prolonged Prolonged APTT Low fibrinogen
Despite the complexity of haemostasis (p. 12), it is possible prothrombin time
to make a general assessment of coagulation with a few Common causes Warfarin Heparin1 DIC
relatively simple first-line tests. As an initial screen of haemo- Liver disease Haemophilia Severe liver disease
static function the following tests should be combined with Vitamin K deficiency vWD
a blood count and film to determine platelet number and DIC DIC
Liver disease
appearance.
Lupus anticoagulant

The prothrombin time (PT) APTT, activated partial thromboplastin time; DIC, disseminated intravascular coagulation; vWD, von
Willebrand disease.
The test is performed by adding thromboplastin to the 1
Unfractionated heparin.
patient’s platelet-poor plasma, warming, and then adding
calcium. The time to clot formation is recorded in seconds
and the PT may be expressed as the ratio of the patient’s time
to a normal control time. The thromboplastin used should
have been calibrated to allow this result to be converted to
the international normalised ratio (INR) – the ratio which
would have been obtained if the international reference prep-
aration for thromboplastin had been used in the test (see p.
80). The PT is essentially a measure of the efficiency of the
extrinsic clotting system (factor VII) in addition to the func-
tioning of factors V and X, prothrombin and fibrinogen.

Activated partial thromboplastin time (APTT)


This test is sometimes referred to as the partial thromboplas-
tin time with kaolin (PTTK) or the kaolin cephalin clotting
time (KCCT). Patient platelet-poor plasma is combined with
contact factors (kaolin, phospholipid) and calcium and the
time to clot formation recorded in seconds. The test measures
the overall efficiency of the intrinsic pathway (i.e. factors VIII,
IX, XI, XII) as well as the function of factors V, X, pro-
thrombin and fibrinogen. Fig 10.1  Measurement of the ESR.

Quantitation of plasma fibrinogen


In most laboratories this has replaced the thrombin time as the acute phase response suggests a physical cause for symp-
a first-line test. Several accurate methods are available for the toms. Possibilities include trauma, infections, neoplasia and
quantitative assay of plasma fibrinogen. Fibrinogen is an acute autoimmune disease. Serial measurements can be useful in
phase reactant (see below) and is frequently elevated in sick monitoring the effects of treatment. The most widely used
patients. Causes of low levels include disseminated intravas- measurements of the acute phase response are the erythrocyte
cular coagulation (DIC) and severe liver disease. sedimentation rate (ESR), the plasma viscosity, and C-reactive
Common clinical causes of abnormal first-line coagulation protein.
tests are shown in Table 10.1. Second-line tests may be needed
for more precise diagnosis. In mixing experiments (or correc- ESR
tion tests) patient plasma is mixed with normal or factor- In this simple and inexpensive test venous blood (in citrate
deficient plasma prior to repeating first-line tests. If a particular anticoagulant) is drawn up into a vertical tube (Fig 10.1) and
coagulation factor is thought to be lacking, a quantitative allowed to stand for 1 hour. The red cells settle out of suspen-
assay can then be performed. A circulating inhibitor of coagu- sion and the length of plasma cleared after the hour is meas-
lation is suggested by failure of the coagulation abnormality ured. The normal values are less than 5 mm/hour in men and
to be corrected by the addition of normal plasma. Many less than 7 mm/hour in women, although values of up to
routine tests are now automated. Most coagulation instru- 15 mm/hour are not infrequent in those over 60 years old.
ments rely on measurement of changes in optical density to The test mainly reflects fibrinogen levels but is also influenced
detect clot formation. by α2-macroglobulin, immunoglobulins and albumin. These
proteins buffer the electrostatic repellent forces on the red cell
membrane and allow the cells to come together and form
Measurement of the acute reversible aggregates or rouleaux which fall more quickly
phase response through the plasma. The ESR result is affected by the haemo-
In assessing patients with ill-defined symptoms it can be globin concentration with high values seen in anaemia and
helpful to measure activation of the acute phase response, the low values in polycythaemia. A fresh sample must be pro-
body’s response to tissue damage. Evidence of activation of cessed as the result also changes over time.
Laboratory haematology II – Coagulation and the acute phase response 21

Table 10.2  Clinical significance of the plasma viscosity


Plasma viscosity (mPa·s)
measured at:
25°C 37°C
Normal range1 1.50–1.72 1.15–1.35
Acute/chronic organic diseases 1.75–2.55 1.36–1.99
(malignancy, infection, etc.)
Need to exclude paraproteinaemias/ >2.55 >2.00
hyperviscosity syndrome

1
Slightly higher levels can be seen in normal older people.

Plasma viscosity
This test also measures the acute phase response indirectly, Fig 10.2  Urine electrophoresis. The highlighted sample demonstrates
the result correlating with fibrinogen and immunoglobulin proteinuria and the presence of Bence Jones (immunoglobulin light chain)
levels. The plasma viscosity has some advantages over the protein (red arrow) in a patient with myeloma and renal failure.
ESR. The normal range is the same in males and females and
the result is independent of haemoglobin concentration. The
sample can be taken from the EDTA anticoagulated blood
count bottle and the test does not need to be performed
immediately. The normal range, which is temperature depend-
ent, is detailed in Table 10.2. Plasma viscosity measurement
has direct pathophysiological relevance in myeloma where
very high values are seen in the hyperviscosity syndrome.

C-reactive protein (CRP)


This easily measured protein is elevated in most types of
tissue injury. The CRP is usually increased within 6–8 hours
of the insult. The normal range is up to 10 mg/L with levels
of 10–40 mg/L in severe viral infections, levels of 40–300 mg/L
in bacterial infections and levels over 300 mg/L in severe
burns. CRP results are not influenced by anaemia.

Other possible measures of the acute phase reaction include


quantitation of fibrinogen, haptoglobins, alpha-1-antitrypsin
and anti-chymotrypsin. These all rise following tissue damage
but some acute phase reactants (notably albumin and trans-
ferrin) actually fall.

Electrophoresis Fig 10.3  Flow cytometry. The use of a combination of myeloid and
lymphoid antibodies shows multiple different cell lines and maturation
Electrophoresis has two routine applications in haematology. stages each in a different colour.
In the diagnosis of haemoglobinopathies (e.g. thalassaemia),
cellulose acetate electrophoresis at alkaline pH is used to
separate the abnormal haemoglobins. Citrate agar electro- conjugated with a fluorochrome, a molecule which emits light
phoresis at a lower pH may be helpful in selected cases. In at a specific wavelength when excited by a laser. The flow
the investigation of myeloma, serum and urine electrophore- cytometer is then used to detect populations of cells labelled
sis is performed to detect the monoclonal immunoglobulin by the fluorescent marker (Fig 10.3). Flow cytometry may be
or light chains characteristic of the disease (Fig 10.2). used in conjunction with molecular methods for the detection
of minimal residual disease in leukaemia.

Flow cytometry
Flow cytometry is essentially the measurement of the charac- Laboratory haematology II
teristics of cells passing in a fluid stream through a detection – coagulation and the acute
apparatus. The automated cell counters described in the previ- phase response
ous section are the major application of the flow cytometry
■ Despite the complexity of haemostasis the coagulation mechanism
principle in haematology but the technique also plays a key
can be assessed with a few relatively simple ‘first-line’ tests.
role in the diagnosis of haematological malignancy. Leukae-
■ The term ‘acute phase response’ describes the body’s response to
mic cells often have a particular ‘immunophenotype’ – a char-
tissue damage; commonly used measures include the ESR, plasma
acteristic pattern of detectable antigens on the cell surface and viscosity and C-reactive protein.
in the cell cytoplasm (see also relevant disease sections). The
■ Electrophoresis
is routinely used in the diagnosis of
antigens are identified by cluster differentiation (CD) numbers
haemoglobinopathies and in the investigation of myeloma.
(e.g. CD13 is a myeloid antigen; see Appendix II). Cells from
■ Flowcytometry methodology is exploited in automated blood cell
peripheral blood or a bone marrow aspirate sample are incu-
counters and plays a key role in the characterisation of leukaemia.
bated with specific CD monoclonal antibodies which are
22 3 ANAEMIA

11 Introduction and classification


have been performed, anaemia is more addition to haemoglobin concentration.
Definition common in women than in men. Par- Abnormal red cell indices should be
The term ‘anaemia’ refers to a reduction ticularly susceptible groups include confirmed by microscopic examination
of haemoglobin or red cell concentra- pregnant women, children under 5 of blood films. The ‘morphological’ clas-
tion in the blood. With the widespread years and the elderly. The majority of sification is based on a correlation
introduction of automated equipment cases in younger people are caused by between red cell indices and the under-
into haematology laboratories the hae- iron deficiency. Anaemia is surprisingly lying cause of anaemia. The most impor-
moglobin concentration has replaced common in the elderly, affecting roughly tant measurements are of red cell size
the haematocrit (or ‘packed cell volume’) 10% of people over 65 years. Up to a (mean cell volume or MCV) and red cell
as the key measurement. Haemoglobin third of these cases remain unexplained haemoglobin concentration (mean cell
concentration can be determined accu- (see p. 92). In developing countries, haemoglobin (MCH) or mean cell hae-
rately and reproducibly and is probably factors influencing the prevalence of moglobin concentration (MCHC)).
the laboratory value most closely cor- anaemia include climate, socioeconomic Anaemias with raised, normal and
related with the pathophysiological con- conditions and, most importantly, the reduced red cell size (MCV) are termed
sequences of anaemia. Thus, anaemia is incidence of coexistent diseases. macrocytic, normocytic and microcytic,
simply defined as a haemoglobin con- respectively. Anaemias associated with
centration below the accepted normal a reduced haemoglobin concentration
range. within red cells are termed hypochro-
General features
The normal range for haemoglobin mic and those with a normal MCH are
concentration varies in men and women In anaemia the blood’s reduced oxygen- termed normochromic. Characteristic
and in different age groups (Table 11.1). carrying capacity can lead to tissue combinations are of microcytosis and
The definition of normality requires hypoxia. The clinical manifestations of hypochromia, and normocytosis and
accurate haemoglobin estimation in a significant anaemia (see also p. 14) are normochromia. As can be seen in Figure
carefully selected reference population. to a large extent due to the compensa- 11.1, this terminology is helpful in nar-
Subjects with iron deficiency (up to 30% tory mechanisms mobilised to counter- rowing the differential diagnosis of
in some unselected populations) and act this hypoxia. Cardiac overactivity anaemia. It is perhaps least helpful in
pregnant women must be excluded or causes palpitations, tachycardia and normocytic anaemia as the possible
the lower level of normality will be mis- heart murmurs. The dyspnoea of severe causes are numerous and diverse.
leadingly low. Normal haemoglobin anaemia may be a sign of incipient car- The value of the blood film in diagno-
ranges may vary between ethnic groups diorespiratory failure. Pallor is due pri- sis should not be underestimated. For
and between populations living at differ- marily to skin vasoconstriction with instance, combined iron deficiency (a
ent altitudes. redistribution of blood flow to tissues cause of microcytosis) and folate defi-
with higher oxygen dependency such as ciency (a cause of macrocytosis) may
the brain and myocardium. cause an anaemia with a normal MCV.
Prevalence Anaemia is one of the most common However, inspection of the film will
The prevalence of anaemia and the aeti- clinical problems presenting in general reveal a dual population of microcytic
ologies vary in different populations. In practice, in hospitals and in medical hypochromic red cells and macrocytic
developed countries where most studies examinations. Usually characteristic red cells.
symptoms and signs prompt a blood
count to confirm the diagnosis but on Aetiological classification
occasion an unexpectedly low haemo- Figure 11.2 illustrates a classification of
Table 11.1  Normal haemoglobin
globin estimation in a ‘routine’ blood anaemia based on cause. It is less imme-
concentrations at different ages count precedes the clinical consultation. diately helpful than the morphological
Age Mean Lower limit Whatever the sequence of events, classification in forming a differential
haemoglobin of normal anaemia is not in itself an adequate diag- diagnosis but it does illuminate the
(g/L) (g/L) nosis; further enquiry to establish the pathogenesis of anaemia. The funda-
Birth (cord blood) 165 135 underlying cause is essential. mental division is between excessive
1–3 days (capillary) 185 145 A logical approach to anaemia loss or destruction of mature red cells,
1 month 140 100 demands a clear understanding of both and inadequate production of red cells
2–6 months 115 95 its possible causes and its clinical and by the marrow.
6 months–2 years 120 105
laboratory features. There are two major Loss of red cells occurs in haemor-
2–6 years 125 115
classifications – both have advantages rhage and excessive destruction in
6–12 years 135 115
and they are best used together. haemolysis. A normal bone marrow will
12–18 years:
respond by increasing red cell produc-
  female 140 120
tion with accelerated discharge of young
  male 145 130 Classification
Adult: red cells (reticulocytes) into the blood.
  female 140 115 Morphological classification Inadequate red cell production may
  male1 155 135 As already discussed (p. 18), modern result from insufficient erythropoiesis
1
Normal haemoglobin concentration probably slightly lower after
electronic laboratory equipment can (i.e. a quantitative lack of red cell precur-
65 years. provide estimations of red cell indices in sors) or ineffective erythropoiesis (i.e.
Introduction and classification 23

Anaemia Microcytic Normocytic Macrocytic


type Hypochromic Normochromic

Red cell MCV and MCH MCV and MCH MCV


indices low normal raised

Blood loss (acute)


Common Iron deficiency Haemolysis1 Megaloblastic
examples Thalassaemia Chronic disease2 anaemias
Marrow infiltration

1 Occasionally macrocytic 2 Occasionally microcytic hypochromic


Fig 11.1  Classification of anaemia based on red cell measurement.

Reduced bone marrow Ineffective red cell


Nutritional deficiency erythroid cells (e.g. aplastic formation (e.g. chronic
(e.g. iron, vitamin B12, anaemia, marrow infiltration inflammation,
folate) by leukaemia or thalassaemia, renal
malignancy) disease)

Increased Failure of Dilution of red


Loss of red cells destruction production of red cells by increased
due to bleeding of red cells cells by the plasma volume
(haemolytic anaemias) bone marrow (e.g. hypersplenism)

Anaemia

Fig 11.2  Classification of anaemia based on cause.

defective erythrocytes destroyed in the Blood transfusion should only be used life-saving in a profoundly anaemic
marrow). Examples of insufficient eryth- where the haemoglobin is dangerously patient but it should be undertaken with
ropoiesis include bone marrow hypo- low, where there is risk of a further dan- great caution as heart failure can be
plasia, as in aplastic anaemia, and gerous fall in haemoglobin (e.g. rapid exacerbated. Mild anaemia in the elderly
infiltration of the marrow by a leukae- bleeding), or where no other effective should not be overlooked as it is a fre-
mia or other malignancy. Inefficient treatment of anaemia is available. quent cause of debility and has been
erythropoiesis is seen in disorders such Prompt blood transfusion can be linked with increased mortality.
as megaloblastic anaemia, thalassaemia
and myelodysplastic syndromes.
The above provides a useful frame-
work for thinking about anaemia. In
reality different mechanisms can operate
simultaneously. The anaemia of thalas- Anaemia: introduction and
saemia is caused by both ineffective classification
erythropoiesis and haemolysis. ■ Anaemia is defined as a haemoglobin concentration below the accepted normal range.
■ The normal range for haemoglobin is affected by sex, age, ethnic group and altitude.
Management ■ Theclinical features of anaemia are largely caused by compensatory measures mobilised to
counteract hypoxia.
The treatment of specific types of
anaemia is discussed in subsequent sec- ■ Anaemia can be classified according to red cell morphology or aetiology.
tions. However, some general state- ■ Red cell indices and morphology correlate with the underlying cause of anaemia.
ments can be made. Whenever possible, ■ Wherever possible the cause of anaemia should be determined before treatment is started.
the cause of anaemia should be deter-
■ Blood transfusion is only required in a minority of cases.
mined before treatment is instituted.
24 3 ANAEMIA

12 Iron deficiency anaemia

Iron
Iron is a constituent of haemoglobin Red
and rate limiting for erythropoiesis. The blood cells
metabolism of iron in the body is domi-
Erythroid bone
nated by its role in haemoglobin synthe- marrow
sis (Fig 12.1). Normally, the total iron
content of the body remains within
Liver Macrophages
narrow limits: absorption of iron from
Spleen
food (usually up to 3–4 mg/day) must
replace any iron losses. Iron is not
excreted as such but is lost in desqua- Serum transferrin-Fe
mated cells, particularly epithelial cells
Absorption Excretion
from the gastrointestinal tract. Menstru-
ating women will lose an additional Gut
highly variable amount of iron, and in
pregnant women the rate of iron loss is Fig 12.1  The normal iron cycle. Iron is absorbed from the gut into plasma where it is transported to
the bone marrow for haemoglobin synthesis. Dying red cells are engulfed by macrophages in the
about 3.5 times greater than in normal
reticuloendothelial system, and iron is recycled into the plasma for reuse. Iron is transported in the
men. The storage forms of iron, ferritin plasma bound to the glycoprotein, transferrin. Transferrin receptors exist on most cells in the body. Of
and haemosiderin, constitute about 13% the total 4–5 g of iron in the body only about 0.1% is being recycled at any given time. The rest is in
of total body iron. The small peptide tissue-specific proteins such as haemoglobin (66% of total body iron) and myoglobin, or stored in ferritin.
hepcidin plays a key role in iron metab-
olism and absorption (see p. 36).

Iron deficiency
Clinically significant iron deficiency is
characterised by an anaemia which can
usually be confidently diagnosed on the
basis of the clinical history and simple
laboratory tests. It cannot be over-
stressed that the diagnosis of iron defi-
ciency is not adequate in itself – a cause Fig 12.3  Glossitis and angular stomatitis in
for the deficiency must always be sought. iron deficiency.

■ Symptoms and signs specific to


Causes Fig 12.2  Carcinoma of the colon. A iron deficiency. Iron is required by
53-year-old man presented to his doctor
The likely cause will vary with the age, complaining only of tiredness. A blood count was many tissues in the body, shortage
sex and geographic location of the consistent with iron deficiency (Hb 76 g/L, MCV particularly affecting endothelial cells.
patient (Table 12.1). Iron deficiency is 69 fl) and this was confirmed by a low serum Patients with long-standing deficiency
usually caused by long-term blood loss, ferritin level. History and examination revealed no may develop nail flattening and
most often gastrointestinal or uterine obvious cause for his iron deficiency. koilonychia (concave nails), sore
Colonoscopy revealed a large bowel carcinoma tongues and papillary atrophy,
bleeding and less commonly bleeding in
which was successfully resected.
the urinary tract or elsewhere. Particu- angular stomatitis (Fig 12.3),
larly in elderly patients, deficiency may dysphagia due to an oesophageal web
infection is the commonest cause of
be the presenting feature of gastrointes- (Plummer–Vinson syndrome) and
iron deficiency worldwide. Malabsorp-
tinal malignancy (Fig 12.2). Hookworm gastritis. Many patients have none of
tion and increased demand for iron, as
these and their absence is thus of
in pregnancy, are other possible causes.
Table 12.1  Causes of iron deficiency little significance. Iron deficiency in
Poor diet may exacerbate iron deficiency
Very common young children can contribute to
but is rarely the sole cause outside the
■ Bleeding from the gastrointestinal tract (e.g. benign psychomotor delay and behavioural
ulcer, malignancy, hookworm) growth spurts of infancy and teenage
problems (see also p. 91).
■ Menorrhagia years.
■ Symptoms and signs due to the
Other
underlying cause of iron
■ Pregnancy
■ Malabsorption (e.g. coeliac disease, Helicobacter Clinical features deficiency. Patients may
pylori gastritis1) spontaneously complain of heavy
These can be conveniently grouped into
■ Malnutrition periods, indigestion or a change in
■ Bleeding from urinary tract three categories:
bowel habit. Once the diagnosis of
■ Pulmonary haemosiderosis
■ General symptoms and signs of iron deficiency is known, it is often
1
May also cause bleeding. anaemia (see pp. 14 and 22). useful to retake the history and
Iron deficiency anaemia 25

Table 12.2  Tests to confirm iron deficiency


Test Result in iron Comment
deficiency
Ferritin Low Level increased in chronic inflammation/liver disease
Transferrin saturation Low Low levels also in elderly and chronic disease
Serum iron Low Levels fluctuate significantly and low in chronic disease
Transferrin concentration1 High Useful test as low in anaemia of chronic disease
Zinc protoporphyrin High Late finding only
BM iron Low Informative but invasive investigation
Serum transferrin receptor level High Also high in haemolysis
Percentage of hypochromic red cells High Limited availability
Reticulocyte haemoglobin content Low Limited availability

BM, bone marrow.


1
Total iron binding capacity (TIBC) may alternatively be used.

Fig 12.4  Blood film from a patient with iron Investigation of Table 12.3  Failure to respond to oral iron
deficiency. The red cells are hypochromic (pale underlying cause – possible causes
staining) and microcytic. Where the likely cause is apparent, ■ Wrong diagnosis (i.e. other cause of anaemia)
re-examine the patient with a view to further investigations can be highly ■ Non-compliance
■ Malabsorption
detecting any clue of an underlying selective. Thus in a young woman with
■ Continued bleeding
disorder. Rectal examination should severe menorrhagia and no other symp-
be routine. toms it can be assumed that uterine
bleeding is the cause of iron deficiency, should be considered (see Table 12.1). In
and investigation of the gastrointestinal 20% of cases of iron deficiency no cause
Diagnosis
(GI) tract is not necessary. A gynaeco- is found.
The diagnosis may be suspected on the logical referral would be adequate. Com-
basis of the history and examination but plaints of indigestion or a change in Correction of iron deficiency
laboratory investigations are required bowel habit should prompt an endos- Oral iron is given to correct the anaemia.
for confirmation. copy or a colonoscopy or barium enema The normal regimen is ferrous sulphate
as first investigations. However, often 200 mg three times a day (providing
The blood count
there are no symptoms suggesting a site 195 mg elemental iron daily). Side-
Iron deficiency causes a hypochromic
of blood loss. The GI tract is by far the effects, including nausea, epigastric pain,
microcytic anaemia. The automated red
most common site in men and post- diarrhoea and constipation, are best
cell analyser generates a report with hae-
menopausal women. Faecal occult blood managed by reducing the dosage rather
moglobin, MCV and MCH values below
testing is inadequately sensitive to than changing the preparation. An ade-
the normal range (see p. 22). There is a
exclude gastrointestinal bleeding and quate response to oral iron is an increase
variation in red cell size (anisocytosis)
therefore a reasonable approach to this in haemoglobin of 20 g/L every 3 weeks.
reflected by a high red cell distribution
common problem is to commence with Iron is given for at least 6 months to
width (RDW). A blood film will show
colonoscopy and, if normal, to proceed ensure body stores are replete. There are
characteristic features (Fig 12.4).
to upper GI endoscopy. If upper GI several possible causes of a failure to
Confirmatory tests endoscopy is performed first in an respond to oral iron (Table 12.3).
Further tests are helpful in confirming elderly patient and shows a benign Parenteral iron (intramuscular or intra-
the diagnosis (Table 12.2) and excluding ulcerative lesion then assessment of the venous) can be used where oral therapy
other causes of a hypochromic micro- lower GI tract should probably still be is unsuccessful because of poor tolera-
cytic anaemia (see p. 23). Measurement performed as coexistent colonic neo- bility or compliance or where there is
of serum ferritin is probably the most plasms are found in a significant minor- continuing blood loss or malabsorption.
useful of these tests: a low level always ity of cases. Anti-tissue transglutaminase Preparations include iron dextran, iron
indicates iron deficiency but a normal (tTG-IgA) is a simple screening method sucrose and ferric caboxymaltose. Ana-
level does not guarantee normal stores for coeliac disease. If the GI tract is phylactic reactions can occur and a test
as ferritin is increased in chronic inflam- normal, rare causes of iron deficiency dose may be indicated.
mation and liver disease. In occasional
difficult cases (e.g. where the patient has
recently been transfused) a bone marrow
aspirate is helpful in showing absence
of iron stores. In practice the most likely Iron deficiency anaemia
confusion is with the anaemia of chronic
■ Iron is a constituent of haemoglobin and is essential for erythropoiesis.
disease (p. 36).
■ Iron deficiency is most often caused by long-term blood loss.
■ Iron deficiency causes a hypochromic microcytic anaemia.
Management
■ The anaemia is usually easily corrected with oral iron supplements.
This is divisible into investigations of the
■ It is important to establish the cause of iron deficiency – it may be the presenting feature
underlying cause and the correction of of gastrointestinal malignancy.
iron deficiency.
26 3 ANAEMIA

13 Megaloblastic anaemia
The megaloblastic anaemias are charac- (‘ineffective haematopoiesis’) or enter vitamin B12 (cobalamin) or folate (pter-
terised by delayed maturation of the the bloodstream as enlarged, misshapen oylmonoglutamate). It is one of the
nucleus of red cells in the bone marrow cells with a reduced survival time. In most common causes of a macrocytic
due to defective synthesis of DNA. clinical practice megaloblastic anaemia anaemia.
Red cells either die in the marrow is almost always caused by deficiency of

Why does deficiency of vitamin B12 or folate lead to megaloblastic anaemia?


Key characteristics of these essential Table 13.1  Vitamin B12 and folate
vitamins are summarised in Table 13.1. Characteristic Vitamin B12 Folate
Both folate and vitamin B12 are neces- Average dietary intake/day (µg) 20 2501
sary for the synthesis of DNA (Fig 13.1). Minimum adequate intake/day (µg) 1–2 1501
Folate is needed in its tetrahydrofolate Major food sources Animal produce only Liver, vegetables
form (FH4) as a cofactor in DNA synthe- Normal body stores Sufficient for several years Sufficient for a few months
sis. Deficiency of B12 leads to impaired Mode of absorption Combined with transport protein (IF) secreted Dietary folate converted to
conversion of homocysteine to methio- by gastric parietal cells – then absorbed methyl THF and absorbed in
nine causing folate to be ‘trapped’ in through ileum via special receptors duodenum and jejunum

the methyl form. The resultant defi- THF: tetrahydrofolate; IF: intrinsic factor.
ciency in methylene FH4 deprives the 1
500 µg daily required in pregnancy.

cell of the coenzyme necessary for DNA


formation.
All dividing cells in the body suffer
dUMP dTMP DNA
from the impaired DNA synthesis of B12
and folate deficiency. However, the
actively proliferating cells of the bone
marrow are particularly affected. As Methylene FH4 FH2
RNA synthesis progresses unhindered
in the cytoplasm, the erythroid cells
develop nuclear–cytoplasmic imbalance
with abundant basophilic cytoplasm and
Methyl FH4 FH4
enlarged nuclei. The chromatin pattern
in the nucleus is characteristically abnor-
mal; one author has described it as
resembling ‘fine scroll work’, another as Homocysteine Methionine
Vit. B12
‘sliced salami’ (Fig 13.2). The slowed syn-
thesis of DNA leads to prolonged cell Fig 13.1  The cause of megaloblastic
cycling and the cells being discharged anaemia. Both vitamin B12 and folate (FH4) are
necessary for normal synthesis of DNA (see text).
into the blood without the normal quota
of divisions. Red cells are enlarged and
egg-shaped and the neutrophils hyper-
segmented due to retention of surplus The clinical hallmarks of pernicious
nuclear material (Fig 13.3). anaemia are gastric parietal cell atrophy
and achlorhydria, a more generalised
Fig 13.2  Bone marrow aspirate in
epithelial cell atrophy and megaloblastic
Clinical syndromes megaloblastic anaemia. The immature red cells
anaemia. The disease is most common show nuclear–cytoplasmic imbalance with
Vitamin B12 deficiency in northern Europe in women greater enlarged abnormal nuclei and basophilic
Pernicious anaemia than 50 years of age and is familial. cytoplasm.
This classic cause of vitamin B12 defi- Affected patients classically have prema-
ciency is an autoimmune disorder. Most ture greying of the hair and blue eyes
patients have IgG autoantibodies tar- and may develop other autoimmune arises from demyelination of the dorsal
geted against gastric parietal cells and disorders including vitiligo, thyroid and lateral columns of the spinal cord.
the B12 transport protein intrinsic factor. disease and Addison’s disease. Slight Patients most commonly complain of
The precise pathogenesis, and particu- jaundice is caused by the haemolysis of an unsteady gait, and if B12 deficiency is
larly the role of the autoantibodies, is ineffective erythropoiesis. not corrected there can be progression
incompletely understood but B12 defi- Patients usually have symptoms of to irreversible damage of the central
ciency ultimately arises from reduced anaemia and the generalised epithelial nervous system with paralysis and
secretion of intrinsic factor (IF) by pari- abnormality can manifest as glossitis dementia. There is a possible increased
etal cells and, hence, reduced availability (Fig 13.4) and angular stomatitis. The incidence of carcinoma of the stomach
of the B12–IF complex which is absorbed archetypal neurological complication – and colorectal cancer in pernicious
in the terminal ileum. ‘subacute combined degeneration’ – anaemia.
Megaloblastic anaemia 27

Table 13.2  The megaloblastic anaemias Other causes of vitamin B12


Vitamin B12 deficiency deficiency
Deficiency of gastric Pernicious anaemia These are mostly abnormalities of the
intrinsic factor Gastrectomy stomach and ileum (Table 13.2). As
Intestinal Ileal resection/Crohn’s disease normal body stores are sufficient for 2
malabsorption Stagnant loop syndrome
years, clinically apparent deficiency
Tropical sprue
Fish tapeworm from any cause will develop slowly.
Congenital malabsorption
Dietary deficiency Vegans Folate deficiency
(rare) Folate deficiency is caused by dietary
Folate deficiency
insufficiency, malabsorption, excessive
Dietary deficiency
Malabsorption Coeliac disease
utilisation or a combination of these
Tropical sprue (Table 13.2). Patients may complain of
Small bowel disease/resection symptoms of anaemia or of an underly-
Increased Pregnancy ing disease. The increased risk of
requirement Prematurity
thrombosis is because of associated
Haemolytic anaemia
Myeloproliferative/malignant/
hyperhomocysteinaemia (see p. 79).
inflammatory disorders There is a macrocytic anaemia and a
Other causes megaloblastic bone marrow. In signifi-
Fig 13.3  Peripheral blood film in
Drug-induced Folate antagonists cant deficiency both serum and red cell
megaloblastic anaemia. There is a macrocytosis
suppression of Metabolic inhibitors folate are usually low but the latter is
and the neutrophils are hypersegmented.
DNA synthesis Nitrous oxide (prolonged use)
the better measure of tissue stores. In
Inborn errors Hereditary orotic aciduria
addition to a thorough dietary history
patients may need investigations for
malabsorption (e.g. jejunal biopsy).
Folate deficiency is treated with oral
4. Autoantibodies. Parietal cell
folic acid 5 mg once daily. This is given
antibodies are found more
for several months at least, the precise
commonly in the serum than IF
duration of therapy depending on the
antibodies (90% vs 50%) but
underlying cause. Folate is prescribed
whereas IF antibodies are almost
prophylactically in pregnancy (400 µg
diagnostic of pernicious anaemia,
daily) with a reduction in neural tube
parietal cell antibodies occur in
Fig 13.4  Painful glossitis in pernicious defects in the fetus and also in groups
anaemia. about 15% of healthy elderly people.
of patients at high risk of deficiency
5. Tests for vitamin B12 absorption. The
(Table 13.2). Before folate is prescribed,
urinary secretion (Schilling) test was
vitamin B12 deficiency must be excluded
formerly much used but radioactive
Diagnosis (or corrected) as subacute combined
cyanocobalamin is not available now
1. Blood count and film. There is a degeneration of the cord can be
and the test is obsolete.
macrocytic anaemia with the typical precipitated.
film appearance of megaloblastic Treatment
anaemia. There may be leucopenia Vitamin B12 levels are usually replen-
and thrombocytopenia. ished by intramuscular injection of the
2. Bone marrow aspirate. This is not vitamin. Several injections of 1 mg
always necessary. It will confirm hydroxycobalamin are given over the
megaloblastic anaemia but will not first few weeks and then either one
illuminate the underlying cause. injection every 3 months or daily oral Megaloblastic
3. Estimation of vitamin B12 and folate vitamin B12 1–2 mg daily for life. The anaemia
levels. In pernicious anaemia the increase in reticulocytes in the blood
■ Megaloblasticanaemia is a common
serum vitamin B12 level is normally peaks 6–7 days after the start of
cause of a macrocytic anaemia.
very low but the assay is not treatment.
■ In clinical practice it is almost always
entirely reliable and a trial of In practice, ill patients with megalob-
caused by deficiency of vitamin B12 or
therapy may be justified where lastic anaemia are often started on both folate.
clinical and blood features strongly B12 and folate supplements after a blood
■ Vitamin B12 deficiency normally arises
suggest deficiency. Serum sample has been taken for assay of the
from malabsorption – the classic
methylmalonate and homocysteine vitamins. When the results are known clinical syndrome is the autoimmune
levels are raised in B12 deficiency but the unnecessary vitamin can be stopped. disorder pernicious anaemia.
their role in diagnosis is limited by Blood transfusion is best avoided as it ■ Folate deficiency is more often due to
their being often increased in may lead to circulatory overload – where frank dietary deficiency or increased
normal people and a range of other judged necessary to correct hypoxia, dietary requirements as in pregnancy.
disorders. Serum folate may be it is undertaken with extreme caution. ■ Vitamin B12 deficiency should be
elevated and the red cell folate Platelet transfusion is used for bleeding excluded or corrected before folate is
reduced (folate is trapped in its caused by severe thrombocytopenia but administered as subacute combined
degeneration of the cord can be
extracellular methyl FH4 form – see this is unusual. Hypokalaemia occasion-
precipitated.
Fig 13.1). ally requires correction.
28 3 ANAEMIA

14 Haemolytic anaemia I – General features and


inherited disorders

General features of haemolysis


The term ‘haemolytic anaemia’ describes Table 14.1  Classification of the haemolytic anaemias
a group of anaemias of differing aetiology Inherited disorders
that are all characterised by abnormal Red cell membrane Hereditary spherocytosis and hereditary elliptocytosis
destruction of red cells. The hallmark of Haemoglobin Thalassaemia syndromes and sickling disorders
these disorders is reduced lifespan of the Metabolic pathways Glucose-6-phosphate dehydrogenase and pyruvate kinase deficiency

red cells rather than underproduction by Acquired disorders


Immune Warm and cold autoimmune haemolytic anaemia
the bone marrow.
Isoimmune Rhesus or ABO incompatibility (e.g. haemolytic disease of newborn, haemolytic
In classification of the haemolytic transfusion reaction)
anaemias there are three main Non-immune and trauma Valve prostheses, microangiopathy, infection, drugs or chemicals, hypersplenism
considerations:
■ The mode of acquisition of the
disease: is it an inherited disorder or
a disorder acquired in later life?
■ The location of the abnormality: is

the abnormality within the red cell


(intrinsic) or outside it (extrinsic)?
■ The site of red cell destruction: red

cells may be prematurely destroyed


in the bloodstream (intravascular
haemolysis) or outside it in the
Fig 14.1  Mild jaundice in a patient with
spleen and liver (extravascular
hereditary spherocytosis.
haemolysis).

The simple classification in Table 14.1 abnormalities including frontal bossing of


relies upon division of the main clinical the skull.
disorders into inherited and acquired Initial laboratory investigations of
types. In general, it can be seen that inher- haemolysis will include an automated
ited disorders are intrinsic to the red cell blood count, a blood film and a reticulo-
and acquired disorders extrinsic. The cyte count. The blood count will show low Fig 14.2  Hereditary spherocytosis.
inherited disorders can be subdivided haemoglobin. Many cases of haemolysis Spherocytes in a blood film.
depending on the site of the defect within have ‘normochromic normocytic’ red cell
the cell – in the membrane, in haemo- indices although some are moderately Red cell
globin, or in metabolic pathways. Acquired macrocytic. The latter observation is lysis (%)
disorders (discussed in the next section) caused by the increased number of large 100
are broadly divided depending on whether immature red cells (reticulocytes) in the
the aetiology has an immune basis. peripheral blood following a compensa-
tory increase in red cell production by the
Diagnosis of a bone marrow. Reticulocytes have a charac-
haemolytic anaemia teristic blue tinge with Romanowsky
Recognition of the general clinical and stains and their presence in the film
laboratory features of haemolysis usually causes ‘polychromasia’. A reticulocyte
precedes diagnosis of a particular clinical count is performed either manually on a
syndrome. Where haemolysis leads to blood film stained with a supravital stain
significant anaemia the resultant symp- or by the automated cell counter.
toms are as for other causes of anaemia. Simple laboratory tests to detect
Sodium chloride concentration
However, the increased red cell break- increased breakdown of red cells are also
down of the haemolytic anaemias causes useful indicators of haemolysis. In addi- Normal Curve in severe
range hereditary spherocytosis
an additional set of problems. Accelerated tion to moderately raised serum bilirubin
catabolism of haemoglobin releases (often 30–50 mol/L), there may be raised
Fig 14.3  Increased osmotic fragility in
hereditary spherocytosis. Spherocytes are
increased amounts of bilirubin into the levels of urine urobilinogen and faecal more fragile than normal red cells and lyse at
plasma such that patients may present stercobilinogen. Bilirubin itself is uncon- higher saline concentrations. The sensitivity of the
with jaundice (Fig 14.1). Where the spleen jugated and therefore does not appear in test is increased by incubating the cells at 37°C.
is a major site of red cell destruction there the urine. Haptoglobin, a glycoprotein
may be palpable splenomegaly. Severe bound to free haemoglobin in the plasma, Haemosiderin is present for several weeks
prolonged haemolytic anaemia in child- is depleted in haemolysis. In intravascular after a haemolytic episode and is simply
hood can lead to expansion of the haemolysis, haemoglobin and haemosi- demonstrated by staining urine sediment
marrow cavity and associated skeletal derin can be detected in the urine. for iron.
Haemolytic anaemia I – General features and inherited disorders 29

Examination of the bone marrow is not usually necessary in In patients with milder disease folate supplements are consid-
the work-up of haemolysis but, where performed, will show an ered but no other treatment is required. In more serious cases
increased number of immature erythroid cells. Formal demon- the spleen is removed. This should ideally be performed after 6
stration of reduced red cell survival by tagging of cells with radio- years of age with counselling regarding the infection risk.
active chromium (51Cr) and in vivo surface counting of
Hereditary elliptocytosis
radioactivity to identify the site of red cell destruction are other
This is generally a mild disorder with similarities to hereditary
possible investigations infrequently performed in practice.
spherocytosis. There is a variable deficiency of spectrin tetram-
ers. Red cells are elliptical except in the rare subtype hereditary
Inherited disorders
pyropoikilocytosis when they are more distorted and heat labile.
Disorders of the red cell membrane Splenectomy may be indicated for severe haemolysis.
Hereditary spherocytosis
Abnormalities of haemoglobin
This is the most common cause of inherited haemolytic disease
These disorders are referred to collectively as the ‘haemoglobin-
in northern Europeans. The disease is heterogeneous with a
opathies’. Thalassaemia and sickle cell syndromes are discussed
variable mode of inheritance. There are many possible gene
in later sections.
mutations with alterations in spectrin, ankyrin and other mem-
brane proteins. In a blood film the red cells are spheroidal Abnormalities of red cell metabolism
(‘spherocytes’) with a reduced diameter and more intense stain- The red cell has metabolic pathways to generate energy and also
ing than normal red cells (Fig 14.2). These abnormal red cells are to protect it from oxidant stress (Fig 14.4). Loss of activity of key
prone to premature destruction in the microvasculature of the enzymes may lead to premature destruction; there are two
spleen. common examples.
The severity of haemolysis is variable and the disease may
Glucose-6-phosphate dehydrogenase (G6PD) deficiency
present at any age. Fluctuating levels of jaundice and palpable
G6PD is a necessary enzyme in the generation of reduced glu-
splenomegaly are common features. Occasionally, patients
tathione which protects the red cell from oxidant stress. Defi-
develop severe anaemia associated with the transient marrow
ciency is X-linked, affecting males; female carriers show half
suppression of a viral infection; this so-called ‘aplastic crisis’,
normal G6PD levels. The disorder is most common in West
which may intervene in any form of chronic haemolysis, is often
Africa, southern Europe, the Middle East and South-East Asia.
caused by parvovirus B19. Prolonged haemolysis may lead to
Patients are usually asymptomatic until increased oxidant stress
bilirubin gallstones.
leads to a severe haemolytic anaemia, often with intravascular
Diagnosis is facilitated by the presence of a family history. The
destruction of red cells. Common triggers include fava beans,
combination of general features of haemolysis and spherocytes
drugs (many, including antimalarials and analgesics) and infec-
in the blood is suggestive of hereditary spherocytosis but not
tions. The disease can alternatively present as jaundice in the
diagnostic as spherocytes may also be seen in autoimmune
neonate. Diagnosis requires demonstration of the enzyme defi-
haemolysis. The two haemolytic disorders are distinguished by
ciency by direct assay – this should not be done during acute
the direct antiglobulin test, which is negative in hereditary sphe-
haemolysis as reticulocytes have higher enzyme levels than
rocytosis and nearly always positive in immune haemolysis.
mature red cells and a ‘false normal’ level may result. Treatment
Useful screening tests for hereditary spherocytosis include
is to stop any offending drug and to support the patient. Blood
measurement of osmotic fragility (Fig 14.3) and flow cytometric
transfusion may be necessary.
analysis of eosin-5-maleimide binding. In difficult cases, gel elec-
trophoretic analysis of red cell membranes is helpful. Pyruvate kinase (PK) deficiency
In this autosomal recessive disorder patients lack an enzyme in
O– H2O the Embden–Meyerhof pathway. Red cells are unable to generate
adequate ATP and become rigid. All general features of haemoly-
sis can be present, but clinical symptoms are often surprisingly
mild for the degree of anaemia as the block in metabolism leads
2GSH GSSG
to increased intracellular 2,3-DPG levels facilitating release of
Glucose oxygen by haemoglobin. Splenectomy may help in reducing
transfusion requirements.
NADP NADPH+H+

Glucose-6-P 6-PG Haemolytic anaemia I –


general features and
Glucose-6-phosphate
dehydrogenase inherited disorders
Fructose-6-P Ribulose 5-P
■ ‘Haemolytic anaemias’ are caused by abnormal destruction of red
cells.
■ Most inherited haemolytic disorders have a defect within the red
2,3.DPG Embden–Meyerhof
pathway cell while most acquired disorders have the defect outside the cell.
Pyruvate Hexose–monophosphate ■ Haemolysis causes characteristic clinical features and laboratory
kinase shunt abnormalities. It may be intra- or extravascular.
Rapoport–Luebering ■ Hereditaryspherocytosis and hereditary elliptocytosis are
Lactate shunt
haemolytic disorders caused by a deficiency in the red cell
Fig 14.4  Schematic diagram of red cell metabolism. This shows the membrane.
key roles of pyruvate kinase in the Embden–Meyerhof pathway (the cell’s ■ Glucose-6-phosphate dehydrogenase and pyruvate kinase are key
source of ATP) and glucose-6-phosphate dehydrogenase in the hexose- enzymes in red cell metabolism; inherited deficiency leads to
monophosphate shunt (the cell’s protection from oxidant stress). The broken haemolysis.
line represents several intermediate steps.
30 3 ANAEMIA

15 Haemolytic anaemia II – Acquired disorders


disorder. It is particularly important to
Autoimmune Table 15.1  Classification of the
autoimmune haemolytic anaemias stop an offending drug – cephalosporin
haemolytic anaemia
Warm AIHA (usually IgG) antibiotics are most commonly impli-
Autoimmune haemolytic anaemia Primary (idiopathic) cated. Where the haemolysis itself requires
(AIHA) is an example of an acquired form Secondary Lymphoproliferative treatment, steroids are normally used (e.g.
disorders
of haemolysis with a defect arising outside prednisolone 1–2 mg/kg daily). In idio-
Other neoplasms
the red cell. The bone marrow produces Connective tissue
pathic AIHA most patients will respond
structurally normal red cells and prema- disorders to steroids with a significant rise in hae-
ture destruction is caused by the pro- Drugs moglobin and diminished clinical symp-
duction of an aberrant autoantibody Infections toms. However, the disease is usually
targeted against one or more antigens on Cold AIHA (usually IgM) controlled rather than cured and relapses
the cell membrane. Once an antibody has Primary (cold often occur when steroids are reduced or
haemagglutinin
attached itself to the red cell, the exact stopped. Where refractoriness to steroids
disease)
nature of the haemolysis is determined Secondary Lymphoproliferative develops, splenectomy is usually indi-
by the class of antibody and the density disorders cated. Other immunosuppressive drugs
and distribution of surface antigens. Infections (e.g. (e.g. azathioprine, ciclosporin), intrave-
IgM autoantibodies cause destruction by mycoplasma) nous immunoglobulin, cytotoxic agents
Paroxysmal cold
agglutination or by direct activation of haemoglobinuria1
and the monoclonal antibody rituximab
serum complement. IgG class antibodies may all be helpful in difficult cases.
generally mediate destruction by binding 1
Caused by a biphasic polyclonal IgG antibody
(Donath–Landsteiner).
of the Fc portion of the cell-bound immu- Cold autoimmune
noglobulin molecule by macrophages in haemolytic anaemia
the spleen and liver. The disparate behav- In cold AIHA the antibody is generally of
iour of different types of autoantibody IgM type with specificity for the I red cell
provides the explanation for a number of antigen. It attaches best to red cells in the
different clinical syndromes. peripheral circulation where the blood
temperature is lower. As is seen in Table
Classification
15.1, this kind of haemolysis can occur in
Table 15.1 shows a simple approach to the
the context of a monoclonal (i.e. malig-
classification of autoimmune haemolytic
nant) proliferation of B-lymphocytes in
anaemia. The disease can be divided into
the so-called ‘idiopathic cold haemaggluti-
‘warm’ and ‘cold’ types depending on
nin syndrome’ or in a variety of lympho-
whether the antibody reacts better with
mas. The other major cause is infection.
red cells at 37°C or 0–5°C. For each of
Fig 15.1  Blood film in warm AIHA. The severity of haemolysis varies and
these two basic types of autoimmune Spherocytes and polychromasia are present. agglutination (clumping) of red cells (Fig
haemolysis there are a number of possible
15.3) may cause circulatory problems
causes and these can be incorporated into
such as acrocyanosis, Raynaud’s phenom-
the classification. A diagnosis of autoim-
enon and ulceration. The haemolysis,
mune haemolysis may precede diagnosis
where longstanding, is often worse in the
of the causative underlying disease.
winter. On occasion red cell destruction is
Clinical presentation and intravascular due to direct lysis by acti-
management vated complement. Where this occurs free
Warm autoimmune haemoglobin is released into the plasma
haemolytic anaemia (haemoglobinaemia) and may appear in
Warm AIHA (Figs 15.1 and 15.2) is the the urine (haemoglobinuria), giving it a
most common form of the disease. The dark colour. Cold AIHA arising from
red cells are coated with either IgG alone, infection is usually self-limiting. Where it
IgG and complement, or complement is chronic the mainstay of treatment is
alone. Premature destruction of these cells keeping the patient warm, particularly in
usually takes place in the reticuloendothe- the extremities. In forms associated with
lial system. Approximately half of all cases
Fig 15.2  Increased reticulocytes in warm lymphoproliferative disorders, cytotoxic
AIHA. The reticulocyte ribosomal RNA is stained
are idiopathic but in the other half there supravitally by brilliant cresyl blue. drugs (e.g. chlorambucil) or rituximab
is an apparent underlying cause (Table may be helpful. Steroids are generally
15.1). The autoantibody is produced by a frequent examination finding in severe ineffective.
polyclonal B-cells and is usually non- cases. The most characteristic laboratory
specific with reactivity against basic mem- abnormality in warm AIHA is a positive
brane constituents present on virtually all
Isoimmune
direct antiglobulin test (DAT) sometimes
red cells. Patients present with the clinical
haemolytic anaemia
known as the Coombs’ test (p. 83). A
and laboratory features of haemolysis dis- major priority in management is the iden- Here alloantibodies (isoantibodies) cause
cussed in the last section. Splenomegaly is tification and treatment of any causative haemolysis as a result of transfusion or
Haemolytic anaemia II – Acquired disorders 31

Table 15.2  Causes of microangiopathic


haemolytic anaemia
Haemolytic uraemic syndrome (HUS)1
Thrombotic thrombocytopenic purpura (TTP)1
Carcinomatosis
Vasculitis
Severe infections
Pre-eclampsia
Glomerulonephritis
Malignant hypertension

1
Some authorities believe that HUS and TTP are effectively a single
disorder TTP-HUS.

multimers which accumulate due to defi-


ciency of a protease (ADAMTS 13). Daily
Fig 15.3  Cold agglutination in the blood plasma exchange is the mainstay of treat- Fig 15.4  Blood film in microangiopathic
film of a patient with cold autoimmune ment; mortality rates are 10–20%. haemolytic anaemia. Fragmented red cells and
haemolytic anaemia. thrombocytopenia.
transfer across the placenta. These anti-
Other acquired
bodies are conventional antibodies spe-
haemolytic anaemias
cific for foreign antigens on incompatible
red cells. Haemolytic blood transfusion Haemolysis associated with red cell frag-
reactions are discussed on page 84 and mentation may also occur due to the
haemolytic disease of the newborn on mechanical effects of defective heart valves
page 90. or in long distance runners who effec-
tively stamp repeatedly on a hard surface
(‘march haemoglobinuria’). Certain drugs
Microangiopathic
(e.g. dapsone and sulfasalazine) can cause
haemolytic anaemia
oxidative intravascular haemolysis in
Collectively, microangiopathic haemolytic normal people if taken in sufficient
anaemia (MAHA) is one of the most fre- dosage. Many infections can cause haemo-
quent causes of haemolysis. The term lysis, either by direct invasion of red cells
describes intravascular destruction of red or via the circulatory changes already dis-
Fig 15.5  Haemosiderinuria caused by
cells in the presence of an abnormal cussed. The anaemia of malaria often has
chronic intravascular haemolysis in PNH
microcirculation. There are many causes a haemolytic component (pp. 98–99). (Perls’ reaction).
of MAHA (Table 15.2) but common trig- Paroxysmal nocturnal haemoglob-
gers are the presence of disseminated inuria (PNH) (Fig 15.5) is a rare example leukaemia. The traditional diagnostic test
intravascular coagulation (DIC), abnor- of acquired haemolysis caused by an exploited the cell’s unusual sensitivity to
mal platelet aggregation and vasculitis. intrinsic red cell defect. In this clonal dis- complement lysis (Ham test). Flow cytom-
Characteristic laboratory findings include order arising from a somatic mutation in etry is now used to show the cells’ charac-
red cell fragmentation in the blood film the PIG-A gene in a stem cell, the mature teristic lack of certain surface proteins
(Fig 15.4) and the coagulation changes blood cells have faulty anchoring of (CD55, CD59) and to quantitate the PNH
seen in DIC (see p. 79). Two specific syn- several proteins to membrane glycophos- clone. Management is largely supportive
dromes merit brief description. pholipids containing phosphatidylinosi- with blood transfusion and anticoagula-
tol. Clinical features are highly variable tion. More recently, eculizumab, a mono-
Haemolytic uraemic
and include intravascular haemolysis, clonal antibody blocking activation of
syndrome (HUS)
pancytopenia and recurrent thrombotic terminal complement, has been given to
HUS mainly affects infants and children.
episodes, including portal vein thrombo- reduce haemolysis and the risk of throm-
The three main features are MAHA, renal
sis. There is coexistent marrow damage bosis. Allogeneic stem cell transplantation
failure and thrombocytopenia. The
and PNH is often associated with aplastic is the only curative option but is used very
disease can occur as seasonal epidemics
anaemia and may even terminate in acute selectively (e.g. in severe marrow failure).
caused by Escherichia coli producing vero-
toxin; it is then preceded by bloody diar-
rhoea. Treatment is essentially supportive Haemolytic anaemia II –
with dialysis for renal failure. Mortality acquired disorders
ranges from 5 to 15%. ■ Autoimmune haemolytic anaemia (AIHA) can be divided into ‘warm’ and ‘cold’ types
dependent on the temperature at which the antibody reacts optimally with red cells.
Thrombotic thrombocytopenic
purpura (TTP) ■ For each type of AIHA there are possible underlying causes which must be identified and
This rare congenital or acquired disorder treated.
has many similarities to HUS. It is charac- ■ The term ‘microangiopathic haemolytic anaemia’ (MAHA) describes the intravascular
terised by MAHA, thrombocytopenia destruction of red cells in the presence of an abnormal microenvironment. Clinical
syndromes associated with MAHA include haemolytic uraemic syndrome and thrombotic
(often severe), fluctuating neurological
thrombocytopenic purpura.
symptoms, fever and renal failure. Platelet
■ Paroxysmal nocturnal haemoglobinuria (PNH) is a rare example of acquired haemolysis
microvascular thrombi are mediated
caused by an intrinsic red cell defect.
by ultra-large von Willebrand factor
32 3 ANAEMIA

16 The thalassaemias
The thalassaemias are a heterogeneous Table 16.1  Classification of thalassaemia but there is often a moderate chronic
group of inherited disorders of haemo- Type of Heterozygote Homozygote haemolytic anaemia (Hb 70–110 g/L)
globin synthesis. They are characterised thalassaemia with splenomegaly and sometimes
by a reduction in the rate of synthesis of α-Thalassaemia1 hepatomegaly. Severe bone changes and
either alpha or beta chains and are clas- α0 (– –/) Thal. minor Hydrops fetalis growth retardation are unusual. The
sified accordingly (i.e. α-thalassaemia, α+ (–α/) Thal. minor Thal. minor blood film shows hypochromic micro-
β-thalassaemia). The basic haematologi- β-Thalassaemia cytic red cells with poikilocytosis, poly-
cal abnormality in the thalassaemias is β0 Thal. minor Thal. major chromasia and target cells. The HbH
a hypochromic microcytic anaemia of β+ Thal. minor Thal. major or molecule is formed of unstable tetram-
variable severity. Unbalanced synthesis intermedia ers of unpaired β chains (β4). It is best
of α- and β-globin chains can damage 1
Compound heterozygosity (– –/–α) leads to HbH disease. detected by electrophoresis but may be
red cells in two ways. Firstly, failure of demonstrated as red cell inclusion
α and β chains to combine leads to β-Thalassaemias are autosomal reces- bodies in reticulocyte preparations.
diminished haemoglobinisation of red sive disorders characterised by reduced
α-Thalassaemia traits
cells to levels incompatible with survival. (β+) or absent (β0) production of β
Deletion of a single α-globin chain leads
Even those hypochromic cells released chains. The heterozygous (‘trait’ or
only to a slight lowering of red cell
into the circulation transport oxygen ‘minor’) form of the disease is usually
mean corpuscular volume (MCV) and
poorly. The second mechanism for symptomless while homozygosity is
mean corpuscular haemoglobin (MCH)
red cell damage is the aggregation of associated with the clinical disease
and even deletion of two genes usually
unmatched globin chains – the inclu- β-thalassaemia ‘major’. Homozygous
only minimally lowers the haemoglobin
sion bodies lead to accelerated apoptosis mild (β+) thalassaemia may, however,
with a raised red cell count and
of erythroid precursors in the bone lead to a less severe clinical syndrome
hypochromia and microcytosis. These
marrow (ineffective erythropoiesis) and termed ‘thalassaemia intermedia’. The
carrier states can be difficult to identify
destruction of more mature red cells in β-thalassaemias are very heterogeneous
in the routine laboratory as haemo-
the spleen (haemolysis). In general, at the molecular level – the large major-
globin electrophoresis is normal. Occa-
the clinical severity of any case of thalas- ity of mutations are single base substitu-
sional HbH bodies may be detected in
saemia is proportionate to the degree of tions (point mutations) and insertions
reticulocyte preparations. Definitive
imbalance of α- and β-globin chain or deletions of one to two bases.
diagnosis requires DNA analysis.
synthesis. Although molecular analysis may be
Thalassaemias are among the most needed, diagnosis of the major syn- β-Thalassaemias
common inherited disorders. Gene carri- dromes is normally possible from con- β-Thalassaemia major
ers have some protection from falci- sideration of the clinical features and The characteristic severe anaemia (Hb
parum malaria. Cases occur sporadically simple laboratory tests. The latter must less than 70 g/L) is caused by α-chain
in most populations but the highest tha- include a blood count and blood film, excess leading to ineffective erythropoi-
lassaemia gene frequency is in a broad and haemoglobin electrophoresis with esis and haemolysis. Anaemia first
geographical region extending from the quantification of the different types of becomes apparent at 3–6 months
Mediterranean through the Middle East haemoglobin (i.e. HbA, HbA2, HbF). when production of HbF declines. The
and India to South-East Asia. Other structural Hb variants may child fails to thrive and develops hepat-
coexist with thalassaemias giving rise to osplenomegaly. Compensatory expan-
a wide range of clinical disorders. Only sion of the marrow space causes the
Classification
the more common thalassaemia syn- typical facies with skull bossing and
The classification illustrated in Table dromes are discussed here. maxillary enlargement (Fig 16.1a). The
16.1 is based on the mode of inheritance ‘hair-on-end’ radiological appearance
of thalassaemia. of the skull (Fig 16.1b) is due to expan-
As the α-globin chain gene is dupli-
Clinical syndromes
sion of bone marrow into cortical bone.
cated on each chromosome there may α-Thalassaemias If left untreated further complications
be total loss of α-globin chain produc- Hb-Barts hydrops syndrome (– –/– –) can include repeated infections, bone
tion (termed α0 or – –/haplotype) or Here deletion of all four genes leads to fractures and leg ulcers. Red cell mem-
partial loss of α-chain production result- complete absence of α-chain synthesis. brane abnormalities contribute to
ing from loss of only one gene (termed As the α-globin chain is needed for fetal hypercoagulability.
α+ or –α/haplotype). haemoglobin (HbF) as well as adult hae- Laboratory testing should precede
The most important clinical syn- moglobin (HbA) (see p. 5) the disorder blood transfusion. There is a severe
dromes are haemoglobin (Hb)-Barts is incompatible with life and death hypochromic microcytic anaemia with
hydrops syndrome (– –/– –), which is occurs in utero (hydrops fetalis). a characteristic blood film (Fig 16.2)
incompatible with life, and Hb H disease and Hb electrophoresis demonstrates
(–α/– –). At the molecular level the HbH disease (–α/– –) absence or near absence of HbA with
α-thalassaemias result from loss of α- This disorder arises from deletion of small amounts of HbA2 and the remain-
gene function due to gene deletion three of the four α-globin genes and is der HbF (Fig 16.3).
or non-deletional mutations; different found most commonly in South-East With intense supportive therapy,
types of mutations may be co-inherited. Asia. The clinical features are variable increasing numbers of patients in the
The thalassaemias 33

Fig 16.2  Blood film in β-thalassaemia major.


(a) (b) Table 16.2  Possible causes of thalassaemia
intermedia
Fig 16.1  β-Thalassaemia major. (a) Typical facies; (b) skull X-ray showing ‘hair-on-end’ appearance. ■ Mild defects of β-globin chain production, e.g.
homozygous mild β+-thalassaemia
Hb type
■ Homozygosity or compound heterozygosity for
severe β-thalassaemia with co-inheritance of
H
α-thalassaemia or genetic factors enhancing γ-chain
A production
■ Heterozygous β-thalassaemia with co-inheritance of

F additional α-globin gene


■ δβ-thalassaemia and hereditary persistence of fetal
haemoglobin
■ HbH disease

β-Thalassaemia trait (minor)


A2
Heterozygotes for β0 or β+ are usually
asymptomatic with hypochromic micro-
cytic red cells and slightly reduced hae-
Normal b thal trait b thal major HbH disease
moglobin levels. The red cell count is
Fig 16.3  Haemoglobin electrophoresis (cellulose acetate, pH 8.5). The patterns obtained in elevated. The key diagnostic feature is a
normality and some common thalassaemia syndromes are shown. Other screening methods include
raised HbA2 level (4–7%). The disorder
high-performance liquid chromatography and isoelectric focusing.
may be confused with iron deficiency
developed world survive into adulthood. production (e.g. erythropoietin, hydroxy- leading to unnecessary investigations. If
Blood transfusion remains the mainstay carbamide) and gene therapy (see p. 103). both parents have β-thalassaemia trait
of management. Raising the haemo- there is a 25% chance of a child having
globin concentration both reduces tissue Thalassaemia intermedia β-thalassaemia major.
hypoxia and suppresses endogenous Thalassaemia intermedia is a clinical
haematopoiesis which is largely ineffec- syndrome which may result from a
Prenatal diagnosis
tive. There is improved growth and variety of genetic abnormalities (Table
development and reduced hepat- 16.2). The clinical features are less severe This depends on early identification of
osplenomegaly. Transfusion is generally than in β-thalassaemia major as the α/β- couples at risk and sensitive counselling.
given to maintain a haemoglobin level globin chain imbalance is less pro- Adequate amounts of fetal DNA can be
of at least 90–100 g/L. Splenectomy can nounced. Patients usually present later obtained around the 10th week of gesta-
reduce the transfusion frequency. With than is the case for β-thalassaemia major tion by chorionic villus sampling.
such regular transfusion iron chelation (often at 2–4 years), and have relatively Current technologies allow reliable
is necessary to minimise iron overload. high haemoglobin levels (80–100 g/L), identification of single point mutations
Without chelation, accumulation of iron moderate bone changes and normal from very small DNA samples. Tech-
damages the liver, endocrine organs and growth. Transfusion may be required niques are being developed to analyse
heart with death in the second or third but requirements are less than in fetal DNA obtained from maternal
decades. The most commonly used β-thalassaemia major. plasma or peripheral blood.
regimen is subcutaneous desferrioxam-
ine given for 5–7 days per week. Compli-
ance may be problematic (especially in The thalassaemias
teenagers) but where good there is a
■ The thalassaemias are a heterogeneous group of inherited disorders where there is a
considerably improved life expectancy. reduction in the rate of synthesis of haemoglobin α chains (α-thalassaemia) or β chains
Oral iron chelators (e.g. deferiprone, (β-thalassaemia).
deferasirox) are emerging as an accept- ■ Theremay be both ineffective erythropoiesis and haemolysis. The basic haematological
able alternative. Endocrine disturbances abnormality is a hypochromic microcytic anaemia.
related to iron overload will require ■ There are several clinical syndromes. In general the severity is proportionate to the degree
appropriate therapy. of imbalance of α- and β-globin chains.
Allogeneic stem cell transplantation
■ β-Thalassaemia major leads to severe anaemia requiring regular blood transfusion and iron
is a serious option. In ‘best risk’ patients chelation.
the probability of survival exceeds 90%.
■ Thalassaemia trait is a symptomless clinical disorder which should not be confused with
Experimental therapies include drugs iron deficiency. Genetic counselling is required in selected cases.
designed to stimulate fetal haemoglobin
34 3 ANAEMIA

17 Sickle cell syndromes


The sickle cell syndromes are a group of 1. Both parents have sickle trait 2. One parent has sickle trait and the other
haemoglobinopathies which primarily is heterozygous for HbC
affect the Afro-Caribbean population. S A S A S A C A
The common feature of these diseases
is inheritance of an abnormal haemo-
globin β-chain gene – the gene is desig-
nated βS. Inheritance of two βS genes
leads to a serious disorder termed sickle
cell anaemia. A similar syndrome can
result from inheritance of the βS gene
with another abnormal β gene such A A S A S S A A S A S C
as the haemoglobin C gene or
Unaffected Sickle trait Sickle cell Haemoglobin
β-thalassaemia gene. Inheritance of the (AS) anaemia (SS) SC disease
βS gene with a normal β-chain gene (βA)
causes the innocuous sickle cell trait Fig 17.1  Inheritance of sickle cell syndromes. Two pedigrees showing inheritance of sickle cell
(Fig 17.1). syndromes. In the first family one child is unaffected, one has sickle cell trait and one has sickle cell
anaemia. In the second family one child has inherited the abnormal sickle gene and the HbC gene; this
double heterozygosity leads to haemoglobin SC disease.
Pathophysiology
The abnormal βS gene has a high inci-
dence in tropical and subtropical regions
as the abnormal haemoglobin produced
(HbS) gives some protection against fal-
ciparum malaria. HbS differs from
normal haemoglobin (HbA) in that
glutamic acid has been replaced by
valine at the sixth amino acid from the
N-terminus of the β-globin chain. The
clinical features of sickle cell anaemia Fig 17.3  Dactylitis in sickle cell anaemia.
arise from the propensity of red cells Fig 17.2  Blood film in sickle cell anaemia. (Reproduced with permission from Linch D C,
containing haemoglobin S to undergo Yates A P 1996 Colour Guide Haematology
‘sickling’. In the deoxygenated state HbS Vascular-occlusive crises Churchill Livingstone, Edinburgh.)
undergoes a conformational change Acute, episodic, painful crises are a
leading to the creation of haemoglobin potentially disabling feature of sickle supply, and hepatic and splenic
tetramers which aggregate to produce cell anaemia. They may be triggered by sequestration.
large polymers. The red cell loses its infection or cold. Patients complain of
normal deformability and becomes musculoskeletal pain which may be Other complications
characteristically sickle-shaped (Fig 17.2). severe and require hospital admission. These are multiple, usually caused by
Damage to the membrane leads to Hips, shoulders and vertebrae are most vascular stasis and local ischaemia.
increased rigidity and the ultimate affected. Attacks are generally self-
■ Genitourinary. Papillary necrosis with
sequestration of the red cell in the retic- limiting but infarction of bone can occur
uloendothelial system causing haemo- haematuria; loss of ability to
and must be distinguished from salmo-
lytic anaemia. The inflexible sickle cells concentrate urine; nephrotic
nella osteomyelitis. Avascular necrosis
also become lodged in the microcircula- syndrome; priapism.
of the femoral head is a crippling com-
■ Skin. Lower limb ulceration.
tion causing stasis and obstruction. plication. Other organs are vulnerable to
■ Eyes. Proliferative retinopathy;
infarction; most serious is neurological
glaucoma.
Clinical syndromes damage which may manifest as seizures,
■ Hepatobiliary. Liver damage; pigment
transient ischaemic attacks (TIAs) and
Sickle cell anaemia (HbSS) gallstones.
strokes. Vaso-occlusion in infancy is
This classic form of sickle cell syndrome
responsible for the ‘hand–foot syn-
is enormously variable in severity. Diagnosis
drome’, a type of dactylitis damaging the
Diagnosis depends on the following:
Haemolytic anaemia small bones of hands and feet (Fig 17.3).
The haemoglobin is generally in the ■ Blood film appearance (see Fig 17.2).
range 60–100 g/L. Because HbS releases Sequestration crises ■ Screening tests for sickling. The blood
oxygen more readily than HbA, the These arise from sickling and infarction sample is deoxygenated (e.g. with
symptoms of anaemia are often surpris- within particular organs. Specific syn- sodium metabisulphate) to induce
ingly mild. Intercurrent infection with dromes include ‘acute chest syndrome’ sickling.
parvovirus or folate deficiency can block with occlusion of the pulmonary vascu- ■ Haemoglobin electrophoresis. In sickle

erythropoiesis and cause a sudden fall lature, ‘girdle sequestration’ caused by cell anaemia (HbSS) there is no HbA
in haemoglobin – the ‘aplastic crisis’. occlusion of the mesenteric blood detectable (Fig 17.4).
Sickle cell syndromes 35

Management needed for severe anaemia or other or β-thalassaemia. HbSC disease is


General. Patients need support in the sickle-cell-related complications. During similar to HbSS but there is a tendency
community and easy access to centres surgery it is important to avoid hypoxia for fewer painful crises and a higher
experienced in the management of and dehydration. Preoperative simple incidence of proliferative retinopathy
sickle cell anaemia. Prophylaxis is transfusion or even exchange transfu- and avascular necrosis. The severity of
important. Patients should avoid factors sion may be appropriate for high-risk sickle cell/β-thalassaemia depends on
known to precipitate crises, take folate procedures. whether patients have the β+ or β0 geno-
supplements (because of chronic Hydroxycarbamide. Increasing the type. The less common HbS/β0 form has
haemolysis) and be prescribed penicillin level of fetal haemoglobin in red cells a similar clinical picture to HbSS.
and pneumococcal vaccine (because of with the antimetabolite hydroxycar-
hyposplenism caused by infarction). bamide can reduce the severity of the Sickle cell trait (HbAS)
Infections require prompt treatment. disease. Clinical trials have been encour- Sickle cell trait normally causes no clini-
Transcranial Doppler ultrasonography aging with significant reductions in cal problems as there is enough HbA in
can identify children at high risk of painful crises, major complications, red cells (approximately 60%) to prevent
stroke. Annual retinal screening is blood transfusion, hospital admissions sickling. However, haematuria occasion-
recommended. and mortality rates. It is important to ally occurs as a result of renal papillary
Painful vascular-occlusive crises. ensure compliance as clinical benefit necrosis and additional care is required
First-line treatment is rest, increased may not be immediate. There are con- during pregnancy and anaesthesia. Diag-
fluids and adequate oral analgesia. Con- cerns regarding the leukaemogenic and nosis is by a sickling test and Hb elec-
stitutional upset or pain not relieved by teratogenic effects of hydroxycarbamide trophoresis (see Fig 17.4). Life expectancy
oral analgesia necessitates hospital but with cautious use and patient educa- is normal although there may be a
admission with continued rest, warmth, tion (e.g. appropriate contraception) the slightly increased risk of sudden death
intravenous fluids and opiate analgesia. risks appear to be low. during intense exercise in young adults.
Psychological support is vital. Stem cell transplantation. Alloge-
Blood transfusion. Acute or chronic neic stem cell transplantation offers the
simple red cell transfusion may be given
Counselling and
possibility of a cure in selected high risk
to relieve severe anaemia and to reduce
prenatal diagnosis
patients but it will not be more widely
the amount of circulating sickle haemo- applicable until the toxicity is reduced Genetic counselling is needed by those
globin. Chronic transfusions are the (see p. 56). affected with either the homozygous
most effective intervention to prevent Gene therapy. Gene therapy has the disease, compound heterozygosity or
recurrent cerebrovascular events. Other potential to provide a cure without the the trait. Prenatal diagnosis is possible
indications for transfusion include com- risks of allogeneic stem cell transplanta- using mutation analysis on PCR-
plications such as chest syndrome and tion (see p. 103). amplified DNA from chorionic villi (see
priapism. Blood transfusion is not p. 100).
usually required for episodes of pain. Prognosis
Exchange transfusion is preferred to The risk of early death is inversely
simple transfusion for rapid reduction
Screening strategies
related to fetal haemoglobin levels. The
of HbS levels or where simple transfu- most common causes of death are infec- Screening of all newborn babies for
sion would cause hyperviscosity or cir- tion in infancy, cerebrovascular acci- sickle cell syndromes is recommended
culatory overload. Blood is phenotypically dents in childhood and adolescence and to reduce the risk of early death from
matched to reduce the chance of alloim- respiratory complications in adult life. infection. Preconception testing and
munisation. Iron chelation may be antenatal testing of pregnant women is
required. Doubly heterozygous sickling performed depending on individual risk
Pregnancy and surgery. Transfu- disorders and the local prevalence of sickle cell
sion is not routinely indicated in an Here patients inherit the βS gene and disease. A similar approach is adopted
uncomplicated pregnancy but may be another abnormal β gene – usually HbC prior to surgery.

Sickle cell syndromes


■ The sickle cell syndromes are a group of haemoglobinopathies
which primarily affect people of African origin.
■ Inheritance of two βS genes leads to the serious clinical disorder
A sickle cell anaemia (HbSS).
■ Clinical
problems in sickle cell anaemia include chronic haemolytic
F anaemia, vascular-occlusive crises, sequestration crises and
S susceptibility to infection.
■ Routine management of sickle cell anaemia entails prophylactic
A2/C measures, supportive care during vascular-occlusive crises and the
selective use of blood transfusion and hydroxycarbamide.
■ Sickle
cell trait (HbAS) is an innocuous clinical disorder but genetic
counselling is often needed.
Fig 17.4  Cellulose acetate electrophoresis to separate
haemoglobins A, F, S and C. Lane 4, control sample; Lanes 2, 3, 6, 7,
normal; Lane 1, sickle cell anaemia; Lane 5, sickle cell trait.
36 3 ANAEMIA

18 Anaemia of chronic disease


Anaemia of chronic disease (ACD) is a questions remain. Key factors in aetiol- reticuloendothelial system limits its
term used to describe a type of anaemia ogy are summarised in Figure 18.2. availability to microorganisms or
seen in a wide range of chronic inflam- Inflammatory cytokines such as tissue tumour cells. Decreased haemoglobin
matory, infective and malignant diseases necrosis factor (TNF) and interleukin-1 levels reduce the oxygen-carrying capac-
(Table 18.1). The anaemia often becomes and -6 are implicated in all of these ity of the blood and might reduce the
apparent during the first few months of processes. oxygen supply to unwelcome microor-
illness and then remains fairly constant There is a modest shortening of red ganisms and cells. Cell-mediated immu-
(Fig 18.1). It is rarely severe (haemo- cell lifespan which leads to an increased nity is probably strengthened by reduced
globin ≥90 g/L; packed cell volume demand for bone marrow production. levels of metabolically active iron in the
(PCV) ≥0.30) but there is some correla- The marrow struggles to respond ade- circulation as iron inhibits the activity of
tion with the intensity of the underlying quately as there is blunting of the IFN-γ.
illness. For instance, in infection the expected increase in erythropoietin
anaemia is often more marked where secretion and also diminished respon-
Diagnosis
there is a persistent fever and in malig- siveness of erythroid precursor cells to
nancy where there is widespread dis- erythropoietin. Hepcidin, a peptide Most patients will have a documented
semination. Patients may suffer no hormone, appears to be an important chronic disorder and a moderate
symptoms from their anaemia or have mediator of ACD. This acute phase reac- anaemia. On occasion the anaemia is a
only slight fatigue. The importance of tant protein is released from the liver more dominant feature and the underly-
this type of anaemia arises not from its following stimulation by interleukin-6. ing cause is not immediately apparent.
severity but from its ubiquity. It is widely Actions of hepcidin include inhibition of The anaemia is usually of normochro-
misunderstood (for such a common dis- microbial infection, macrophage iron mic normocytic type although it can be
order) and ill patients are frequently recycling and intestinal iron absorption. slightly hypochromic microcytic. The
subjected to excessive haematological Its role in iron balance and transport is blood film appearance is often unre-
investigation and unnecessary treat- mediated via binding to ferroportin, the markable but there may be changes
ment with haematinics. The term ACD major cellular iron efflux protein. ‘reactive’ to the underlying disorder such
should not be used to describe other Patients with inflammation and anaemia as a neutrophil leucocytosis, thrombocy-
causes of anaemia such as haemolysis or have elevated serum and urine levels of tosis and rouleaux formation. There
bleeding which may also complicate hepcidin. Abnormalities of iron metabo- is a reticulocytopenia. Inflammatory
chronic disorders. It has been argued lism are well documented in ACD. markers such as C-reactive protein
that the designation ACD is inappropri- These include: (CRP) and erythrocyte sedimentation
ate but other suggested terms (e.g. rate (ESR) are often raised. Serum iron
■ reduced iron absorption from the
anaemia of inflammation) appear even concentration and transferrin concen-
gastrointestinal tract
less satisfactory. The anaemia of chronic tration are usually reduced. The serum
■ decreased plasma iron concentration
renal failure is variously referred to as ferritin level is normal or high (as an
■ excessive retention of iron in
ACD although it has its own specific acute phase reactant). In practice, ACD
reticuloendothelial cells
features (see p. 96). is most commonly confused with mild
(macrophages) with diminished
iron deficiency anaemia, particularly if
release to erythroid cells.
the MCV and MCH are reduced.
Incidence The high prevalence of ACD has However, the two forms of anaemia
Because its causes are common, ACD is led to the suggestion that it may have should be distinguishable as in uncom-
probably only second to iron deficiency some benefits for those with chronic plicated iron deficiency the transferrin
as a cause of anaemia. It has been esti- inflammation. Perhaps withdrawal of concentration is elevated and the ferritin
mated to account for approximately half iron by increased storage in the level is low. In difficult cases the serum
of all hospital cases of anaemia not
explained by blood loss.
Haemoglobin (g/L)

120
Pathophysiology
The causation of the anaemia of chronic 100
disease has been extensively studied but
80

Table 18.1  Common causes of the anaemia


60
of chronic disease
■ Malignancy
■ Rheumatoid arthritis 40
■ Various connective tissue disorders
■ Chronic infection
Fig 18.1  ACD in a
20 patient with chronic
■ Extensive trauma
1
■ Chronic renal failure
infection. The rate of
■ Chronic heart failure 0 development of anaemia
0 3 6 9 12 and its final severity are
1
See also page 96. Months since onset of infection typical of ACD.
Anaemia of chronic disease 37

Fig 18.2  Overview of the aetiology of


Blunted response to Inhibition of red cell ACD. Cytokines such as TNF, interleukin-1
erythropoietin precursors and interleukin-6 and the peptide hepcidin
play key roles (see text).

Impaired red cell production Impaired iron mobilisation


in marrow and utilisation

ACD Reduced red cell


survival

Table 18.2  Comparison of clinical and laboratory findings in ACD


and iron deficiency anaemia
Characteristic ACD Iron deficiency
Severity of anaemia Hb usually ≥90 g/L Very variable
Symptoms of anaemia Usually mild May be severe
Coexistent chronic disease Yes Variable
Red cell indices (MCV, MCH) Normochromic Hypochromic
Normocytic1 Microcytic
Blood film appearance Often normal or reactive2 Hypochromia
Microcytosis
Poikilocytosis
Target cells
Serum iron Reduced Reduced Fig 18.3  Bone marrow aspirate stained with Perls stain showing
Transferrin concentration Reduced or normal Increased increased reticuloendothelial iron stores in ACD.
Ferritin Normal or increased Reduced3
Serum transferrin receptor Normal Increased
Serum transferrin Low High
receptor-ferritin index4 Occasionally, patients cannot adequately compensate for
Marrow iron stores Normal or increased Reduced the anaemia and require blood transfusion. Recombinant
human erythropoietin and its derivatives can be effective in
1
May be slightly hypochromic microcytic.
2
relieving anaemia, particularly in rheumatoid arthritis and
‘Reactive’ changes in a blood film may accompany the underlying disorder; possible abnormalities
include rouleaux formation, a neutrophil leucocytosis and thrombocytosis. malignancy. Their use is restricted by concerns of increased
risk of thromboembolic events and higher rates of tumour
3
Unless there is a coexistent acute phase response when the ferritin level may be normal.
4
Transferrin receptor concentration divided by serum ferritin concentration (or log of plasma ferritin
concentration). recurrence; they should only be used selectively and in the
lowest effective dose. In the absence of coexistent iron defi-
transferrin receptor concentration and the serum transferrin ciency, oral iron supplements are rarely helpful. Intravenous
receptor-ferritin index may be useful (Table 18.2). Measure- iron may be beneficial, especially if combined with erythro-
ment of the percentage of hypochromic red cells or reticulo- poietin, but there is limited experience outside the field of
cyte haemoglobin content can be helpful in detecting renal medicine. Further studies are needed to evaluate the
coexistent iron-restricted red cell production in a patient with effect of amelioration of the anaemia on the course of the
ACD. Reliable hepcidin assays are under development and are underlying disease. Possible future therapies for ACD include
likely to enter routine clinical practice. Bone marrow examina- alternative stimulators of erythropoiesis, hepcidin antagonists
tion is not routinely required but where performed will show and novel anti-inflammatory agents.
normal or increased marrow iron stores with decreased
marrow sideroblasts (Fig 18.3).
It should be remembered that anaemia in a patient with a
chronic medical disorder may be of multifactorial origin. It is Anaemia of chronic disease
important not to misdiagnose ACD as something else but (ACD)
equally it cannot be assumed that every patient with long-
standing disease and a low haemoglobin has only ACD. ■ ACD is seen in a wide range of chronic malignant, inflammatory
and infective disorders.
■ The pathogenesis of ACD is complex. There is a reduction in both
Management red cell production and survival. Hepcidin is likely to be a key
mediator.
As the anaemia is usually non-severe and not progressive, the
■ The anaemia is usually of normochromic, normocytic type,
management is primarily that of the underlying disorder. The
non-progressive and is rarely severe.
rationale for treating the anaemia itself is to avoid immediate
■ Treatment is primarily that of the underlying disorder. Blood
deleterious effects (e.g. on the cardiovascular system), to
transfusion and erythropoietin may help in selected cases. Iron
improve quality of life, and possibly to improve the prognosis
supplementation has a limited role.
of the underlying condition.
38 4 LEUKAEMIA

19 Introduction
The leukaemias are a heterogeneous group of malignant incompletely understood, this protein causes deregulated
blood disorders. In this introductory section, general charac- myeloid cell growth.
teristics such as definitions, aetiology and classification are
discussed. Each of the more common types of leukaemia is Chromosome deletions and additions
subsequently described in more detail. A chromosome may be completely or partly deleted, for
example monosomy 7 in acute myeloid leukaemia (AML).
Here a normal gene may be lost, allowing expression
Definition
of a recessive cancer gene. Conversely, an additional chromo-
Leukaemia is a type of cancer caused by the unregulated some may be gained.
proliferation of a clone of immature blood cells derived from
mutant haematopoietic stem cells. The disease is the result of Submicroscopic mutations
multiple acquired genetic and epigenetic events which can A change in the base sequence of certain oncogenes may
vary widely between patients. Leukaemic transformation is predispose to leukaemia. The RAS oncogene which encodes
assumed to occur at or near the level of the leukaemic stem a protein vital in signal transduction is mutated in 50% of
cell prior to definite lineage commitment. The leukaemic cells cases of AML.
do not differentiate normally. They may avoid standard mech-
anisms of cell death (apoptosis) and they may also retain the Epigenetic mechanisms
stem cell signature of self-renewal. This relentless proliferat- Epigenetic changes, where there is a change in gene function
ing clone of aberrant cells eventually squeezes out normal (e.g. altered DNA methylation) but not structure, may play a
cells from the bone marrow causing marrow failure and role in leukaemia.
death.

Lung 157,000
Incidence
Leukaemia is not a common disorder but it is a significant Colorectal 49,000
cause of death from cancer (Fig 19.1). There is a male prepon-
derance in most types of leukaemia. Geographic variations Breast 40,000
exist; for instance, chronic lymphocytic leukaemia is the pre-
Pancreas 37,000
dominant form of leukaemia in the Western world but is
much less frequent in Japan, South America and Africa.
Prostate 34,000

Leukaemia 22,000
Aetiology
As for other malignancies, the evolution of leukaemia is likely Non-Hodgkin’s 19,000
to be a multistep process. Thus, accumulated genetic muta- lymphoma
tions corrupt normal cellular pathways controlling prolifera- 0 100000
tion and differentiation and lead to the production of an Annual deaths
autonomous proliferating stem cell clone (‘clonal evolution’). Fig 19.1  Annual causes of death from malignancy in the year 2011
It is easiest to think about the aetiology in terms of these (estimated data from United States).
acquired genetic abnormalities and other more general pre-
disposing factors.

Genetic abnormalities
Cytogenetic analysis and particularly molecular genetic tech-
niques have revealed various acquired non-random chromo-
somal derangements which play a fundamental role in
leukaemogenesis (Fig 19.2). There are a number of different
types of possible chromosomal change.

Chromosomal translocations
One chromosome breaks and donates a fragment to another
chromosome which reciprocates by returning a fragment of
its own. Such translocations can result in the movement of
proto-oncogenes to new sites where they have the capacity to
cause leukaemic transformations. The classical example of a
balanced translocation is the ‘Philadelphia chromosome’,
found in 95% of cases of chronic myeloid leukaemia (CML),
where breakages in chromosomes 9 and 22 result in the crea- Fig 19.2  Fluorescence in situ hybridisation (FISH) study of a
tion of a new fusion gene (BCR-ABL) which encodes a novel complex karyotype (including t(8;16) ) in a patient with acute
protein with intense tyrosine kinase activity. In a manner myeloid leukaemia.
Introduction 39

Table 19.1  Factors predisposing


to leukaemia
Radiation exposure
Previous chemotherapy (particularly alkylating agents)
Occupational chemical exposure (e.g. benzene)
Some genetically determined disorders (e.g. Down
syndrome)
Viral infection (only HTLV-1 proven as a causative
factor)
Myelodysplastic and myeloproliferative disorders
Other possible (e.g. cigarette smoking)

Particular chromosome changes are


often associated with specific types of
leukaemia (e.g. the Philadelphia chro-
mosome in CML). However, few abnor-
malities are entirely specific – the
Philadelphia chromosome can be found
in cases of acute leukaemia. It should Fig 19.3  Peripheral blood film in a young Fig 19.4  Bone marrow trephine appearance
also be noted that not all cases of leu- woman with acute myeloid leukaemia. in human T-cell leukaemia lymphoma. The
She had received chemotherapy for only human leukaemia with a known viral
kaemia have a detectable cytogenetic
choriocarcinoma several years previously. causation.
abnormality. The incidence of abnor-
mality is partly dependent on the labora- patients also treated with radiotherapy. leukaemias into their cell of origin (i.e.
tory expertise available. The best established occupational leu- myeloid or lymphoid) and refers to the
kaemogenic exposure is undoubtedly to microscopic appearance (morphology)
Predisposing factors benzene. A number of genetically deter- of the leukaemic cells. The traditional
In a small subpopulation of leukaemic mined diseases also predispose to leu- classification of the acute leukaemias is
patients there is another obvious predis- kaemia. Here the liability to leukaemia that of the FAB group – the abbreviation
posing factor – the more common of is probably caused by factors such as being for the French, American and
these are listed in Table 19.1. increased chromosomal breakage (e.g. British nationalities of the terminolo-
The incidence of acute leukaemia and Fanconi’s anaemia) and immunosup- gists – but this has been overtaken by
chronic myeloid leukaemia increases pression (e.g. ataxia telangiectasia). the World Health Organization (WHO)
with radiation dose exposure in all age Viruses are known to be the main system. Basic morphological techniques
groups. Classic studies have included cause of leukaemia in many animals but (e.g. microscopic inspection of a blood
people exposed to the atomic bombs in in humans the only well-proven associa- film and bone marrow sample) remain
Japan and patients receiving radiother- tion is of the HTLV-1 virus with the rare important in the initial diagnosis but
apy for ankylosing spondylitis in the disorder T-cell leukaemia lymphoma cytogenetic and molecular genetic tech-
middle years of the 20th century. Results (Fig 19.4). Myelodysplastic syndromes niques are becoming increasingly
from studies of diagnostic radiation and (pp. 50–51) and myeloproliferative dis- important in classification as acquired
adult leukaemia are inconsistent and in orders (pp. 64–67) may transform to genetic changes frequently have prog-
appropriate radiological procedures acute myeloid leukaemia. nostic significance and can guide
the benefit is likely to outweigh what treatment.
appears to be at most a very small risk. In the following pages are discussed
Classification acute myeloid leukaemia, acute lym-
Paternal preconception exposure to ion-
ising radiation has been associated with In such a potentially complex group of phoblastic leukaemia, chronic myeloid
an increased incidence of acute leukae- disorders it is helpful to use a relatively leukaemia and chronic lymphocytic leu-
mia in offspring. simple classification. The leukaemias kaemia. Together these four diseases
Cytotoxic chemotherapy, particularly can most broadly be divided into acute constitute the overwhelming majority of
with alkylating agents, leads to an and chronic types depending on their leukaemias in clinical practice. A few
increased risk of leukaemia (Fig 19.3). clinical course. The classification illus- rarer types of leukaemia are discussed
The risk appears to be greatest in older trated here (Table 19.2) further divides separately.

Leukaemia: introduction
■ Leukaemia is a type of cancer caused by the unregulated
Table 19.2  Classification of leukaemia1 proliferation of a clone of immature blood cells.
Acute leukaemia Acute myeloid leukaemia ■ Leukaemia is a heterogeneous group of clinical disorders classified
Acute lymphoblastic leukaemia on the basis of their clinical course (acute or chronic) and their cell
Chronic leukaemia Chronic myeloid leukaemia of origin (myeloid or lymphoid).
Chronic lymphocytic leukaemia ■ The aetiology of leukaemia is likely to be multifactorial with known
Other types Hairy cell leukaemia predisposing factors such as radiation exposure present in only a
Prolymphocytic leukaemia minority of cases. Acquired genetic and epigenetic abnormalities
T-cell leukaemia lymphoma play a fundamental role in leukaemogenesis with certain changes
associated with particular types of leukaemia.
1
See specific disease sections for more detail.
40 4 LEUKAEMIA

20 Acute myeloid leukaemia

Introduction Table 20.1  WHO classification of acute


myeloid leukaemia
Acute myeloid leukaemia (AML) is a AML with recurrent genetic abnormalities1
malignant clonal disorder of haemat- AML with t(18;21)(q22;q22)
opoietic progenitor cells. Leukaemic AML with inv (16)(p13;q22) or t(16;16) (p13.1;q22) (M4
Eo)
transformation usually occurs at a very
AML with t(15;17)(q22;q12) (M3:M3V)
early stage of myeloid development, AML with t(9;11) (p22;q23)
probably at or near the haematopoietic AML with t(6;9) (p23;q34)
stem cell, but it may develop in a more AML with inv(3) (q21;q26.2) or t(3;3) (q21;q26.2)
mature cell. The disease is heterogene- AML (megakaryoblastic) with t(1;22) (p13;q13)
AML with myelodysplasia-related changes Fig 20.1  Gum infiltration in acute
ous. The malignant cells acquire genetic
Therapy related myeloid neoplasms monocytic (M5) leukaemia.
alterations which upset their normal
AML not otherwise specified2
mechanisms of self-renewal, prolifera-
AML with minimal differentiation (M0)
tion and differentiation. Ultimately the AML without maturation (M1)
malignant clone causes marrow failure. AML with maturation (M2)
AML is rare in childhood and the inci- Acute myelomonocytic leukaemia (M4)
dence increases with age; two-thirds of Acute monoblastic/ monocytic leukaemia(M4/5)
Acute erythroid leukaemia (M6)
cases occur in people aged over 60 years.
Acute megakaryoblastic leukaemia (M7)
Acute basophilic leukaemia
Acute panmyelosis with myelofibrosis
Classification Myeloid sarcoma (a)
Myeloid proliferation related to Down syndrome
The WHO system has now largely
Blastic plasmacytoid dendritic cell neoplasm
superseded the French-American-British
(FAB) classification. The newer classifica- 1
See also Table 2 for genetic changes.
2
FAB equivalent is shown in parentheses.
tion reduces the bone marrow leukae-
mic blast cell percentage differentiating
■ AML with t(15;17)(q22;q12) (M3,
AML from myelodysplastic syndrome
M3V). More traditionally referred to
(see p. 50) from 30% to 20%. Other key
as acute promyelocytic leukaemia, this
changes include the creation of specific
disease is associated with a high
subtypes with non-random cytogenetic (b)
incidence of disseminated
or equivalent molecular abnormalities,
intravascular coagulation (DIC) and a
and the distinction of patients with mul-
high risk of spontaneous bleeding
tilineage dysplasia and also previous
into vital organs.
chemotherapy. The major FAB subtypes
are included in the ‘other’ category with Tissue infiltration is more common in
the exception of acute promyelocytic subtypes with monocytic morphology
leukaemia (previously FAB M3) which is and immunophenotypic features (i.e.
now in the ‘recurrent translocations’ FAB M5) – patients often present with
group due to the inevitable presence of gum infiltration (Fig 20.1), lymphaden- (c)
t(15;17). It can be seen (Table 20.1) that opathy, skin deposits and hepatosplenom-
occasional cases of AML show meg- egaly. Central nervous system (CNS) Fig 20.2  Bone marrow appearance in
different FAB subtypes of AML. (a) AML M2:
akaryocytic or erythroid differentiation. disease is rare in AML but most frequent
the leukaemic blast cells show some granulocytic
Gene mutations are likely to become in monocytic/monoblastic leukaemia. differentiation. (b) AML M3 (promyelocytic): the
increasingly important in classification. leukaemic cells show marked cytoplasmic
granularity. (c) AML M4 (myelomonocytic): some
Diagnosis of the leukaemic cells have monocytic features.
Clinical features Diagnosis depends on a logical sequence
infiltrated by leukaemic blast cells
of tests.
In practice there is little uniformity in (Fig 20.2). In more immature forms
presentation. Some patients are remark- 1. Blood count and film. The white of AML morphological
ably asymptomatic while others are seri- cell count (WCC) is usually elevated differentiation from acute
ously ill. Bone marrow infiltration by (up to 200 × 109/L) but may be lymphoblastic leukaemia (ALL) can
leukaemic blast cells usually leads to normal or low. There is often be difficult.
anaemia, neutropenia and thrombocy- anaemia and thrombocytopenia. 3. Cytochemistry. Special stains are
topenia. Thus, patients often have Usually there are leukaemic blast used on bone marrow and blood
symptoms of anaemia, infection and cells although occasionally these are smears to help differentiate myeloid
haemorrhage. absent. There may be dysplastic and lymphoid blast cells. In AML
One subtype of AML deserves special changes in other cells. there is positivity with Sudan black
consideration as it must be treated as a 2. Bone marrow aspirate and and myeloperoxidase – these stains
medical emergency: trephine. The bone marrow is are negative in ALL. AML with
Acute myeloid leukaemia 41

monocytic features will stain Table 20.2  Common genetic abnormalities in AML
positively with a non-specific Abnormality Genes involved Associated subtype Prognosis1
esterase stain. t(8;21) AML1-ETO (RUNX1 M2 Good
4. Immunophenotyping. Both -RUNX1-T1)
surface and intracellular antigens t(15;17) PML-RARα M3 Good
are analysed. Characteristic ‘myeloid’ inv 16 CBFB-MYH11 M4 Good
antigens include CD13 and CD33 t(9;11) MLL M4/5 Poor
while CD34 positivity indicates a 5 and 7 (various) Unknown Secondary AML2 Poor
particularly immature cell of origin. 1
Compared with AML with no detectable genetic abnormality.
Modern multicolour flow cytometry 2
Antecedent events include chemotherapy, myelodysplastic syndrome and myeloproliferative disorders.
techniques allow quantitation of 100
blast cells and correlate with both
90
morphology features and the
80
common balanced translocations.
5. Cytogenetics. A bone marrow 70
sample is sent for analysis. 60 Secondary
Chromosome abnormalities are 50 Favourable
associated with particular AML
40 Intermediate
subtypes and also give vital
30 Adverse
prognostic information (see Tables
20.1 and 20.2). 20
6. Molecular biology. Molecular 10
techniques are increasingly 0
important in classification, 0-14 15-34 35-44 45-59 60+
determining prognosis, and in Fig 20.3  Relationship between age of presentation and characteristics of AML. Older patients
monitoring response of disease to have a higher incidence of ‘poor risk’ disease. (Reprinted with permission from Smith ML, Hills RK,
treatment (see p. 100). Sequential Grimwade D 2011 Independent prognostic variables in AML. Blood Reviews 25: 40.)
RT-PCR monitoring (e.g. in patients longer-term ‘maintenance’ treatment is include anti-CD33 antibodies, FLT3
with the t(15;17) subtype in clinical rarely given. The well tested combination inhibitors and demethylating agents.
remission) can predict the of an anthracycline (e.g. daunorubicin)
likelihood of relapse. Numerous and cytosine arabinoside is standard
Prognosis
genetic abnormalities are being induction therapy. Higher doses of cyto-
identified – mutation of the tyrosine sine are often used as consolidation The major factors determining outcome
kinase receptor gene FLT3 is the therapy. Acute promyelocytic leukaemia are age, initial response to treatment
commonest finding in patients with (t(15;17)) is additionally treated with the and genetic abnormalities. Approxi-
normal cytogenetics and carries a differentiating agent all-trans-retinoic mately 80–90% of younger patients will
poorer prognosis. Other genetic acid (ATRA), which reduces the risk of achieve a CR with conventional chemo-
mutations (e.g. nucleophosmin 1 early death from bleeding and improves therapy. Younger patients with ‘standard
(NPM1), two mutations of CEPPA long-term survival compared with chem- risk’ disease have 5-year survivals of
gene) can favourably influence otherapy alone. 40–45% with optimal therapy; this com-
prognosis. Autologous stem cell transplantation pares with around 70% for ‘good risk’
(SCT) can be used to intensify chemo- and 20% for ‘poor risk’ groups. Older
therapy but the benefit has proved dif- patients have a greater incidence of
Management ficult to quantify. Surprisingly, the adverse cytogenetics (Fig 20.3) and toler-
Supportive care precise role of allogeneic SCT is also not ate chemotherapy less well, and CR and
This includes red cell transfusion for clear-cut – most clinicians would con- cure rates are much lower (see p. 93).
anaemia, platelet concentrates for sider a transplant from an available Indeed, it may be kinder not to use
thrombocytopenia and broad-spectrum HLA-matched sibling in a younger chemotherapy in some elderly patients.
intravenous antibiotics for infection. patient with high-risk (see below) or In children, intensive chemotherapy
An indwelling central venous catheter relapsed disease. Novel molecular tar- gives 5-year survival rates of around
facilitates support during and after geted therapies under exploration 50%.
chemotherapy.
Acute myeloid leukaemia
Chemotherapy and stem
■ AML arises out of the malignant transformation of a myeloid precursor cell.
cell transplantation
The first objective of treatment with cyto- ■ The WHO classification emphasises the prognostic significance of non-random
toxic drugs is to achieve a ‘complete chromosome abnormalities.
remission’ (CR) – defined as less than 5% ■ Symptoms mainly result from anaemia, neutropenia and thrombocytopenia.
blast cells in a normocellular bone ■ Prognosis largely depends on age, initial response to treatment, and genetic abnormalities.
marrow. Initial cytotoxic drug treatment
■ Chemotherapy leads to CR rates of 80–90% in younger patients but cure rates are lower,
is termed ‘induction’. A CR is followed around 45%. Allogeneic stem cell transplantation is considered in younger patients at high
by a second sequence of drugs termed risk of relapse.
‘consolidation’. Induction and consolida- ■ Older patients tolerate chemotherapy less well and cure is rarely achievable.
tion take at least several months, but
42 4 LEUKAEMIA

21 Acute lymphoblastic leukaemia


Acute lymphoblastic leukaemia (ALL) is a clonal malignancy Incidence rate
of lymphoid precursor cells. In over 80% of cases the malig- (per 100000 population)
nant cells are primitive precursors of B-lymphocytes and the 10.0
remainder are T-cell leukaemias. The abnormal cell may arise 8.0 Male
at various stages of early lymphocyte differentiation (see p. 8). Female
6.0
ALL has a peak incidence in childhood with a gradual rise 4.0
in incidence in later years (Fig 21.1). The disease has distinct
2.0
characteristics in children and adults. Childhood ALL is often
0.0
curable by chemotherapy whereas cure is elusive in adult ALL. 0 10 20 30 40 50 60 70 80
Poorer outcome in adult ALL is due to a combination of a Age (years)
greater frequency of high-risk leukaemia with more drug Fig 21.1  Incidence of ALL at different ages.
resistance, and less effective treatment regimens.

Table 21.1  Classification of ALL


Classification
Morphological classification1
The French-American-British (FAB) L1 Small uniform blast cells with
morphological classification is based on scanty cytoplasm
characteristics of the blast cells, includ- L2 Large heterogeneous blast cells
ing cell size, nuclear–cytoplasmic ratio, with nucleoli and low
nuclear–cytoplasmic ratio
number and size of nucleoli and the
L3 Basophilic vacuolated blast cells
degree of cytoplasmic basophilia (Fig
Immunological classification
21.2). Morphological classification is (a)
B-lineage ALL
now less important than that based on
  Early pre-B
immunophenotyping, cytogenetics and
  Pre-B ALL
molecular analysis (Table 21.1).
  B-cell
Definition of the immunological sub- T-lineage ALL
types of ALL depends on the presence   Early
or absence of various cell surface and   Cortical
cytoplasmic antigens. A commonly used   Mature
classification divides ALL into early
1
See also Figure 21.2.
pre-B, pre-B, B-cell and T-cell subtypes.
Mature B-cell ALL typically has L3 in ALL than in AML and patients can (b)
morphology. The current WHO classifi- present with symptoms of raised intrac-
cation divides most ALL subtypes ranial pressure (headache, vomiting) or
into B- or T-lymphoblastic leukaemia/ cranial nerve palsies (particularly VI and
lymphoma under the heading ‘precur- VII). Examination findings may include
sor lymphoid neoplasms’. Mature B-ALL pallor, haemorrhage into the skin and
is included as Burkitt lymphoma. mucosae, lymphadenopathy and mod-
In the selection of treatment it is erate hepatosplenomegaly. In males the
important to differentiate between three testes can be involved and should be
broad groups; T-cell ALL, mature B-ALL routinely examined. (c)
and all other types of B-lineage ALL. Fig 21.2  Morphology of ALL blast cells.
Genetic abnormalities are becoming (a) L1 type; (b) L2 type; (c) L3 type. Note that
increasingly important in classification
Diagnosis the L2 cells have more cytoplasm and more
of ALL as they give vital prognostic 1.  Blood count and film prominent nucleoli than L1 cells. L3 type cells
have cytoplasmic vacuolation.
information (Table 21.2). The white cell count may be raised,
normal or low. Only 20% have white cell Cytochemistry is useful in distinguish-
counts greater than 50 × 109/L. Anaemia ing precursor B and B-ALL from T-ALL.
Clinical features
and thrombocytopenia are common. Reactivity with the acid phosphatase
These can be very variable. Accumula- The proportion of blast cells in the stain is seen in malignant T-lymphocytes
tion of malignant lymphoblasts in the white cell count varies from 0% to 100%. but not in B-cells which may show peri-
marrow leads to a scarcity of normal odic acid Schiff (PAS) block positivity.
cells in the peripheral blood and symp- 2.  Bone marrow aspirate
toms may include those associated with and trephine 4.  Immunophenotyping
anaemia, infection and haemorrhage. This is essential to confirm the diagnosis Useful reagents for establishing the
Other common complaints are anorexia and for classification. diagnosis and identifying the immuno-
and back or joint pain. T-cell ALL is asso- logical subtype include antibodies to
ciated with a large mediastinal nodal 3.  Cytochemistry CD19, CD79A and CD22 (found in most
mass and pleural effusions which result Stains which classically show positivity B-lineage ALLs), CD10 (the ‘common
in dyspnoea. Central nervous system in AML – Sudan black and myeloper- ALL antigen’), CD3 and CD7 (found in
(CNS) involvement is more often seen oxidase – are negative in ALL. T-lineage ALLs).
Acute lymphoblastic leukaemia 43

Table 21.2  Chromosomal abnormalities in ALL Table 21.3  Factors predicting poor
Abnormality Prognostic significance prognosis in ALL
Numerical change ■ Increasing age1
High hyperdiploidy (over 50 chromosomes) Favourable ■ High white cell count at presentation

Hyperdiploidy (47–50) Intermediate ■ Certain cytogenetic abnormalities (see Table 21.2)


■ Poor response to treatment2
Pseudodiploidy (46 with structural/numerical change) Intermediate
Hypodiploidy (less than 46) Poor 1
With the exception of children under 1 year who have a worse
Structural abnormality Genes involved prognosis than older children.
2
Assessed from the bone marrow appearance after 14 days of
Philadelphia chromosome, t(9;22)1 BCR-ABL Poor chemotherapy.
t(12;21)2 TEL-AML1 (ETV6-RUNX1) Good
t(1;19) E2A-PBX1 Good intensive chemotherapy. With improved
t(v;11q23) MLL-AF4, ENL-MLL Poor cure rates the long-term side-effects of
t(8;14)3 MYC Good the drugs, including endocrine prob-
1
Must be distinguished from the lymphoid blast crisis of chronic myeloid leukaemia.
lems, secondary leukaemia and cardio-
2
Occurs in 20% cases of childhood ALL. Not detectable by standard cytogenetics. toxicity, are becoming increasingly
3
Seen in B-ALL with L3 morphology.
relevant. Wherever feasible, the use of
a period of ‘maintenance’ using meth- agents with the safest profiles is
5.  Cytogenetics desirable.
otrexate and mercaptopurine. The
Cytogenetic analysis is doubly useful as
greater chance of CNS disease in ALL
structural abnormalities correlate with ALL in adults
(than in AML) necessitates prophylactic
particular subtypes of ALL and both The majority of adult patients enter
treatment to prevent CNS relapse. The
structural and numerical abnormalities remission but are not curable with
usual method is intrathecal and systemic
give prognostic information (see Table chemotherapy alone and less than 40%
chemotherapy with the possible addi-
21.2). Varying patterns of cytogenetic will become long-term survivors. Most
tion of cranial irradiation in those at
abnormality may partly explain the dif- chemocurable patients are aged between
highest risk.
ferent prognosis in children and adults. 16 and 25 years with other good prog-
The ultimate choice of management
The Philadelphia chromosome, regarded nostic features. This ‘good risk’ sub-
is influenced by a number of prognostic
as a marker of ‘incurability’ by chemo- group resembles childhood ALL and
factors which have changed with
therapy, is found in 20–30% of adult chemotherapy alone is a reasonable
improving treatment (Table 21.3). Where
cases but in only 2% of children. initial policy with cure rates of around
clinical and laboratory features predict a
75%. For adults with higher-risk disease
6.  Molecular techniques poor response to chemotherapy alone,
the hope of cure is likely to depend on
Molecular analysis yields complemen- more intensive treatments such as allo-
even more intensive therapy with either
tary and additional information to con- geneic stem cell transplantation (SCT)
autologous or allogeneic SCT. Alloge-
ventional cytogenetics (see Table 21.2). are considered. Of all the prognostic
neic SCT from an HLA-matched family
The cryptic t(12;21) creates a TEL-AML1 indices the most influential is age.
donor performed in first remission gives
(ETV6-RUNX1) fusion gene – this is
ALL in children long-term survival of around 50%.
the commonest genetic rearrangement
The majority of children are curable SCT using an unrelated HLA-’matched’
in childhood ALL and it can only
with current chemotherapy regimens. donor is more risky but can be success-
be detected by molecular techniques.
The standard strategy is intensive induc- ful. In Philadelphia chromosome posi-
Although not yet routinely available in
tion therapy, CNS prophylaxis, and tive ALL the tyrosine kinase inhibitor
most laboratories, global gene expres-
maintenance treatment for 2–2.5 years. imatinib is useful adjunctive therapy
sion profiling reveals distinct patterns in
In children receiving the most intensive (see p. 45). Optimum management of
specific subtypes of ALL (see p. 100).
protocols, 5-year disease-free survivals of adult ALL has yet to be defined and
nearly 90% are now achievable. Autolo- there is a need for careful consideration
Management and gous and allogeneic SCT is best reserved of all the known prognostic factors in
outcome for relapse after chemotherapy or for each case. More elderly patients (over 60
patients with poor prognostic features. years) tolerate chemotherapy less well
General principles and cure rates are very low. In these
New methods for detecting minimal
Patients with ALL require supportive
residual disease during treatment (e.g. cases it is often kinder to concentrate on
care. Chemotherapy is the mainstay of
after induction) allow early identifica- palliation of symptoms and provision of
treatment. Drug schedules vary but
tion of patients at high risk of relapse. a short period of good quality life rather
remission induction classically relies on
Mature B-ALL is a special case best than undertaking aggressive chemother-
three agents: vincristine, a glucocorticoid
treated with short-term fractionated apy with a negligible chance of success.
(e.g. prednisolone) and asparaginase.
The anthracycline daunorubicin may be
Acute lymphoblastic leukaemia
included in the induction regimen and
other drugs, notably methotrexate, cyclo- ■ ALL is a clonal malignancy of lymphoid precursor cells.
phosphamide and cytosine arabinoside, ■ There is a peak incidence in childhood and a gradual rise in later years.
then added in ‘intensification’ (‘consoli- ■ Accumulation of lymphoblasts in the bone marrow often leads to anaemia, infection and
dation’) (see p. 54 for more detail of haemorrhage. CNS involvement is more common than in acute myeloid leukaemia.
individual drugs). The rationale for early
■ The majority of children are curable with standard chemotherapy regimens and CNS
intensification of treatment is to reduce prophylaxis.
the leukaemic cell population quickly
■ In adults, cure by chemotherapy alone is much less frequent. Autologous or allogeneic
and reduce the likelihood of drug resist- stem cell transplantation may be considered in ‘high-risk’ cases.
ance. Therapy is usually completed with

tahir99-VRG & vip.persianss.ir


44 4 LEUKAEMIA

22 Chronic myeloid leukaemia

Chronic myeloid leukaemia (CML) is


a clonal myeloproliferative disorder
Diagnosis and
which results from an acquired genetic
monitoring
change in a pluripotential stem cell. The The major laboratory abnormality in
disease is characterised by a gross over- CP-CML is an elevated white cell count;
production of neutrophils and their this often exceeds 100 × 109/L. The blood
precursors (Fig 22.1). It is unusual in film shows an increase in morphologi-
having three clinical phases: a relatively cally normal myeloid cells at all stages
benign ‘chronic phase’ is followed by of differentiation but with greatest
an ominous ‘accelerated phase’ and, numbers of myelocytes and neutrophils
finally, an almost invariably fatal acute (Fig 22.3). There is usually an absolute
leukaemic phase termed ‘blast crisis’. basophilia. Thrombocytosis and nucle-
The annual incidence of CML is ated red cells may be present.
around one per 100 000 with presenta- The bone marrow appearance is less
tion most common in the fifth and sixth informative than the blood film; pro-
decades of life. The diagnosis is increas- nounced hypercellularity and abnormal
ingly made in asymptomatic patients myelopoiesis is characteristic but not
having routine blood tests. specific for CML. The key diagnostic
abnormality is the presence of the Ph
Fig 22.1  Blood sample (right) from a chromosome. Patients with apparent
Pathogenesis patient with CML. Note the greatly increased CML with Ph chromosome negativity
The hallmark of CML cells is the pres- white cell component (‘buffy coat’) compared need careful review as they may repre-
ence of a Philadelphia (Ph) chromosome with the normal sample. sent an atypical myeloproliferative or
– the t(9;22)(q34;q11) chromosomal myelodysplastic disorder.
translocation. Over 95% of classical The accelerated phase is characterised
CML cases are Ph positive. The Ph trans- is usually associated with an insidious by an increase in the number of imma-
location causes the fusion of the ABL deterioration in the patient’s health and ture cells in the peripheral blood and in
proto-oncogene from chromosome 9 to the need for more intense treatment to blast crisis the blood appearance is dom-
the interrupted end of the breakpoint control splenic size and white cell count. inated by the presence of myeloblasts
cluster region (BCR) of chromosome 22
(Fig 22.2). The chimeric BCR-ABL gene
created on the Ph chromosome (22q−)
encodes a protein with considerably greater tyrosine kinase
activity than the normal counterpart. In chronic phase CML, Chromosome
BCR ABL
cells in the progenitor pool have increased proliferation due 9 9q+ 22 9
to over-expression of BCR-ABL. The mechanism by which the 5' 3' DNA
BCR-ABL oncogene affects stem cell kinetics is not well
understood. It presumably deregulates signalling pathways
involved in proliferation, apoptosis, cellular adhesion and BCR-ABL
genomic stability. Progression to blast crisis with production chimeric RNA
of leukaemic stem cells requires complex additional events 3'BCR
including increased proliferation and self-renewal capacity ABL
avoidance of cell death, a block in differentiation and bypass- SIS
Chimeric fusion
ing of normal immune responses. protein with tyrosine
kinase activity

Clinical features
22 22q-
Patients usually present in chronic phase. Typical symptoms
? Mechanism
are of anaemia, anorexia and weight loss. Splenomegaly is the
most common physical finding and is often marked, causing
pain, bloating and satiety. The occasional patient presents BCR 5' 5'BCR
3'
with gout or hyperviscosity associated with a very high white Proliferation
cell count. Neutropenia and thrombocytopenia are not nor- ABL of myeloid cells
SIS in bone marrow
mally features of chronic phase and infection and haemor-
rhage are rare. Fig 22.2  The Philadelphia chromosome. Chromosomal and molecular
abnormalities in chronic myeloid leukaemia. In a translocation between
After a period of stability in chronic phase, patients develop
chromosomes 9 and 22 (t(9;22) ) the oncogene ABL on chromosome 9 is
blast crisis with symptoms typical of acute leukaemia. Between moved to the breakpoint cluster region (BCR) of chromosome 22. The
chronic phase (CP) and blast crisis is an intervening period resulting BCR-ABL hybrid gene encodes a protein with high tyrosine kinase
of ‘acceleration’. The accelerated phase is poorly defined but activity.

tahir99-VRG & vip.persianss.ir


Chronic myeloid leukaemia 45

Table 22.1  Evaluation of response to imatinib in chronic phase CML


Time after start of Response
treatment Optimal Suboptimal Failure
3 months CHR and at least minor CyR No CyR No CHR
6 months At least partial CyR Less than partial CyR No CyR
12 months CCyR Partial CyR Less than partial CyR
18 months MMR Less than MMR Less than CCyR
Any time Stable or improving MMR Loss of MMR. Presence of Loss of CHR. Loss of CCyR.
BCR-ABL mutations Clonal evolution1

CHR: complete haematological remission; CyR: cytogenetic response; CCyR: complete cytogenetic response; MMR: major molecular response.
1
Additional chromosome abnormalities in Ph+ cells.

milestones shown in Table 22.1 have an Autologous stem cell transplantation


excellent prognosis, with long-term sur- can induce Ph-negative haematopoiesis
vival rates exceeding 90% and a very low but the therapeutic value is unproven.
risk of transformation to accelerated Choice of treatment in chronic
phase or blast crisis. For patients who phase. The number of allogeneic SCTs
are unable to tolerate imatinib or who performed for CP-CML has fallen
become resistant, so-called second gen- sharply in the imatinib era. TKI therapy
Fig 22.3  Blood film in CML showing eration TKIs such as dasatinib and nilo- is now unquestionably the treatment of
myeloid cells of varying maturity.
tinib can be effective. Both these drugs choice in the vast majority of patients.
are more potent than imatinib and are Allogeneic SCT still has a role in patients
(65% of cases) or lymphoblasts (35%). likely to be increasingly used as initial who fail TKI therapy although it is not
The most widely used staging system, treatment. Third generation agents (e.g. currently clear how this failure should
devised by Sokal, is based on patient age, bosutinib) are under investigation. be defined. Patients who do not respond
spleen size, blood blast cell count Stem cell transplantation (SCT). to two TKIs should probably receive
and platelet count. Monitoring of the Allogeneic SCT is at present the only allogeneic SCT if feasible.
response to treatment is now central to proven curative treatment for CML.
management. This involves examination Patients have survived for more than 10 Advanced disease
of the peripheral blood, bone marrow years after SCT with no detectable BCR- In the accelerated phase and blast crisis,
metaphase cytogenetics and measure- ABL transcripts in blood or bone options remain limited. Patients may be
ment of BCR-ABL transcripts using real- marrow. The 5-year leukaemia-free sur- helped by allogeneic SCT but results are
time quantitative polymerase chain vival after HLA- identical sibling SCT is much inferior to those achieved in CP.
reaction (RQ-PCR). The results allow around 60%. Results have been best Blast crisis can be treated with the com-
the patient’s response to be defined at when SCT has been performed in bination chemotherapy regimens used
key time points (see Table 22.1). Where chronic phase within 1 year of diagno- in acute leukaemia, and some patients
there is failure or a suboptimal response, sis. The use of low intensity condition- (particularly those with lymphoblastic
alternative therapy is considered. ing prior to allogeneic transplantation transformation) will initially respond
(see p. 57) potentially allows the proce- and return to chronic phase. Unfortu-
dure in older patients. In younger nately, such ‘remissions’ are usually
Treatment
patients the use of an unrelated HLA- short-lived. Imatinib can also give good
Recent advances have revolutionised the matched donor is possible but results responses but these are rarely
management of chronic phase CML. It are poorer than for sibling donor SCT. sustained.
has been transformed from a disease
with a very poor prognosis to a chronic
subclinical disorder controlled with oral
medication.
Chronic myeloid leukaemia
Chronic phase
Patients presenting with a very high ■ CML is a clonal myeloproliferative disorder arising from an acquired genetic change in a
white cell count may have symptoms of pluripotential stem cell.
hyperviscosity and can benefit from ■ The hallmark of CML cells is the Philadelphia chromosome (t(9;22) ) and the resultant
leucapheresis. chimeric BCR-ABL gene.
Drug therapy. Hydroxycarbamide ■ There is gross overproduction of neutrophils and their precursors.
can also be used to rapidly reduce an ■ CML has an indolent chronic phase followed by a period of acceleration and a final,
initial high white cell count. Tyrosine generally fatal, acute leukaemic phase.
kinase inhibitors (TKIs) are the treat- ■ Tyrosine kinase inhibitors (e.g. imatinib) have much improved the prognosis of CP-CML.
ment of choice. Imatinib at a dose of
■ Allogeneic stem cell transplantation is the only well proven curative treatment but is
400 mg daily orally is normally used associated with significant mortality.
as first-line. Patients achieving the

tahir99-VRG & vip.persianss.ir


46 4 LEUKAEMIA

23 Chronic lymphocytic leukaemia


Chronic lymphocytic leukaemia (CLL)
is a disease characterised by a clonal pro-
liferation of antigen-stimulated mature
B-lymphocytes. It is the most frequent
form of leukaemia in the Western world
and is a disease of the elderly; almost all
patients are over 50 years old at diagno-
sis. Recent research has highlighted the
biological diversity of CLL. The disease
can be most broadly divided into two
types dependent on whether the leukae-
mic cells have a mutation of the immu-
noglobulin heavy chain variable region
(IgVH) gene. Patients with cells lacking
this mutation tend to have more aggres-
sive disease with shortened survival.

Clinical features
Many patients survive long periods with
(a) Fig 23.2  Blood film in CLL. The malignant
cells resemble mature lymphocytes but are prone
minimal symptoms, while others have a to burst during film preparation leading to the
rapid demise with bone marrow failure, formation of smear cells.
bulky lymphadenopathy and hepat-
osplenomegaly. Fortunately, the former
group is in the majority. Indeed, the
Table 23.1  Binet staging system for CLL
diagnosis is increasingly made by chance
Stage A No anaemia or thrombocytopenia
on a routine blood count. Elderly
Fewer than three lymphoid areas1 enlarged
patients with early CLL are very likely Stage B No anaemia or thrombocytopenia
to die from other causes. Three or more lymphoid areas enlarged
Where problems do arise, patients (b) Stage C Anaemia (Hb less than 100 g/L) and/or
commonly complain of symptoms of platelets less than 100 × 109/L
Fig 23.1  CLL is a cause of acquired
anaemia, lymphadenopathy, unusually immunosuppression. (a) Oral candidiasis; 1
Lymphoid areas are cervical, axillary and inguinal
persistent or severe infections and (b) severe chickenpox. lymphadenopathy (uni- or bilateral), spleen and liver.
weight loss. The most frequent findings
on examination are lymphadenopathy cells’ (Fig 23.2). Unexplained persisting immunophenotye is identical to CLL
and splenomegaly. In more advanced lymphocytosis in an elderly person and some of these patients will progress
cases other tissues such as skin, the gas- should always suggest CLL. The diagno- to CLL over time.
trointestinal tract, the central nervous sis is made by proving that the lym-
system, lungs, kidneys and bone may be phocytosis is a proliferation of clonal
infiltrated by leukaemic cells. Occasion-
Staging
B-cells; this is most simply demon-
ally there is transformation into a poorly strated by using in situ or flow cytom- Staging is important in CLL as it helps
differentiated large cell lymphoma etry techniques (see p. 21) to show that in making a rational decision as to
which carries a poor prognosis (Richter the cells have characteristic B-lymphocyte whether to commence treatment, and it
syndrome). The immunodeficiency in antigens and that a single immunoglob- also gives useful prognostic informa-
CLL is caused mainly by hypogamma- ulin light chain (kappa or lambda) exists tion. The easiest method is the Binet
globulinaemia, which predisposes to on the cell surface (i.e. it is a monoclonal adaptation of the previous Rai system
infections (Fig 23.1) and also accounts population). The bone marrow aspirate (Table 23.1); this is simple to apply and
for an increased incidence of other shows increased numbers of small lym- correlates closely with survival.
malignancies. phocytes and a trephine biopsy is worth- Other variables are increasingly
while as the pattern of lymphocyte important in predicting prognosis. As
infiltration gives prognostic informa- gene sequencing is expensive and time-
Diagnosis
tion. The blood film appearance may consuming, expression of the signalling
The diagnosis is suggested by a high suggest autoimmune haemolysis or molecule ZAP-70 can be used as a sur-
lymphocyte count confirmed by the autoimmune thrombocytopenia, both rogate marker for unmutated IgVH
blood film appearance. Lymphocyte of which can complicate CLL. genes and a poor prognosis (Table 23.2).
counts in CLL exceed 5 × 109/L and may The term monoclonal B-cell lymphocy-
reach levels of 500 × 109/L or more. The tosis is used where there are fewer than
cells resemble normal mature lym- 5 × 109/L monoclonal B-lymphocytes in
Management
phocytes but are often slightly larger the blood in the absence of other disease When to start treatment
with a tendency to burst during prepara- features such as lymphadenopathy. There has to be a reason to start treat-
tion of blood films, resulting in ‘smear In the majority of cases the ment in CLL – many patients with early

tahir99-VRG & vip.persianss.ir


Chronic lymphocytic leukaemia 47

stage disease are completely well and Table 23.2  Other prognostic factors in CLL1
need reassurance as to its relatively Factor Prognosis
benign nature. Early treatment may Good Poor
slow progress but does not improve sur- Age Younger Older
vival and can lead to significant side- Response to therapy (e.g. CR2) Yes No
effects including other neoplasms, and Lymphocyte morphology Typical Atypical
the emergence of resistant disease. Histopathology of marrow trephine Non-diffuse infiltration Diffuse infiltration
No. of lymphocytes in blood Low (e.g. <50 × 109/L) High (e.g. >50 × 109/L)
Choice of treatment Lymphocyte doubling time Long (e.g. >12 months) Short (e.g. <12 months)
Treatment should be commenced when Immunophenotype FMC7− CD38− FMC7+ CD38+
the patient develops significant symp- β2-microglobulin Low High
toms, when the disease is progressing Cytogenetics del 13q14 del 11q22, trisomy 12, del 17p133
rapidly or when it is already at an Mutation IgVH genes Yes No
advanced clinical stage. For many years, ZAP-70 expression No Yes
oral chlorambucil (usually given inter- 1
Factors not included in Binet staging system.
mittently) has been the traditional first- 2
Complete remission.
3
p53 mutation/deletion.
line agent for treatment. Chlorambucil
is still useful in older patients and where
there is significant comorbidity but it has Treatment Chlorambucil Fludarabine
now been mostly replaced in first-line (longstanding) (6 courses)
Chest
treatment by the more effective purine infection
Cervical
analogue fludarabine. The combination lymph nodes
of fludarabine with other agents has
brought benefits with higher levels of
Spleen
complete remission (Fig 23.3) translat-
ing into a survival advantage; the current 50
favoured regimen is a combination of Lymphocyte
count
fludarabine, cyclophosphamide and the (x109/L)
monoclonal antibody rituximab (anti- 10
CD20). The alkylating agent bendamus-
0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16
tine is a promising new approach to Months
first-line treatment. Steroids (e.g. pred-
Fig 23.3  Efficacy of fludarabine in CLL. Fludarabine, usually in combination with other agents, is
nisolone) are best reserved for patients
now the favoured first-line treatment. Its general superiority over chlorambucil is demonstrated in this
with pancytopenia or autoimmune com- patient – the chest infection is a reminder that fludarabine is, however, more immunosuppressive than
plications such as haemolysis or immune chlorambucil.
thrombocytopenia. Treatment decisions
are increasingly influenced by risk 100 Fig 23.4  The
factors (see Table 23.2) in addition to combination of
90
stage – for instance, patients with 17p different agents as
deletions are known to respond poorly 80 first-line treatment for
to fludarabine and may be considered CLL leads to improved
70 response rates.
for more novel therapies such as alem- CR%
60 (Reproduced from Hallek
tuzumab (anti-CD52) and ibrutinib. OR% M 2009 State-of-the-art
Radiotherapy can be used as pallia- 50 treatment of chronic
tion, particularly where enlarged 40 lymphocytic leukaemia.
lymph nodes or spleen cause compres- American Society of
30 Haematology Educational
sive problems. Splenectomy can be ben-
20 Book). CR, complete
eficial for painful splenomegaly or
remission; OR, overall
autoimmune cytopenia. In hypogamma- 10 response; Chl,
globulinaemia and recurrent infection, chlorambucil; F,
0
regular intravenous immunoglobulin fludarabine; C,
Chl F FC FCR
has been shown to be well tolerated and cyclophosphamide; R,
quality of life is often improved. rituximab.
None of the above drug regimens or
other treatment modalities will cure Chronic lymphocytic leukaemia
CLL; the emphasis is on control of
■ CLL is the commonest form of leukaemia in the Western world. It is a disease of the elderly.
symptoms and prolongation of life. In
the rare younger patient with CLL a ■ There is a clonal proliferation of B-lymphocytes.
more aggressive approach to treatment ■ Symptoms/signsinclude anaemia, recurrent infections, weight loss, lymphadenopathy and
may be justified to try and eradicate hepatosplenomegaly.
disease. Allogeneic stem cell transplan- ■ The clinical course is often indolent but it can be more aggressive in advanced stages.
tation should be considered in younger ■ Chemotherapy is often not immediately needed in early CLL.
fit patients with refractory CLL or with
■ The combination of fludarabine, cyclophosphamide and rituximab (FCR) is the initial
very poor prognostic features (e.g. 17p treatment of choice in most cases.
deletion).

tahir99-VRG & vip.persianss.ir


48 4 LEUKAEMIA

24 Other leukaemias
of lymphoid cells with blood-filled
Hairy cell leukaemia spaces lined by hairy cells (the pathog-
Prolymphocytic
nomonic ‘pseudosinuses’ of HCL).
leukaemia
Hairy cell leukaemia (HCL) is a rare
chronic B-cell leukaemia characterised There is no specific karyotypic Prolymphocytic leukaemia (PLL) may
by distinctive biological features and abnormality. be connected with chronic lymphocytic
unusual sensitivity to treatment. The leukaemia (p. 46), but it more often
name of the disease is a reference to the Management presents de novo and is best regarded as
distinctive appearance of the malignant A minority of patients (perhaps 10%) a distinct disease. The malignant cell is
cell (Figs 24.1 and 24.2). are asymptomatic and in the first usually of B-lineage and is more mature
instance may require no intervention. than the B-CLL cell. Thus, in addition to
Clinical features Treatment options are as follows. characteristic B-cell antigens, the cells
Patients often have non-specific symp- show a high density of surface immu-
toms including fatigue and weight loss. Nucleoside analogues noglobulin and clonal rearrangements
Infection, the main cause of morbidity Purine analogues are highly effective of both heavy and light chain immu-
and mortality, and bleeding are other treatment, producing a more complete noglobulin genes. TP53 mutations are
possible presentations. The spleen is the and durable response than other thera- commonly found. Approximately 20%
probable site of origin of the malignant pies (Fig 24.3). Both cladribine and of cases are of T-cell lineage.
clone and splenomegaly is found in over pentostatin give excellent long-term sur-
Clinical features and diagnosis
80% of cases. This may be massive and vivals (90–95% at 10 years) and death
PLL is very much a disease of the elderly
is usually not accompanied by lymphad- from HCL is rare.
with a maximum incidence in the eighth
enopathy. The liver is enlarged in 50% of
decade of life. The most common clini-
patients. Interferon alfa
cal presentation of B-PLL is massive
Although less effective than nucleosides,
splenomegaly. Lymphadenopathy is
Diagnosis interferon alfa may still be used in
usually not conspicuous. In T-PLL,
Most cases of HCL have a pancytopenia patients with severe cytopenia or where
involvement of lymph nodes and other
and there may be circulating hairy cells the myelosuppressive effects of nucleo-
tissues including liver and skin is more
in the blood film. Neutropenia and sides are unacceptable. In the latter
common. The characteristic PLL blood
monocytopenia are often particularly patients, G-CSF may also be helpful.
abnormality is a marked lymphocytosis
marked, accounting for the frequency of Major side-effects of interferon alfa
(normally greater than 100 × 109/L).
infection. Hairy cells strongly express include systemic symptoms such as
Anaemia is normal but platelet numbers
the markers of mature activated B-cells: pyrexia, lethargy and depression. Fol-
are often well preserved. Prolym-
CD19, CD11c, CD20, CD22 and CD25 lowing cessation, the disease normally
phocytes are large cells recognised by
and CD103. The bone marrow is nor- slowly relapses but it will often respond
their condensed nucleus with a single
mally difficult to aspirate because of to reintroduction of the drug.
prominent nucleolus surrounded by
increased fibrosis; the trephine will
abundant cytoplasm. B- and T-cell types
show a variable number of infiltrating Splenectomy
are not distinguishable by routine
hairy cells. Where splenectomy is per- This produces improvement in symp-
microscopy.
formed, the sinuses and cords are seen toms and cytopenia but responses are
to be infiltrated by a uniform population not particularly durable and it is now Management
less used than previously. PLL has a poorer prognosis than chronic
lymphocytic leukaemia. The disease is
usually aggressive and the median sur-
vival is 3 years with an even bleaker
outlook in the T-cell variant. Most
patients are elderly and the disease is
frequently refractory to conventional
chemotherapy. Palliative options include
splenic irradiation, splenectomy and
leucapheresis to control the high white
cell count. Survival in B-PLL may be
improved by use of fludarabine and
rituximab while alemtuzumab (anti-
CD52 monoclonal antibody) is emerg-
ing as first-line treatment for T-PLL.

T-cell large granular


lymphocyte leukaemia
Fig 24.2  Hairy cells seen with electron T-cell large granular lymphocyte leukae-
Fig 24.1  Normal lymphocyte and hairy cell. microscopy. mia (T-LGL) is a clonal disorder

tahir99-VRG & vip.persianss.ir


Other leukaemias 49

Bone marrow 1 2 3 Table 24.1  Common clinical and


150 laboratory features in ATLL
140 Clinical
130 Lymphadenopathy
120 Skin lesions
Haemoglobin
(g/L) 110 Splenomegaly
100 Hepatomegaly
90 Pulmonary lesions
80 Laboratory
70 High white cell count
140 Anaemia
Thrombocytopenia
Platelets 100 Hypercalcaemia
(x 109/L)
50

10
6 Diagnosis
5 This requires morphological examina-
White cells 4 tion and immunophenotyping of blood
(X 109/L) 3 Total count lymphocytes or a lymph node biopsy.
2
1 Hairy cells HTLV-1 positivity is established by sero-
0 logical testing and by DNA analysis of
0 1 2 3 4 5 6 7 8 9 10
Months from diagnosis affected tissue where available. Chromo-
Interferon 2-CDA some abnormalities are found in up to
Fig 24.3  Response of hairy cell leukaemia to treatment with cladribine (2-CDA). Bone marrow 90% of cases but are not specific for
examinations 1 and 2 showed numerous hairy cells, and bone marrow 3 a remission. ATLL.

Management
of cytotoxic T-lymphocytes. It is often or a lymphoma. In the most acute form Therapy is offered to patients with
associated with autoimmune disorders presentation is with a frank leukaemia. acute, lymphomatous or unfavourable
such as rheumatoid arthritis. The most The malignant cells in the blood are prognosis chronic type ATLL while
common blood manifestation is severe pleomorphic but often have very irregu- patients with typical chronic or smoul-
chronic neutropenia although anaemia lar polylobulated nuclei. Even within the dering disease are normally first
and thrombocytopenia may also occur. leukaemic group there is great heteroge- observed. Treatment is unsatisfactory
Patients can suffer from recurrent infec- neity with chronic and smouldering and the median survival in aggressive
tions, ‘B symptoms’ such as fever and forms. In 25% of cases the disease is disease is less than 1 year. The lym-
weight loss, and hepatosplenomegaly. better described as a lymphoma as there phoma type of ATLL has a slightly better
Although the disorder is unequivocally is no demonstrable blood involvement. outlook than the leukaemia type. Acute
a leukaemia rather than a reactive Despite the variability of the pathology forms are frequently resistant to conven-
process, it is generally indolent with there are well-defined clinical and labo- tional lymphoma chemotherapy proto-
responses to immunosuppressive ratory features which should prompt cols (e.g. CHOP) and more intensive
treatment. consideration of the diagnosis, particu- regimens with central nervous system
larly in a person from an HTLV-1 prophylaxis are generally recom-
endemic area (Table 24.1). In practice, mended. The combination of interferon
Adult T-cell
lymphoma-type ATLL may be confused alfa and the antiviral agent zidovudine
leukaemia lymphoma
with other forms of T-non-Hodgkin’s may give responses where chemother-
Adult T-cell leukaemia lymphoma lymphoma. Leukaemic ATLL must be apy has failed. Allogeneic stem cell
(ATLL) is a malignant disorder of rela- distinguished from Sezary syndrome, transplantation can be considered in
tively mature T-lymphocytes. It is rare a lymphoproliferative disorder with younger patients with a suitable donor.
but of great interest as it is conclusively circulating T-cells and skin changes The chronic and smouldering leukae-
caused by a virus. The majority of including erythroderma and exfoliative mia forms can run a protracted course
patients with ATLL have antibodies to dermatitis. but eventually transform to an acute
HTLV-1 and definitive evidence for the phase. Skin lesions may be helped by
aetiological role of this retrovirus has extracorporeal photochemotherapy.
come from studies showing monoclonal
integration of proviral DNA in the leu-
kaemic cells. Other leukaemias
The disease is mostly seen in areas ■ Hairy cell leukaemia (HCL) is a malignant proliferation of B-cells with a characteristic hairy
endemic for HTLV-1, notably in parts of appearance. Pancytopenia and splenomegaly are common. Nucleoside analogue drugs are
Japan and in the islands of the Carib- usual first-line treatment and the prognosis is good.
bean. There is a long latent period from ■ Prolymphocytic leukaemia is a B-cell (or less often a T-cell) malignancy typified by
infection to overt disease and less than presentation in the elderly, a high white cell count, splenomegaly and a poor prognosis.
5% of infected people actually develop ■ T-cell LGL leukaemia most commonly manifests as chronic severe neutropenia.
ATLL. Patients most commonly present
■ Adult T-cell leukaemia lymphoma is a malignant disorder of T-lymphocytes associated with
in the fifth decade and, as its name sug- infection by the HTLV-1 virus. It may present as leukaemia or lymphoma.
gests, ATLL may behave as a leukaemia

tahir99-VRG & vip.persianss.ir


50 4 LEUKAEMIA

25 The myelodysplastic syndromes


The myelodysplastic syndromes (MDS) are a group of clonal common in the subtypes with increased blast cells. CMML,
disorders of the bone marrow. Their common feature is bone like the acute monocytic leukaemias, has specific features
marrow failure as a result of ineffective haematopoiesis rather including splenomegaly (rare in other forms of MDS), skin
than reduced haematopoietic activity. A hypercellular marrow infiltration and serous effusions.
and peripheral blood cytopenia with characteristic dysplastic
morphological abnormalities form the basis for diagnosis. In
Diagnosis
early disease there is increased apoptosis of marrow progeni-
tors causing peripheral blood cytopenia but in later MDS Morphology
there is actually decreased apoptosis with characteristic gene The diagnosis of MDS depends on careful morphological
mutations, enhanced survival of myeloblasts and potential examination of the blood film and bone marrow aspirate and
expansion of a leukaemic clone. Patients may develop frank trephine specimens (Fig 25.2). Common abnormalities
acute myeloid leukaemia (AML; see p. 40). Increased marrow include:
angiogenesis and autoimmunity also occur.
■ Peripheral blood. Red cells – anisopoikilocytosis,
MDS is predominantly a disease of the elderly, although it
macrocytosis. Neutrophils – hypogranulation, pseudo-
may affect all ages. It can arise de novo or follow previous
Pelger forms. Platelets – giant forms.
chemotherapy or radiotherapy for another malignancy. It
■ Bone marrow. Erythroid cells – multinuclearity, nuclear
seems to be increasing in incidence.
budding, ring sideroblasts. Myeloid cells – hypogranularity,
increased blast cells. Megakaryocytes – giant forms or
Classification micromegakaryocytes.

This is not straightforward. The French-American-British Where there are changes in all three lines the term ‘triline-
(FAB) classification divides MDS into five subtypes depending age dysplasia’ is used. The bone marrow trephine biopsy
on morphological features and particularly the number of usually confirms marrow hypercellularity, although fibrosis
blood and marrow leukaemic blast cells. The more recent and even hypocellularity may occur.
WHO system (Table 25.1) divides MDS into unilineage or
multilineage dysplasia, refractory anaemia with ring siderob- Genetics
lasts and dysplasia with excess blasts. The FAB entity chronic Around 50% of cases of MDS show cytogenetic abnormali-
myelomonocytic leukaemia (CMML) is now included in the ties. Common changes include monosomy 7 or 7q−, trisomy
overlap ‘MDS with myeloproliferative disorder’ category. It is 8, monosomy 5 or 5q−, and loss of the Y chromosome. The
likely that cytogenetic and molecular abnormalities will be incidence of chromosome abnormalities increases with the
increasingly incorporated into the classification; a current severity of the disease and risk of leukaemic transformation.
example is 5q− syndrome, a distinct subtype of MDS associ- Work is ongoing to better characterise the complex molecu-
ated with a response to novel therapy and a good lar basis of MDS. Mutations of genes encoding cell surface
prognosis. receptors (e.g. KIT), signal transduction proteins (e.g. RAS),
transcription factors (e.g. AML1), epigenetic modifiers (e.g.
MLL) and protein degradation pathways (e.g. CBL) have been
Clinical features
identified.
The diagnosis may follow a routine blood count in an asymp-
tomatic patient. Where symptoms do occur they range from
Prognostic factors
a mild anaemia to the consequences of severe marrow failure
with profound anaemia, leucopenia and thrombocytopenia The outcome is closely linked to the classification and the risk
(Fig 25.1). Abnormal haematopoiesis can cause functional of transformation to leukaemia. The International Prognostic
abnormalities of cells, and infection and haemorrhage may be Scoring System (IPSS) – based on the number of blood cyto-
more severe than would be predicted from the degree of penias, percentage of bone marrow blasts and karyotype – is
cytopenia. Pronounced symptoms are predictably more a simple prognostic tool which can be used to direct

Table 25.1  WHO classification of myelodysplastic syndrome


Refractory anaemia with unilineage dysplasia
Refractory anaemia (RA)
Refractory neutropenia (RN)
Refractory thrombocytopenia (RT)
Refractory anaemia with ring sideroblasts (RARS)
Refractory cytopenia with multilineage dysplasia (RCMD)
Refractory anaemia with excess blasts (RAEB)1
MDS with isolated del (5q)
MDS, unclassifiable
Myelodysplastic/myeloproliferative neoplasms (MDS/MPN)
Chronic myelomonocytic leukaemia (CMML)
Atypical chronic myeloid leukaemia
Refractory anaemia with ring sideroblasts and thrombocytosis (RARST)
Fig 25.1  Purpuric rash in myelodysplastic syndrome. The patient was
1
Defined as 5–10% blasts in bone marrow. thrombocytopenic.

tahir99-VRG & vip.persianss.ir


The myelodysplastic syndromes 51

(a) (b) (c)


Fig 25.2  Myelodysplastic syndromes. Morphological changes in the blood film and bone marrow aspirate. (a) Pseudo-Pelger neutrophil with bilobed
nucleus. (b) Dysplastic megakaryocyte in the bone marrow. (c) Iron stain of the bone marrow showing ‘ring sideroblasts’.

treatment (Table 25.2). Median survivals Table 25.2  The International Prognostic Scoring System for Primary MDS
vary from 6 years in the low risk group Score Value
to less than a year in patients at higher Prognostic variable 0 0.5 1 1.5 2 Risk group Score
risk. Patients with low risk disease have Bone marrow blasts (%) <5 5–10 – 11–20 21–30 Low 0
an average period of 10 years before the Int-1 0.5–1
onset of AML whereas for high risk Karyotype Good1 Intermediate Poor2 – – Int-2 1.5–2
patients this is only a few months. Cytopenias 0/1 2/3 – – – High ≥2

Int: Intermediate
Treatment 1

2
E.g. normal, del (5q).
E.g. complex or chromosome 7 abnormalities.

Supportive care
In patients with significant marrow particularly effective in patients with del prolong survival and delay transforma-
failure, supportive care is crucial to (5q), often leading to resolution of tion to AML. In younger patients,
ameliorate symptoms and prolong life. anaemia and transfusion independence. AML-type chemotherapy and allogeneic
Regular blood transfusion is often nec- After a long period when low-risk MDS stem cell transplantation, the only
essary to control symptoms of anaemia, patients received only supportive care, it potentially curative treatment, are pos-
and haemorrhage is managed with is likely that many will now receive at sible options. The hypomethylating
platelet transfusions. Patients receiving least a trial of growth factors or a novel agent azacitidine has shown efficacy
multiple blood transfusions can benefit agent. in higher risk subgroups with signifi-
from iron chelation therapy. This may cantly prolonged survivals and delay of
reduce not just iron overload but also High-risk MDS onset of AML. It is given subcutaneously
transfusion dependency. Infections In high-risk MDS (e.g. RAEB; see as outpatient therapy and is generally
require swift intervention with broad- Table 25.1), the primary aim is to well tolerated.
spectrum antibiotics.

Specific treatments
Treatment needs to be individualised
according to the type of disease and age The myelodysplastic syndromes
of the patient.
■ MDS is a heterogeneous group of clonal disorders of the bone marrow; the abnormal clone
differentiates ineffectively, leading to a hypercellular marrow and blood cytopenia.
Low-risk MDS
■ MDS may affect all ages but is predominantly a disease of the elderly.
In patients with low-risk MDS, the
main goal of treatment is to improve ■ Diagnosisdepends on the presence of characteristic morphological changes in the blood
cytopenias. Anaemia may respond to and marrow.
erythropoietin alone or combined with ■ Classification into subtypes relies on bone marrow morphology and cytogenetics.
G-CSF. Immunosuppression with anti- ■ Prognosis is highly variable.
thymocyte globulin (ALG) can give
■ Supportive care remains crucial but growth factors and other specific therapeutic agents
good results in younger patients with (e.g. lenalidomide, azacitidine) are increasingly used. Chemotherapy and stem cell
marrow hypocellularity. The immu- transplantation may be considered in younger patients with high-risk disease
nomodulatory agent lenalidomide is

tahir99-VRG & vip.persianss.ir


52 4 LEUKAEMIA

26 Aplastic anaemia
The term aplastic anaemia is a misno- very short telomeres. This is also tendency (caused by thrombocytope-
mer in that the disorder so described is observed in 10–15% of patients with nia). The onset may be gradual or fulmi-
characterised by a pancytopenia arising acquired AA. nant. Symptoms or signs of an
from failure of production of all the Infections known to predispose to AA underlying systemic disorder (e.g. Fan-
normal cells of peripheral blood. The include viral hepatitis and parvovirus coni’s) or possible trigger (e.g. hepatitis)
underlying cause is a reduction in infection. Exposure to chemicals, drugs may be present. An exhaustive history,
the number of pluripotential stem cells. and radiation can damage stem cells. including drug and occupational expo-
This deficit may be exacerbated by an Drugs may depress haematopoiesis idi- sure, and a thorough examination are
abnormality in the marrow microenvi- osyncratically or predictably (Table mandatory.
ronment or an autoimmune reaction 26.2). In roughly two-thirds of patients,
against the abnormal haematopoietic no cause is apparent and AA is termed
Diagnosis
tissue. ‘idiopathic’. Improved haematopoiesis
Aplastic anaemia is rare (approxi- following immunosuppression (see There are really two questions. Is the
mately 2–5 cases/million/year world- below) suggests that in at least some pancytopenia due to aplastic anaemia?
wide) and affects all ages. It must be cases the abnormal stem cell compart- Is this idiopathic AA or aplasia second-
emphasised that it is not a subtype of ment is further compromised by poorly ary to an identifiable cause (Table 26.3)?
leukaemia. However, the disease’s pre- defined immune phenomena. A reasonable sequence of investiga-
senting clinical characteristics, the man- tions is as follows:
agement problems of marrow failure
Clinical features 1.  Blood count and film
(including fulminating septicaemia and
There is a pancytopenia and reticulocy-
haemorrhage) and the possible evolu- Patients with marrow failure predictably
topenia. To define AA there must be at
tion to a clonal marrow disorder dictate present with anaemia, unusually
least two of the following: (1) haemo-
its inclusion in this section. frequent or severe infections (caused
globin less than 100 g/L; (2) neutrophils
by neutropenia) and a haemorrhagic
less than 1.5 × 109/L; (3) platelets less
Classification and Table 26.1  Classification of than 50 × 109/L. There are no abnormal
aetiology aplastic anaemia cells in the blood film.
1.  Idiopathic AA
Aplastic anaemia (AA) may be part of a 2.  Bone marrow aspirate
2.  Congenital AA Fanconi’s anaemia
congenital syndrome, be secondary to and trephine
Dyskeratosis congenita
well-defined insults to the bone marrow, This is the key diagnostic test. The
3.  Secondary AA Drugs – idiosyncratic or
or arise apparently spontaneously with dose-related marrow aspirate can be highly sugges-
no identifiable cause. A simple classifica- Chemicals tive of aplasia with grossly hypocellular
tion is shown in Table 26.1. The most Ionising radiation
common congenital disorder is Fanco- Infection
Table 26.3  Causes of pancytopenia
ni’s anaemia. Affected children suffer Marrow failure or infiltration
from defective DNA repair and the Table 26.2  Drugs associated with aplastic ■ Aplastic anaemia
aplasia often coexists with skeletal anaemia1 ■ Myelodysplastic syndrome
deformities, skin pigmentation (Fig Predictable Cytotoxic agents ■ Leukaemia

26.1) and renal abnormalities. To date, Idiosyncratic Chloramphenicol ■ Myelofibrosis


Sulfonamides ■ Infiltration by other malignancy (e.g. lymphoma,
fifteen genes (termed FANC) have been
Phenylbutazone carcinoma)
identified. Dyskeratosis congenita, ■ Infection (e.g. tuberculosis)
Indometacin
another form of constitutional aplasia, is Gold salts ■ Megaloblastic anaemia
distinguished by a later onset, nail dys- Penicillamine ■ Prolonged starvation

trophy, leukoplakia of mucosal surfaces Carbamazepine Hypersplenism


and a high incidence of epithelial Phenytoin ■ Portal hypertension
Carbimazole ■ Felty syndrome (rheumatoid arthritis)
tumours. There is defective telomere
■ Storage disorders
maintenance and patients usually have 1
This is a selective list of more commonly implicated agents.

(a) (b)
Fig 26.1  Fanconi’s anaemia. (a) Digital abnormalities in brothers with the syndrome. (b) Skin pigmentation.

tahir99-VRG & vip.persianss.ir


Aplastic anaemia 53

particles but a trephine biopsy is neces- cyclophosphamide and the anti-CD52


sary to confirm the diagnosis and quan- monoclonal antibody alemtuzumab. As
tify the degree of hypocellularity (Fig further discussed below, there is concern
26.2). Aplasia may be patchy and if the that immunosuppression may stimulate
trephine is surprisingly cellular in the haematopoiesis but not necessarily cure
context of the blood count, then further the disease.
samples should be obtained. In practice
the only likely confusion is with hypocel- Stem cell transplantation (SCT)
lular myelodysplastic syndrome (see p. The object of allogeneic SCT is to repop-
50) or an atypical presentation of acute ulate the patient’s marrow with normal
leukaemia, the latter particularly in stem cells from a healthy compatible
childhood. donor. Transplantation from an HLA-
identical sibling donor in younger
3.  Tests for an underlying cause patients (less than 45 years) produces
These include liver function tests, viral long-term survival (possibly cure) in
titres, vitamin B12 and folate levels and about 75–90% of cases. It is important
tests for paroxysmal nocturnal haemo- to transplant early in the course of the
globinuria (see p. 31) and the inherited disease as multiple transfusions lead to
bone marrow failure syndromes. sensitisation and an increased chance of
Cytogenetic analysis of the hypocellular
Fig 26.2  Bone marrow trephine in severe graft rejection. Minimal-intensity condi-
aplastic anaemia. The cellularity is markedly tioning regimens (see p. 57) are being
bone marrow is often problematic but reduced. (Compare with normal bone marrow
will reveal abnormal cytogenetic clones explored.
appearance, p. 2.)
in some patients. Immunosuppression or SCT?
Table 26.4  Severity of aplastic anaemia Younger patients (less than 30 years)
Measurement of severity AA is defined as non-severe (NSAA) unless it is: with SAA and a matched sibling donor
1. Severe (SAA). Requires two of three peripheral should be transplanted. In VSAA (see
This is crucial as the severity defined blood criteria: Table 26.4) in this age group, the lack of
from peripheral blood and bone marrow ■ Neutrophils less than 0.5 × 109/L a family donor should prompt a search
measurements predicts the response to ■ Platelets less than 20 × 109/L
for an HLA-matched unrelated donor.
treatment and survival (Table 26.4). The ■ Reticulocytes less than 20 × 109/L
Within the bone marrow: In patients 30–40 years with SAA,
median survival of untreated severe AA sibling SCT and immunosuppression
■ Less than 25% haematopoietic cells, or 25–50%
is 3–6 months with only 20% of patients haematopoietic cells and less than 30% cellularity produce similar survivals over 2–5 years.
surviving longer than 1 year. 2. Very severe (VSAA). Requires a neutrophil count However, a significant proportion of
less than 0.2 × 109/L in the presence of at least one
patients receiving immunosuppression
other peripheral blood criterion and the bone
Management marrow features given for SAA alone, approximately 25% at 10 years,
evolve to clonal marrow diseases such
Removal of cause as paroxysmal nocturnal haemoglob-
Where an agent such as a drug or a around 75%. Responses are poorer in inuria, myelodysplastic syndrome and
chemical is implicated this should be younger patients and in severe aplastic acute myeloid leukaemia. Thus in this
removed. anaemia (SAA). The best regimen is a group matched sibling SCT probably
combination of antithymocyte globulin gives a better long-term prognosis. In
Supportive care (ATG) and ciclosporin. Both drugs are older patients with SAA, and patients
Blood and platelet transfusion may be potentially toxic – ATG can produce with non-severe AA, immunosuppres-
life-saving but should be used judi- pyrexia, rashes and hypotension while sion is generally the treatment of choice.
ciously as patients with AA have intact ciclosporin may cause nephrotoxicity
cellular and humoral immunity and can and hypertension. Complete or partial Growth factors
become sensitised to histocompatibility responses to immunosuppressive Growth factors cannot rectify the stem
antigens. Iron chelation therapy may be treatment can take several months. cell defect and are therefore not rou-
required. Infection in neutropenic Other immunosuppressive drugs tinely used in AA. G-CSF may be useful
patients requires prompt expert man- which may be considered include in severe infection.
agement (see p. 87).
Aplastic anaemia
Restoring normal
haematopoiesis ■ AA is characterised by a pancytopenia arising from the failure of production of normal cells
There are two major options: immu- by the bone marrow.
nosuppression and stem cell ■ There is a reduction in the number of pluripotential stem cells; there may also be an
transplantation. abnormal marrow microenvironment and ill-defined autoimmunity.
■ AA can be congenital, secondary to well-defined insults (e.g. drugs) or idiopathic.
Immunosuppression ■ Prognosis relates to severity, which is defined from blood and bone marrow indices.
AA is thought to be at least in part an Evolution to a clonal marrow disorder such as leukaemia may occur.
autoimmune disease and immunosup- ■ Good supportive care is vital.
pressive agents provide worthwhile
■ Major treatment modalities are immunosuppression and SCT – the choice of treatment is
responses and prolonged survival in based on patient age, disease severity and availability of a stem cell donor.
60–80% of patients with 5-year survival

tahir99-VRG & vip.persianss.ir


54 4 LEUKAEMIA

27 Chemotherapy and related treatments


some antimetabolic activity. The main
General principles
toxic effect of the group is myelosup-
The life cycle of the normal cell is shown pression. Fludarabine is significantly
schematically in Figure 27.1. Conven- immunosuppressive. Mitosis
tional anti-leukaemic and lymphoma (division)
cytotoxic drugs can be broadly divided Topoisomerase poisons
G2 period
into those agents active during only one and inhibitors (DNA repair) G1 period
phase of the cell cycle (‘phase-specific’) This broad class of drugs includes the (preparation)
and those acting at all stages (‘phase anthracyclines (doxorubicin, daunoru-
non-specific’). In practice, most anti- bicin, mitoxantrone, idarubicin) and the
leukaemic drugs act predominantly epipodophyllotoxins (etoposide). The S phase
(DNA synthesis)
against proliferating cells and therefore anthracyclines are cell cycle non-specific
affect a fraction of the malignant cell drugs. The acute dose-limiting toxicity is
population. Thus, if in advanced acute bone marrow suppression but the
leukaemia the total number of malig- cumulative dosage is limited by cardio-
Fig 27.1  The cell cycle.
nant cells is 1010, a single course of chem- toxicity. Etoposide is a phase-specific
otherapy could be expected to kill drug (active in G2) with myelosuppres-
Table 27.1  Common adverse effects of
between 2 and 5 log of cells, leaving sion the major toxicity. cytotoxic drugs
between 105 and 108 residual leukaemic Short-term effects Long-term effects
cells. It can be seen that the chance of Spindle poisons Myelosuppression Infertility
eradication of the disease by chemother- Key agents in this group are the two Nausea and vomiting Secondary malignancy
apy is favoured by early treatment when vinca alkaloids, vincristine and vinblast- Alopecia
the leukaemic mass is small and by ine. They are cell-cycle phase-specific, Mucositis
repeated courses of cytotoxic drugs. It is exerting a cytotoxic effect by binding to
Note: If extravasated from the vein some drugs can cause severe
also logical to combine different agents cellular microtubular protein and inhib- tissue injury.
to maximise the anti-leukaemic activity iting mitosis. Vincristine’s major adverse
and exploit different toxicities (‘combi- effects are mixed motor-sensory and malignancy including leukaemia and
nation chemotherapy’). autonomic neuropathies (patients solid tumours. Alkylating agents are
usually initially complain of ‘pins and particularly leukaemogenic.
needles’ in the fingers or toes and
Major classes
constipation); vinblastine is less neuro-
of conventional Multi-drug resistance
toxic but causes more bone marrow
cytotoxic drugs
suppression. The major problem in the treatment of
Alkylating agents leukaemia and other haematological
Despite their variable structure, all malignancies is the emergence of cells
alkylating agents appear to have a
Side-effects of
resistant to chemotherapy. Genes
common mechanism with cross-linking
conventional
capable of conferring resistance to cyto-
of DNA the principal cytotoxic action.
cytotoxic drugs
toxic drugs have been characterised. Of
Agents commonly used in haematologi- Some toxic effects are common to many particular note is the P-glycoprotein or
cal practice include melphalan, chloram- cytotoxic drugs and must be discussed multi-drug resistance gene (MDR1), as
bucil, cyclophosphamide, busulfan and with all patients receiving a relevant its over-expression can lead to resistance
bendamustine. These are toxic to rapidly single agent or combination chemother- to many of the agents used in the treat-
proliferating cells and the dose-limiting apy (Table 27.1). Myelosuppression and ment of leukaemia. The MDR1 gene
toxicity is myelosuppression. Other alopecia are often unavoidable. However, encodes a membrane protein which
side-effects include infertility, haemor- nausea and vomiting can usually be acts as an ATP-dependent efflux pump
rhagic cystitis (cyclophosphamide) minimised or even completely avoided transporting organic compounds out of
and an increased risk of secondary by modern antiemetic protocols. The the cell. Elevated MDR1 levels appear
malignancy. probability of infertility is influenced by to predict a poor prognosis in acute
the agents used, the total dosage, the myeloid leukaemia. A number of MDR-
Antimetabolites duration of administration and the age reversing agents (e.g. ciclosporin, PSC-
These drugs are compounds which and sex of the patient. Strategies to mini- 833, zosuquidar) have been given in
interfere with the utilisation of a natural mise infertility include prechemother- conjunction with normal chemotherapy
metabolite by virtue of the similarity of apy storage of germ cells (unfortunately, in AML but so far with little benefit.
their chemical structure. Most are ana- fertility is often abnormal at presenta-
logues of nucleic acid precursors. Com- tion) or choice of regimens which are
monly used examples are the folinic relatively non-sterilising. Gonadal failure
Other treatments for
acid analogue methotrexate, the purine occurs more commonly in women and
haematological
analogues 6-mercaptopurine and may be managed by hormone replace-
malignancy
fludarabine, and the pyrimidine ana- ment therapy; androgens are used Conventional chemotherapy currently
logue cytosine arabinoside. The alkylat- in men. Chemotherapy is associated plays the major role in the treatment of
ing agent bendamustine also has with an increased risk of secondary most haematological malignancies but,
Chemotherapy and related treatments 55

as these selected examples demonstrate, proliferative neoplasms and FLT3 inhibi- anti-cancer activities. It has been exten-
there is increasing emphasis on more tors in acute myeloid leukaemia. sively used in CML and hairy cell leu-
targeted therapies which exploit particu- kaemia but is now being largely
lar characteristics of tumour cells and Proteosome inhibitors supplanted by more effective and better
cause fewer systemic side-effects. The proteosome is an enzyme complex tolerated agents.
that plays a crucial part in cell-cycle
Differentiating agents control and gene expression by regu-
In most cases, attempts to induce matu- lating cellular protein degradation.
Haematopoietic growth
ration of malignant cells have been dis- Inhibition of the proteosome ulti-
factor therapy
appointing. One notable exception is mately results in cell death. Bortezomib Several haematopoietic growth factors
the drug all-trans-retinoic acid (ATRA) (Velcade) is a proteosome inhibitor that are routinely used in clinical haematol-
in acute promyelocytic leukaemia (asso- is able to kill tumour cells selectively. It ogy. Their main use is in haematological
ciated with t(15;17)). At pharmacological is effective in myeloma and it also has malignancy.
concentrations ATRA overcomes the activity in non-Hodgkin’s lymphomas.
suppressive effect of the PML-RARα The most troublesome side-effect is Supportive care in patients with
fusion protein, allowing leukaemic pro- peripheral neuropathy. blood cytopenia
myelocytes to differentiate into neu- G-CSF (see p. 2) is most commonly
trophils. Addition of ATRA to normal Epigenetic therapies used to accelerate the production of
chemotherapy reduces the severity of The term ‘epigenetics’ refers to heritable neutrophils following chemotherapy or
APL-related coagulopathy and gives a changes in gene expression which are stem cell transplantation. The shortened
significantly better survival than chemo- not coded in the DNA sequence. Epige- period of neutropenia reduces the inci-
therapy alone. The major side-effect is netic mechanisms, including DNA and dence of infections and the length of
‘ATRA syndrome’ where a rising white histone modifications, are potentially stay in hospital. It can also help main-
blood cell count accompanies systemic reversible. The hypomethylating agents tain the dose intensity of chemotherapy.
upset with cardiopulmonary and renal azacitidine and decitabine have activity Erythropoietin is mainly used to treat
problems. in myelodysplastic syndrome and acute the anaemia of renal failure but may
myeloid leukaemia. ameliorate anaemia in selected patients
Monoclonal antibodies with myelodysplastic syndrome and
Monoclonal antibodies (MoAbs) Anti-angiogenic agents myeloma. Early trials of thrombopoietin-
promise tumour-targeted therapy with Myeloma is known to be associated like agents to treat thrombocytopenia
minimal toxicity. They may be unconju- with increased angiogenesis in the bone were unsuccessful due to the develop-
gated or conjugated to a toxin or radio- marrow. The anti-angiogenic agents tha- ment of neutralising antibodies. Throm-
isotope. One potential target is the lidomide and lenalidomide now play bopoietin receptor agonists are used in
CD20 antigen which is present in over key roles in the treatment of this malig- immune thrombocytopenia (see p. 69)
90% of B-cell lymphomas. An unconju- nancy. Both drugs have other mecha- but they have no definite role in throm-
gated chimeric human-mouse anti- nisms of action including alteration of bocytopenia due to marrow failure.
CD20 MoAb (rituximab) is widely used tumour cell cycle progression and
in association with chemotherapy in immunomodulation. Stem cell mobilisation
follicular and diffuse large B-cell non- G-CSF is used in conjunction with
Hodgkin’s lymphomas. Newer anti- Interferon alfa chemotherapy to ‘mobilise’ stem cells
CD20 agents (e.g. ofatumumab) are Interferon alfa is an antiviral protein from the bone marrow to the blood
being investigated. The linking of a radi- with immunomo dulatory and prior to harvesting (see also p. 56).
onucleotide to anti-CD20 may improve
efficacy. Other MoAbs in clinical prac-
tice include anti-CD52 (alemtuzumab)
in chronic lymphocytic leukaemia, anti-
CD33 (Mylotarg) in acute myeloid leu- Chemotherapy and related
kaemia and anti-CD30 (brentuximab) in treatments
Hodgkin’s lymphoma.
■ There are several classes of conventional cytotoxic drugs with different mechanisms of
action.
Tyrosine kinase inhibitors (TKIs) ■ In
leukaemia and lymphoma it is usual to combine cytotoxic drugs in repeated courses to
Imatinib mesilate (Glivec), a specific maximise anti-tumour activity and exploit different toxicities.
small molecule inhibitor of BCR-ABL, is
■ Cytotoxic drugs have predictable short-term and long-term side-effects.
very effective drug therapy for chronic
■ Conventional chemotherapy is increasingly being supplemented, or even replaced, by
myeloid leukaemia (CML) (see also p.
therapies targeting particular characteristics of tumour cells (e.g. tyrosine kinase inhibitors
45). In patients who develop resistance
in CML).
to imatinib more potent BCR-ABL
■ Erythropoietin may be used to treat anaemia in selected patients with haematological
inhibitors (e.g. dasatinib, nilotinib) can
malignancy.
give good responses. Other TKIs
■ G-CSF is used to shorten the duration of neutropenia after intensive chemotherapy and to
entering clinical practice include JAK2
mobilise stem cells for harvesting.
inhibitors (e.g. ruxolitinib) in myelo­
56 4 LEUKAEMIA

28 Stem cell transplantation


The term ‘stem cell transplantation’ family size in the Western world, only attacking antigens in the recipient and
(SCT) is used to describe a number of around 30% of patients will have an can be abrogated by removal of
different procedures. In allogeneic SCT HLA-identical sibling. Transplants from T-lymphocytes from the donor stem
the haematopoietic stem cells are pro- HLA-haploidentical relatives have been cells. Such ‘lymphocyte depletion’ may
vided by another individual, either a associated with a high rate of morbidity lead to an increased risk of relapse of the
family member or an unrelated donor. and mortality. An alternative strategy is underlying malignancy, confirming that
In autologous SCT the patient’s own to search for an unrelated volunteer much of the curative potential of alloge-
stem cells are used to re-establish hae- donor who is a phenotypic HLA match. neic SCT is due to the presence of immu-
matopoiesis. For many years bone From the worldwide databases of 18 nologically active donor cells rather than
marrow (BM) harvested from the pelvis million HLA-typed volunteers, around the preparative regimen (see also non-
was the only source of stem cells but 50% of Caucasian patients can be found myeloablative SCT). The profound and
now they are more commonly harvested a suitable donor. Success rates are lower prolonged immunosuppression of allo-
from the peripheral blood (PB) by in other ethnic groups. Use of a HLA- geneic SCT renders the patient vulnera-
leucapheresis. mismatched donor leads to an increase ble to life-threatening fungal and viral
in the incidence and severity of the (e.g. cytomegalovirus) infections. Causes
adverse effects of SCT. Weaker trans- of late death include relapse of disease,
Allogeneic and syngeneic
plant reactions result from mismatch chronic GVHD and secondary cancers
(twin) SCT
for minor histocompatibility antigens, related to previous chemotherapy and
The allogeneic procedure is outlined in single peptides derived from polymor- SCT conditioning. Peripheral blood
Figure 28.1. The patient’s own haemat- phic proteins which may differ between stem cells are now used more often than
opoietic stem cells, immune system and donor and recipient. bone marrow for allogeneic procedures.
residual tumour cells are conventionally After conditioning treatment there is a For related donors, PBSC give quicker
destroyed by conditioning treatment period of about 3 weeks before ‘engraft- engraftment and probably reduced
with high-dose chemotherapy and ment’ during which the patient is disease relapse and improved survival
(usually) radiotherapy prior to intrave- severely pancytopenic and immunosup- although there is some increase in
nous infusion of stem cells harvested pressed and requires intensive support- GVHD. Common indications for alloge-
from the healthy donor. The ideal ive care with blood products and neic SCT include acute leukaemia and
patient has a disease curable by alloge- aggressive treatment of any infection. chronic myeloid leukaemia.
neic SCT but not by less toxic treatment Major adverse events include graft failure
and is young (less than 40 years old). with rejection arising from a failure to
Autologous SCT
The ideal donor, excepting the rare pres- immunosuppress the patient adequately,
ence of a twin, is a sibling genotypically and graft-versus-host disease (GVHD). The procedure is outlined in Figure 28.1.
matched with the recipient for HLA-A, GVHD is a potentially life-threatening High-dose chemotherapy, and some-
B and DR. The genes for HLA are found disorder predominantly affecting the times radiotherapy, is followed by rein-
on chromosome 6 and so inheritance skin (Fig 28.2), gastrointestinal tract and fusion of previously stored patient stem
follows the rules of simple Mendelian liver which may occur early after trans- cells. Autologous SCT has less toxicity
inheritance; two siblings have a one in plantation (acute GVHD) or after a few than allogeneic SCT and therefore can
four chance of sharing the same two months (chronic GVHD). GVHD results be performed in older patients. It also
HLA haplotypes. With relatively small from donor immunocompetent cells has the advantage that the patient is the

(a) Allogeneic
Patient
High-dose chemotherapy
± irradiation Very intensive
Stem cells given supportive care and
± additional intravenously GVHD prophylaxis
immunosuppression

Donor Possible lymphocyte depletion


(usually HLA-matched sibling)
Harvest of stem cells

(b) Autologous
Patient

Harvest of stem cells High-dose chemotherapy Stem cells given Moderately intensive
± irradiation intravenously supportive care

Storage of stem
cells ± 'purging'

Fig 28.1  Stem cell transplantation.


Stem cell transplantation 57

Fig 28.2  Acute GVHD of the skin after


allogeneic SCT.

donor and therefore donor unavailabil-


ity and GVHD are not issues. The main Fig 28.3  Peripheral blood stem cells being harvested by leucapheresis.
disadvantage of autologous SCT com-
pared with allogeneic SCT is an
increased incidence of relapse of malig- The rationale of reduced intensity SCT
nant disease. It is not clear whether this is to avoid potentially harmful intensive Umbilical cord blood
arises from resistance to the condition- pre-transplant conditioning and to transplantation
ing treatment or infusion of residual instead harness the anti-leukaemic (or Umbilical cord blood (UCB) is an
tumour cells in the graft. The ‘purging’ other tumour) effects of the donor allo- important source of haematopoietic
of tumour cells from grafts does not reactive T-lymphocytes. These cells are stem cells now being used for transplan-
appear to alter survival. Peripheral blood capable of killing leukaemic stem cells tation. UCB is readily available and large
stem cells (PBSC) have now largely and other tumour cells resistant to con- banks of stored frozen UCB are signifi-
replaced the use of bone marrow in ventional chemotherapy. Patients are cantly extending the application of SCT.
autologous procedures. PBSC can be given moderate doses of chemotherapy Immunocompetent cells in CB are less
harvested from the patient’s blood by and immunosuppressive drugs to mature than the T- and B-lymphocytes
leucapheresis during the recovery phase engender a state of host versus graft tol- in an adult and there is a lower risk of
from moderate doses of chemotherapy erance before the infusion of allogeneic inducing GVHD, allowing the possibil-
(Fig 28.3). The growth factor G-CSF is donor stem cells. As the procedure is of ity of HLA-mismatched UCB transplan-
often used to facilitate the ‘mobilisation’ relatively low toxicity it allows alloge- tation. A cord blood unit provides a
of stem cells. Compared with the tradi- neic SCT in older patients. There is, relatively low stem cell dose leading to
tional bone marrow autologous trans- however, still a significant risk of acute slower haematopoietic and immune
plant, PBSCT gives more rapid recovery and chronic GVHD. Reduced intensity recovery. Results are better where higher
of a normal blood count with fewer conditioning regimens are being devel- cell doses are available. Emerging strate-
infections, less intensive supportive care oped to try and minimise the toxicity gies include the use of double cord
and shortened hospital stay. PBSCT is and to maximise GVT activity. Donor blood transplants and in vitro tech-
currently widely used as a convenient lymphocyte infusions (DLIs) are another niques to increase the number of
and relatively safe method of escalating form of allogeneic cell therapy. In haematopoietic stem cells. UCB trans-
the dose of chemotherapy in patients patients with leukaemia relapsed after plantation has been mainly used in chil-
with acute leukaemia, lymphoma and allogeneic SCT, a simple infusion of dren. In adults it is considered when
myeloma. Its role in the treatment of lymphocytes from the original donor other donor cells are unavailable or not
non-haematopoietic solid tumours may induce a further remission. quickly accessible.
and autoimmune disorders is less
established.

Stem cell transplantation


Recent developments
■ Stem cell transplantation (SCT) procedures may be undertaken using an HLA-matched
Non-myeloablative allogeneic family donor or unrelated donor (allogeneic SCT), an identical twin donor (syngeneic SCT)
SCT (‘reduced intensity’) or the patient’s own stored stem cells (autologous SCT).
As allogeneic SCT has evolved it has ■ Stem cells may be sourced from the bone marrow but are now more commonly derived
become increasingly apparent that its from peripheral blood (PBSC).
curative potential is not simply due to ■ Allogeneic SCT is a more effective anti-leukaemic treatment than autologous SCT but is
the killing of tumour cells by intensive associated with greater toxicity including graft rejection and graft versus host disease.
conditioning regimens. The major ther-
■ The greater curative potential of allogeneic SCT in leukaemia is largely due to a ‘graft versus
apeutic component of allogeneic SCT is leukaemia (GVL)’ effect mediated by donor T-lymphocytes.
thought to be donor T-lymphocytes
■ More recent developments include reduced intensity conditioning regimens exploiting
mediating ‘graft versus tumour (GVT)’ GVL effects and alternative sources of stem cells (e.g. umbilical cord blood).
or ‘graft versus leukaemia (GVL)’ effects.
58 5 LYMPHOMA AND MYELOMA

29 Hodgkin’s lymphoma
The lymphomas are malignant disor-
ders of lymphoid tissue subdivided into
two broad groups – Hodgkin’s lym-
phoma (HL) and non-Hodgkin’s lym-
phoma (NHL).
Hodgkin’s disease was first described
by Thomas Hodgkin in 1832. In devel-
oped countries there is a bimodal age
distribution with peak incidences in
young adults (15–35 years) and the
more elderly (over 50 years). The disease
is commoner in men.

Aetiology
Hodgkin’s lymphoma is an unusual
malignancy in that the malignant cells, Fig 29.1  Reed–Sternberg cells in a lymph node biopsy. This giant cell is binucleated or
termed Reed–Sternberg cells (Fig 29.1), multinucleated with large inclusion-like nucleoli and abundant cytoplasm.
and mononuclear Hodgkin’s cells form
only a minority of the tumour. The
Table 29.1  WHO classification of Hodgkin’s lymphoma
remainder is composed of very variable
Histological subtype Histological pattern Immunophenotype
numbers of other cells including lym-
Lymphocyte predominant Polylobulated Reed–Sternberg (RS) cells CD30−
phocytes, granulocytes, fibroblasts and nodular Nodular growth pattern CD20+
plasma cells. This inflammatory cell CD45+
infiltrate presumably reflects an immune Classical
response by the host against the malig- Lymphocyte-rich Classical or lacunar type. RS cells in CD15+
Nodular sclerosis inflammatory cell background. In nodular CD30+
nant cells. Reed–Sternberg (RS) cells
Mixed cellularity sclerosis type are often fibrous bands CD45−
appear to originate from germinal-centre Lymphocyte depletion
B-lymphocytes. In classical HL the RS
cells are ‘crippled’ germinal-centre B-cells
incapable of secreting immunoglobu- nodes usually gradually enlarge but may
lins, while in lymphocyte predominant fluctuate in size. Patterns of disease
nodular HL RS cells the coding regions suggest contiguous spread via the lym-
of the immunoglobulin genes are intact phatic chain. Mediastinal involvement is
and potentially functional. a particular feature of the nodular scle-
Epstein–Barr virus (EBV) may play rosing histological subtype (Fig 29.2).
a role in classical Hodgkin’s lym- Splenomegaly and hepatomegaly occur
phoma, particularly the mixed cellular- but massive enlargement is rare.
ity subtype. When the disease occurs in Significant systemic upset affects a
patients with HIV infection and after minority of patients (20–30%) at presen-
solid organ transplantation it is often tation. This includes fever, sweating
EBV-associated. There is no specific (often at night), weight loss, pruritus
chromosomal translocation associated and fatigue.
with Hodgkin’s lymphoma.
Lymphocyte predominant
nodular Hodgkin’s lymphoma
Classification
Most cases present with cervical aden-
Fig 29.2  Chest X-ray showing mediastinal
It is acknowledged in the World Health opathy and early stage disease. The
lymphadenopathy in nodular sclerosing
Organization (WHO) classification (see disease is more indolent than classical Hodgkin’s lymphoma.
also p. 60) that ‘Hodgkin’s disease’ com- HL with long survivals common.
prises two distinct ‘Hodgkin’s lympho- However, late relapse and transforma- examination. This is needed to distin-
mas’ with different clinical features: tion to diffuse large B-cell NHL can guish Hodgkin’s lymphoma from other
classical HL and lymphocyte predomi- occur. Treatment of advanced disease is causes of lymphadenopathy.
nant nodular HL (Table 29.1). similar to that used for classical HL (see
Staging
below).
Optimal treatment is determined by
Clinical presentation the stage of disease (Fig 29.3), which

Classical Hodgkin’s lymphoma


Diagnosis and staging is derived from the following
investigations:
Asymmetrical and painless lymphaden- Diagnosis
opathy, most often in the cervical region, The key investigation is biopsy of 1. Blood count and bone marrow
is the most common presentation. The a lymph node for histological investigation. A mild normochromic
Hodgkin’s lymphoma 59

Stage I Stage II Stage III Stage IV


Fig 29.3  Staging of Hodgkin’s lymphoma. Stage I, node involvement in single lymph
node area (e.g. cervical); II, two or more lymph node areas on one side of diaphragm; III,
nodal involvement above and below diaphragm; IV, involvement outside node areas
(e.g. liver, bone marrow). The stage is followed by the letter A (absence) or B (presence)
pertaining to significant systemic symptoms (one or more of: unexplained fever above 38°C,
night sweats, loss of more than 10% body weight in 6 months). A single extranodal site is Fig 29.4  PET scan appearance in Hodgkin’s
designated E. lymphoma.

or microcytic anaemia and blood disease) and the radiation field may be Table 29.2  Factors predicting
eosinophilia may be present. Bone more restricted when combined with a poor prognosis
marrow aspiration and trephine chemotherapy. There are various chem- ■ Advanced stage (most important)
biopsy to detect infiltration by otherapy protocols but most common is ■ B symptoms
disease is necessary in more ABVD (doxorubicin, bleomycin, vinblas- ■ Increased tumour bulk
■ Increased sites of disease
advanced cases. tine, dacarbazine), which is given intra-
■ Advanced age
2. Imaging. A whole body computed venously as 4-weekly cycles. ■ Extranodal disease
tomography (CT) scan is the central ■ Poor response to chemotherapy (e.g. after two
staging procedure. In difficult cases Advanced stage disease cycles)
this may be supplemented by All patients with stage III or IV disease ■ Early relapse
■ Elevated erythrocyte sedimentation rate (ESR)
magnetic resonance imaging (MRI). require chemotherapy with possible
■ Lymphopenia
Positron emission topography (PET) addition of radiotherapy for bulky ■ Anaemia
scanning is increasingly used in disease or palliation of symptoms.
staging and also to assess response ABVD is the regimen of choice for most
during and after treatment (Fig patients. This is being compared in clini-
29.4). Clinical trials are needed to cal trials with ‘alternating’ or ‘hybrid’ prognostic factors influence outcome
better define its role. regimens containing a larger number of (see Table 29.2). Cure rates for early
drugs. Autologous stem cell transplanta- stage disease are around 90% while even
tion is the best choice for younger more advanced disease is curable in up
Management of classical patients who fail induction chemother- to 80% of patients with optimal manage-
Hodgkin’s lymphoma apy or who have early relapse. Novel ment. As in other haematological
targeted therapies under investigation malignancies, elderly patients tolerate
The challenge is to improve current
include the anti-CD30 monoclonal chemotherapy less well and cure rates
high cure rates while reducing the inci-
antibody-drug conjugate brentuximab are more modest. In long-term survivors
dence of the serious late complications
vedotin. there is a risk of secondary malignancy.
of radiotherapy and chemotherapy.
Young women receiving mediastinal
irradiation are at particularly high risk
Early stage disease Prognosis and late effects of breast cancer. Other possible late
Patients with stage I or II disease who of treatment effects of treatment include cardiac
lack adverse features (Table 29.2) have
Survival rates are closely linked to disease, lung damage, sterility and endo-
been traditionally treated with radio-
stage although within each stage other crine dysfunction.
therapy alone. This is given over an
extended field using a linear accelerator.
Nodes above the diaphragm are treated
using the ‘mantle’ field (like the mantle Hodgkin’s lymphoma
on a suit of armour) while the ‘inverted ■ The term Hodgkin’s lymphoma describes a group of lymphomas distinct from the
Y’ field includes all nodes below the ‘non-Hodgkin’s’ lymphomas.
diaphragm. ■ The presumed malignant cells, Reed–Sternberg and mononuclear Hodgkin’s cells, compose
Radiotherapy alone fails to cure a minority of tumour cells.
disease in 20–30% of patients and there ■ Common clinical presentations are palpable lymphadenopathy and constitutional
is now more widespread use of chemo- symptoms.
therapy in early stage disease – either ■ Prognosis is largely determined by the stage of the disease.
alone or in combination with radiother-
■ Chemotherapy leads to high cure rates even in advanced disease. The late side-effects of
apy. Chemotherapy may be used in such treatment (e.g. secondary malignancy) are significant.
shorter courses (than for advanced
60 5 LYMPHOMA AND MYELOMA

30 Non-Hodgkin’s lymphoma
Malignant solid tumours of lymphoid Table 30.1  The WHO classification of
tissue which are not Hodgkin’s lym- lymphoid malignancy1
phoma are termed non-Hodgkin’s With respect to NHL, approximately 90% of cases are
lymphomas (NHL). This group of lym- of B-cell type and 10% of T-cell type. The commonest
phomas is even more heterogeneous NHL entities are follicular (20–25% of all cases) and
diffuse large B-cell (30–35%)
than Hodgkin’s lymphoma. The disease
B-cell neoplasms
is the most common haematological Precursor B-cell neoplasms
malignancy and is currently the fifth Precursor B-lymphoblastic lymphoma/leukaemia
most common cancer in the Western Mature B-cell neoplasms
world. It appears to be increasing in inci- Chronic lymphocytic leukaemia/small lymphocytic
lymphoma
dence. NHL may occur at any age but
Lymphoplasmacytic lymphoma
the median age of presentation is 55–60 Hairy cell leukaemia
years. Plasma cell myeloma/plasmacytoma
Marginal zone lymphoma
Follicular lymphoma
Aetiology Mantle cell lymphoma
Diffuse large B-cell lymphoma
The cause of the majority of cases of Burkitt’s lymphoma
NHL is obscure. However, specific chro- B-cell proliferations of uncertain malignant
mosomal translocations are closely potential
T-cell and putative NK-cell neoplasms
associated with particular histological
Precursor T-cell neoplasms
types. Thus, the majority of Burkitt’s Precursor T-lymphoblastic lymphoma/leukaemia
lymphoma cases demonstrate the t(8;14) Mature T-cell and NK-cell neoplasms Fig 30.1  Axillary lymphadenopathy in
abnormality in which the MYC onco- T-cell prolymphocytic leukaemia non-Hodgkin’s lymphoma.
gene on chromosome 8 is moved next T-cell large granular lymphocyte leukaemia
Adult T-cell lymphoma/leukaemia
to the immunoglobulin heavy chain
Mycosis fungoides/Sezary’s syndrome
region on chromosome 14. Almost Enteropathy-type T-cell lymphoma requiring no immediate treatment, or an
90% of follicular low-grade lymphomas Angioimmunoblastic T-cell lymphoma aggressive, rapidly fatal malignancy.
are characterised by t(14;18) where the Peripheral T-cell lymphoma unspecified
■ Nodal involvement. Painless
BCL2 gene on chromosome 18 is moved Anaplastic large cell lymphoma
Hodgkin’s lymphoma (see p. 58) lymphadenopathy (Fig 30.1), often in
to the immunoglobulin heavy chain
the cervical region, is the most
region. This leads to excessive expres- 1
See text for discussion.
common presentation of NHL.
sion of BCL2, an oncogene known
Enlarged nodes may cause
to inhibit apoptosis (programmed cell clinical course but are often curable.
complications such as superior vena
death). It is likely that such chromo- Low-grade tumours are composed of
cava syndrome and hydronephrosis.
some rearrangements require further smaller, better differentiated cells. They
■ Extranodal involvement. Intestinal
events – perhaps co-expression of a are more indolent clinically but have a
lymphoma can present with vague
second proto-oncogene or antigenic tendency to repeatedly relapse.
abdominal pain, anaemia (caused by
stimulus – to produce the clonal malig- The current WHO classification
bleeding) or dysphagia. CNS disease
nant cell. Possible triggering antigens avoids the overly simplistic high-grade/
frequently leads to headache and
include Helicobacter pylori in gastric low-grade split and divides lymphomas
cranial nerve palsies and may cause
MALT lymphoma and hepatitis C in into more specific subtypes based on
spinal cord compression. Lymphoma
marginal zone lymphoma. The aggres- clinical features, morphology, immu-
may arise in the skin (e.g. mycosis
sive extranodal lymphomas seen in nophenotype, karyotype and molecular
fungoides). Bone marrow
AIDS are likely to result from a combi- characteristics. In addition to NHL and
involvement is more common in
nation of immunosuppression (due Hodgkin’s lymphoma the WHO scheme
low-grade lymphomas and can result
to the HIV virus), deregulation of a contains a number of other lymphoid
in pancytopenia.
proto-oncogene (MYC) and secondary neoplasms occurring mainly at extran-
■ Systemic symptoms. Sweating and
viral infection (Epstein–Barr virus). odal sites that are discussed elsewhere
significant weight loss occur in less
Similar tumours may follow organ (e.g. myeloma, hairy cell leukaemia).
than a quarter of patients and, where
transplantation. Some of the major entities are shown in
present, usually indicate advanced
Table 30.1.
disease. Occasionally, patients present
Classification with metabolic complications such as
This is complex and ever-changing with
Clinical presentation hyperuricaemia, renal failure and
hypercalcaemia.
a real risk of ‘heart-sink’ for the uniniti- NHL is essentially a disease of lymph
ated. In simplest terms NHL can be nodes but it has a more diverse presen-
divided into ‘high-grade’ and ‘low-grade’ tation than Hodgkin’s lymphoma with
types. High-grade tumours are com-
Diagnosis and staging
more irregular spread and a higher
posed of large poorly differentiated incidence of extranodal involvement. It Diagnosis depends on obtaining a
lymphoid cells. They have an aggressive may be an indolent disorder, perhaps tissue biopsy, usually a lymph node, for
Non-Hodgkin’s lymphoma 61

histological examination (Fig 30.2). being a rapidly enlarging nodal mass. A


Immunophenotyping is used to identify few cases will have transformed from
the degree of maturation of the malig- previous follicular NHL. Localised
nant cell and determine whether it is of disease is generally treated with a com-
B- or T-cell origin. B-cell antigenic bination of shortened chemotherapy
‘markers’ include CD19, 20 and 22 and and local radiotherapy. Disseminated
T-cell markers CD2, 3, 5 and 7. Gene disease requires full course chemother-
rearrangement studies also aid identifi- apy and an anthracycline-based regimen
cation. B-cell lymphomas have their combined with rituximab (R-CHOP;
immunoglobulin genes clonally rear- cyclophosphamide, doxorubicin, vinc-
ranged while in T-cell lymphomas there ristine, prednisolone) is the standard.
is clonal rearrangement of the T-cell Approximately 70–80% of patients will
receptor genes. Molecular techniques achieve remission and 60% will be
(see p. 100) are being increasingly used cured. In patients with high-risk disease
to detect chromosome abnormalities (based on IPI or genetic abnormalities)
and to derive prognostic information. or relapse, use of more intensive chemo-
The staging system is similar to that therapy with or without stem cell
used in Hodgkin’s lymphoma. Patients support can potentially improve the
are staged with CT scanning (Fig 30.3), prognosis.
MRI or PET, and a bone marrow aspi-
Fig 30.2  Section of a cervical lymph node
showing extensive infiltration with large
rate and trephine. However, in NHL the poorly differentiated lymphoid cells typical Selected other
stage plays a more modest role in man- of diffuse large B-cell non-Hodgkin’s lymphoma subtypes
agement than in Hodgkin’s lymphoma. lymphoma. Mantle cell lymphoma is heterogene-
The histological type of the tumour is ous but is typically disseminated with
more closely related to the likely clinical marrow involvement and a poor
course and other factors impinge upon response to treatment. Median survival
prognosis. An international prognostic is only 4 years. Marginal zone lym-
index (IPI), based on age, stage, bulk of phoma is indolent and includes extran-
disease, performance status and serum odal tumours of mucosa-associated
lactate dehydrogenase (LDH) level, is lymphoid type (MALT). MALT lym-
commonly used (see Appendix III). phoma of the stomach is associated with
Helicobacter pylori and antibiotic treat-
ment to eliminate the bacterium may
Management and
lead to lymphoma regression. Burkitt’s
prognosis
lymphoma is treated with intensive
Only some of the commoner NHL sub- combination chemotherapy with a good
types will be discussed. chance of cure in younger patients.
There is a high risk of tumour lysis syn-
Follicular lymphoma Fig 30.3  CT scan of the abdomen showing drome. Peripheral T-cell lymphomas
Follicular lymphoma is usually a ‘low- enlargement of lymph nodes in a patient have a high incidence of extranodal
grade’ tumour. There is typically dis- with non-Hodgkin’s lymphoma. disease and, overall, a worse prognosis
seminated disease at presentation, an than B-cell NHL. The CHOP regimen
initial good response to therapy, but after induction treatment appears to is commonly used but in view of
then recurrent relapses at decreasing improve survival but cure remains the disappointing results a number
intervals. Median survival is between elusive. of experimental agents are under
10 and 15 years. Patients may initially investigation.
require no treatment. Local disease Diffuse large B-cell lymphoma
(unusual) may be treated with radio- This is the commonest type of ‘high-
therapy. For disseminated disease grade’ NHL, the classic presentation
requiring intervention there is a wide
range of possibilities. There is a move
towards rituximab (anti-CD20 mono-
clonal antibody)-containing regimens
(e.g. R-CVP: rituximab, cyclophospha- Non-Hodgkin’s lymphoma
mide, vincristine, prednisolone) but oral
■ The term NHL encompasses solid tumours of lymphoid tissue which are not Hodgkin’s
agents such as chlorambucil and fludara-
lymphoma.
bine are still widely used. For relapsed
■ Histologicalclassification is complex. There is great clinical heterogeneity with indolent and
disease in younger patients either
aggressive types of disease.
autologous or allogeneic stem cell trans-
■ Indolent (e.g. follicular) NHL often initially responds well to chemotherapy but cure is
plantation should be considered. Radio-
elusive.
immunotherapy (the combination of a
■ Aggressive (e.g. diffuse large B-cell) NHL may be cured with conventional chemotherapy
monoclonal antibody with a radioiso-
combined with rituximab (R-CHOP); autologous stem cell transplants are increasingly used
tope) is a promising alternative. Rituxi- for ‘high-risk’ and relapsed disease.
mab maintenance therapy for 2 years
62 5 LYMPHOMA AND MYELOMA

31 Myeloma

Introduction
Multiple myeloma is a malignant disorder in which there is an
uncontrolled proliferation of clonal plasma cells in the bone
marrow. Secretion of a variety of proteins by the malignant
cells leads to characteristic symptoms and signs. Myeloma
constitutes 10–15% of all haematological malignancies and is
essentially a disease of the elderly – only 2% of cases are diag-
nosed in patients less than 40 years old.

Basic biology
The initial step in the development of myeloma is the appear-
ance of a small number of clonal plasma cells (the clinical
syndrome is ‘monoclonal gammopathy of uncertain signifi-
Fig 31.1  The blood film in myeloma. There is marked rouleaux
formation and increased background staining.
cance’ (MGUS) ). Approximately 50% of patients with MGUS
have translocations involving the immunoglobulin heavy
chain locus on chromosome 14q32. With progression to frank
myeloma, more complex genetic events occur in the neoplas-
tic plasma cells. Changes in the bone marrow microenviron-
ment include the induction of angiogenesis, the suppression
of cell-mediated immunity and increased secretion of
interleukin-6, a powerful growth factor for myeloma cells.
Bone lesions result from osteoclast activation. Myeloma cells
secrete a monoclonal immunoglobulin or immunoglobulin
fragments (‘M-proteins’ or ‘paraproteins’) composed of a
single heavy chain class and a single light chain class, kappa
or lambda. Most myelomas produce IgG or IgA but light
chains alone are produced in over 10% of cases. Free light
chain appearing in the urine is termed Bence Jones protein.
Fig 31.2  The bone marrow in myeloma. The malignant plasma cells
Occasionally myeloma is non-secretory with no detectable show varying degrees of maturity.
M-protein. Localised plasma cell tumours in the absence of
systemic myeloma are termed ‘plasmacytomas’.

Clinical features
More than two-thirds of patients have bone pain at presenta-
tion. Pain is most common in the back and chest and may be
attributed to ‘arthritis’. More advanced bone disease can lead
to pathological fractures or vertebral collapse with loss of
height. Infiltration of the bone marrow by plasma cells may
lead to symptoms of anaemia or bleeding due to thrombocy-
topenia. Infections are common due to immune paresis (low
Fig 31.3  Electrophoretic strip showing serum paraprotein bands.
level of normal immunoglobulins) and other complications
Patient 1 has an IgM paraprotein (Waldenström’s macroglobulinaemia),
which may lead to symptoms include hypercalcaemia, amy- patient 2 IgA myeloma and patient 3 IgG myeloma.
loidosis and renal failure. The major cause of the nephropathy
is deposition of obstructive tubular casts composed of immu- myeloma requires evidence of such impairment; typically
noglobulin light chains – other possible factors include dehy- increased calcium, renal insufficiency, anaemia, or bone
dration, infection and amyloid. lesions (Table 31.1 and Fig 31.3). Bony disease is increasingly
assessed by MRI scanning in addition to traditional X-rays
(‘skeletal survey’) (Fig 31.4). Patients who have a paraprotein
Diagnosis and staging
in the serum but who do not meet the criteria for myeloma
Myeloma is an easy malignancy to miss as the early symp- are diagnosed as having MGUS. They have a rate of progres-
toms such as malaise and backache are common in the popu- sion to myeloma of 1% per year. Monoclonal gammopathy is
lation. The combination of backache and a high erythrocyte associated with other diseases such as lymphoma, non-
sedimentation rate (ESR) should be taken seriously as it may haematopoietic malignancies and connective tissue disorders
indicate myeloma or another metastatic malignancy. but it is also quite common in healthy elderly people (approxi-
In asymptomatic (‘smouldering’) myeloma there is gener- mately 5% over 70 years of age).
ally a serum monoclonal protein >30 g/L and/or bone marrow The prognosis of myeloma can be predicted from present-
clonal plasma cells >10% but no related organ or tissue ing clinical and laboratory features (Table 31.2). The combina-
impairment (Figs 31.1 and 31.2). A diagnosis of symptomatic tion of a high β2-microglobulin level and a low albumin level
Myeloma 63

Table 31.1  Diagnostic criteria for


symptomatic myeloma
Monoclonal protein in serum and/or urine (Fig 31.3)
Bone marrow clonal plasma cells1 or plasmacytoma
(Fig 31.2)
Related organ or tissue impairment (end organ
damage including bone lesions)

1
If flow cytometry is performed most plasma cells (>90%) will show a
‘neoplastic’ phenotype.

Table 31.2  Myeloma: poor prognostic factors


Low haemoglobin
High calcium Fig 31.5  The fundus in hyperviscosity
High M-protein or Bence Jones protein level (a)
syndrome complicating Waldenström’s
Multiple lytic lesions on X-ray
macroglobulinaemia.
High creatinine (i.e. renal failure)
High β2-microglobulin
(see p. 94). Additional bisphosphonate
Low albumin
Poor response to chemotherapy therapy is helpful in hypercalcaemia,
Adverse cytogenetics (e.g. del(17p) ) and anaemia can respond to erythropoi-
etin. Renal failure often responds to
carries a particularly poor prognosis. rehydration and chemotherapy but
Cytogenetic and molecular genetic pro- haemodialysis may be required.
files of the malignant cells also predict
myeloma behaviour. Hyperdiploid and Palliative treatment –
t(11;14) mutations define standard risk a team approach
disease while non-hyperdiploid, t(4;14), Particular emphasis is placed on pain
del(17p) and del(13q) mutations indi- relief and the maintenance of independ-
cate inferior outcome. ence (see pp. 94–95).

Management and Waldenström’s


outcome macroglobulinaemia
(b)
Myeloma may be diagnosed by chance This disease is a form of indolent lym-
Fig 31.4  X-rays of the skull (a) and left
on laboratory screening in patients with radius and ulna (b) in a patient with phoma. It is appropriately considered
limited disease and no symptoms. In myeloma. Numerous lytic lesions are seen. with myeloma as the malignant cells,
this group, about 20% of all patients, which show features of lymphocytes
the disease may remain stable for additionally receive a bisphosphonate. and plasma cells, secrete an IgM para­
several years and there is no advantage In the very elderly or in patients with protein. Patients may complain only of
in early intervention. Where treatment significant comorbidity, a gentler fatigue, but high IgM levels can lead to
is required this generally entails drug approach (e.g. low dose melphalan and the ‘hyperviscosity syndrome’, with con-
therapy, management of specific com- prednisolone) may be justified. In the fusion and neurological symptoms. In
plications, and palliation. rare very fit young patient allogeneic these cases retinal examination reveals
stem cell transplantation is a potentially engorged veins, haemorrhages, exudates
Drug therapy curative but very toxic option. (Fig 31.5) and rarely papilloedema.
Myeloma remains incurable with Other possible physical signs include
current standard treatment but there Management of complications lymphadenopathy and hepatosplenom-
has been recent progress with the intro- The pain of bone disease may require egaly. Where treatment is required,
duction of novel therapeutic agents local radiotherapy in addition to analge- options include chemotherapy (e.g.
targeting myeloma cells and their micro- sia. In spinal compression, radiotherapy chlorambucil or fludarabine) and mono-
environment. Treatment algorithms are and high-dose steroids usually obviate clonal antibodies (e.g. rituximab).
evolving rapidly but the immunomodu- the need for laminectomy. Spinal Significant hyperviscosity requires
latory agent thalidomide is frequently pain may be alleviated by vertebroplasty plasmapheresis.
used in first-line regimens (often com-
bined with dexamethasone and cyclo- Myeloma
phosphamide). In younger fitter patients
■ Myeloma is a malignant proliferation of plasma cells.
(<65–70 years) induction therapy is gen-
erally followed by stem cell harvesting ■ Diagnostic features include an ‘M-protein’ in the serum and/or urine, osteolytic bone
lesions and infiltration of the bone marrow by malignant plasma cells.
and intensification of treatment with
high dose melphalan and autologous ■ Bone pain is the most common presenting symptom.
stem cell transplantation. This strategy ■ Complications include renal failure, hypercalcaemia and amyloidosis.
gives a median survival of 5 years. ■ Commonly used agents include thalidomide, lenalidomide and bortezomib. Autologous
Other agents increasingly used in stem cell transplantation is performed in younger fitter patients.
induction and maintenance therapy ■ Good palliative care, especially pain relief, is crucial.
include lenalidomide, pomalidomide
■ Waldenström’s macroglobulinaemia is a form of indolent lymphoma with secretion of an
and the proteosome inhibitor borte- IgM paraprotein and possible hyperviscosity.
zomib (Velcade). All patients should
64 6 MYELOPROLIFERATIVE NEOPLASMS

32 Polycythaemia
Plasma
Introduction volume (PV)
In simple terms, polycythaemia (or
Red cell
erythrocytosis) means an increase in red mass (RCM)
cell count, haemoglobin and packed cell Upper limit
volume (PCV) above the normally red cell mass
accepted levels. Polycythaemia due to an
absolute increase in red cell mass may
occur as a myeloproliferative neoplasm
(polycythaemia vera (PV) ) or secondary (a) (b) (c)
to hypoxia or an abnormal focus of Fig 32.1  Red cell mass and plasma volume in normality, true polycythaemia and apparent
erythropoietin secretion. In ‘apparent polycythaemia. (a) Normal red cell mass (RCM) and plasma volume (PV). (b) True polycythaemia:
polycythaemia’ the raised haemoglobin there is a significant increase in RCM and total blood volume. (c) Apparent polycythaemia: RCM and PV
and PCV are not accompanied by a sig- are at the upper and lower limits of the normal range with a resultant increased haematocrit.
nificantly raised red cell mass; usually
the plasma volume is relatively reduced
(Fig 32.1). Male > 0.52
Raised packed
Polycythaemia
cell volume Female > 0.48
An approach to the
patient with
polycythaemia
The initial decision to investigate further No Apparent
Raised red cell mass
is taken on the basis of a persisting polycythaemia
raised PCV (haematocrit) or haemo-
globin level. If true polycythaemia is Yes
confirmed by measurement of red cell
mass and plasma volume then the next Yes
Causes of secondary polycythaemia Secondary
step is to determine whether this is present1 (see Table 32.2) polycythaemia
primary or secondary. The full sequence
of investigations is not required in all
No
cases. For example, in a patient with
known respiratory disease causing
Diagnostic criteria for polycythaemia Yes
chronic hypoxia, a degree of polycythae- Polycythaemia
mia is predictable and does not require vera present (see Table 32.1) vera
investigation (Fig 32.2).
No

Clinical syndromes
Polycythaemia vera (PV) Idiopathic erythrocytosis
PV is a myeloproliferative neoplasm;
other diseases in this category are essen- Fig 32.2  Approach to the patient with polycythaemia.
tial thrombocythaemia and myelofibro- 1
If there is any doubt as to the secondary aetiology, investigations for polycythaemia vera (e.g. JAK2
sis (see p. 66). In PV a pluripotential stem testing) should still be performed.
cell is mutated. Almost all patients with
the disease (and some with essential dysfunction may cause a bleeding ten- hypercellularity but there may be no
thrombocythaemia and myelofibrosis) dency. The increased cell turnover can pathognomonic features. Testing for the
have an identical acquired point muta- lead to gout (Fig 32.3). Patients are char- JAK2 V617F mutation is now central to
tion in the Janus kinase 2 (JAK2) gene. acteristically plethoric and may have the diagnosis of PV (Table 32.1). Around
Clinical features. The raised red cell rosacea (Fig 32.4). Palpable splenomeg- 95% of patients with PV are positive. In
mass and total blood volume with asso- aly may be present. the rare negative PV cases, mutations in
ciated hyperviscosity causes the symp- Diagnosis. The diagnostic challenge exon 12 of JAK2 have been found. It is
toms and signs of the disease. Common is to differentiate PV from a secondary likely that other genetic events (e.g.
complaints include headaches, dizziness, polycythaemia. Splenomegaly and ele- MPL, TET2 mutations) are required for
lethargy, sweating and pruritus (the vated white cell and platelet counts are disease development.
latter particularly after a hot bath). Most suggestive of PV. Increased erythropoi- Management. The dual purpose of
importantly, there is an increased risk of esis can lead to iron deficiency. Erythro- treatment is to relieve symptoms and to
arterial and venous thrombosis, particu- poietin estimation by radioimmunoassay reduce the risk of complications such as
larly strokes. Paradoxically, a combina- is normal or low. The bone marrow thrombotic disease and bleeding. Aspirin
tion of hyperviscosity and platelet aspirate and trephine in PV show (75 mg/day) should be given unless
Polycythaemia 65

Table 32.1  Diagnostic criteria for


polycythaemia vera (PV)1
The diagnosis of PV requires (1) both major criteria and
one minor or (2) the first major and two minor criteria.
Major criteria
Significantly raised haematocrit2
Presence of JAK2 mutation
Minor criteria
1. Bone marrow trephine biopsy showing
hypercellularity for age and other features of
myeloproliferation
2. Low serum erythropoietin level Fig 32.5  Clubbing in a patient with
Fig 32.3  Gout complicating severe PV. 3. Endogenous erythroid colony formation cyanotic congenital heart disease and
secondary polycythaemia.
1
World Health Organization 2008.
2
Haemoglobin level or red cell mass may also be used.

essentially that of the underlying cause,


although cases with very high PCVs
Table 32.2  Causes of secondary may benefit from venesection.
polycythaemia
Hypoxia High altitude Idiopathic erythrocytosis
Hypoxic lung disease This is a small heterogeneous group of
Cyanotic congenital heart
patients who have an absolute poly-
disease (Fig 32.5)
Smoking
cythaemia without features of either PV
Abnormal Hb with increased or secondary polycythaemia. Venesec-
O2 affinity tion may be required.
Inappropriate Renal disease (e.g. tumours,
secretion of cysts, hydronephrosis) Apparent polycythaemia
erythropoietin Hepatoma
This condition has accumulated several
Cerebellar haemangioblastoma
Phaeochromocytoma names including spurious, stress or rela-
Fig 32.4  The face is a diagnostic clue in
Uterine fibroids tive polycythaemia, pseudopolycythae-
polycythaemia vera. Patients are frequently
plethoric and may have rosacea. Other Androgens mia and Gaisbock’s syndrome. The
Neonatal polycythaemia usual cause is an increase in red cell
Hypertransfusion mass and a decrease in plasma volume
contraindicated. The PCV is reduced Note: In practice hypoxia is by far the commonest cause. Renal within the normally accepted limits (see
below 0.45 by venesections (up to tumours are a rare cause but important to exclude. Neonatal Fig 32.1). Patients are most frequently
polycythaemia is discussed on page 90.
450 mL of blood removed) which may male and middle-aged. Other common
initially be required twice weekly. In characteristics are excess weight, hyper-
more severe disease the need for vene- tension, diuretic use and significant con-
section can become intolerable and Secondary polycythaemia sumption of alcohol and tobacco. The
cytotoxic drugs are used to suppress This is due to either a physiological adoption of a healthier lifestyle often
erythropoiesis. Hydroxycarbamide is response to hypoxia or an inappropriate leads to resolution of polycythaemia.
the usual choice. Interferon alfa can be secretion of erythropoietin. Causes are Venesection is not routine but is consid-
useful in younger patients and in preg- numerous and are listed in Table 32.2 ered where there are thrombotic risk
nancy. Busulfan is effective given inter- (and see Fig 32.5). Treatment is factors.
mittently but is best limited to older
patients as there is a significant risk of
secondary malignancy. Drug treatment
is particularly important when there is Polycythaemia
a need to control coexistent thrombocy-
tosis or progressive splenomegaly. JAK2 ■ Polycythaemia means an increase in haemoglobin and PCV above normally accepted limits.
inhibitors are under investigation. ■ Polycythaemia can be absolute (with an increased red cell mass) or apparent (with a
PV is a relatively benign disorder and normal red cell mass). The absolute form can be primary or secondary.
if well controlled is compatible with a ■ Polycythaemia vera is a myeloproliferative neoplasm associated with mutations in the JAK2
median survival of greater than 10 years. gene. Secondary polycythaemia arises from a physiological response to hypoxia or
However, it is a clonal disease and a few inappropriate secretion of erythropoietin.
patients eventually transform to mye- ■ Management of PV is by venesection alone or with cytotoxic drugs.
lofibrosis (10%) or even acute leukaemia ■ Treatment of secondary polycythaemia is essentially that of the underlying cause.
(5%). The risk of the latter is increased ■ Apparent polycythaemia may respond to adoption of a healthier lifestyle.
by treatment with alkylating agents.
66 6 MYELOPROLIFERATIVE NEOPLASMS

33 Essential thrombocythaemia and myelofibrosis


ET as opposed to a ‘reactive’ thrombocy- pluripotential stem cell. Abnormal meg-
Essential tosis. Even where there is an acquired akaryocytes are produced in increased
thrombocythaemia JAK2 gene mutation, other myeloprolif- numbers and it is these cells which
Essential thrombocythaemia (ET) is erative disorders must be excluded. release cytokines such as platelet-derived
a chronic myeloproliferative neoplasm Bone marrow examination is worth- growth factor (PDGF) and transforming
characterised by a persistent increase in while to exclude chronic myeloid growth factor-β, which stimulate fibrob-
platelet count. It is thought to be a clonal leukaemia (absence of Philadelphia last proliferation and build-up of colla-
stem cell disorder although recent chromosome), myelofibrosis or myelo- gen in the bone marrow. The scarred
studies suggest that it is heterogeneous. dysplasia, and to check iron stores. marrow is unable to function normally
Almost half of ET patients are positive Patients with polycythaemia vera may and haematopoietic stem cells move to
for the JAK2 V617F mutation (see p. 64). have thrombocytosis, while patients the spleen and liver (extramedullary
They appear to have distinct clinical fea- with ET can have an increased red cell haematopoiesis).
tures including a closer link to poly- mass. In practice such patients are better
cythaemia vera and a higher incidence diagnosed as having myeloproliferative
of thrombosis. ET may be associated neoplasms rather than forced into either
with either thrombotic or haemorrhagic category. Only about 5% of all raised
complications, the latter caused by platelet counts are due to ET, but per-
abnormal platelet function. The average sistence of the count above 1000, par-
age of presentation is around 60 years. ticularly with coexistence of thrombosis
The prognosis is generally good or haemorrhage, makes it the likely
although there is a risk of transforma- diagnosis. Abnormal platelet function
tion to myelofibrosis, polycythaemia tests suggest ET rather than a reactive
and acute myeloid leukaemia. thrombocytosis.

Clinical features Management


ET may be asymptomatic and discov- Management is not straightforward.
ered accidentally on routine blood The decision whether to treat at all must
testing. Symptoms commonly arise follow consideration of the patient’s age,
from disturbances of the microcircula- the degree of thrombocytosis and the
tion. Patients may complain of burning presence or perceived risk of significant
sensations in the soles and palms, cold thrombotic or haemorrhagic events.
peripheries and varied neurological Any clinical benefit must be weighed
symptoms including headache and against potential toxicity of cytotoxic
dizziness. Arteriolar occlusion can cause drugs. In a patient of more advanced age Fig 33.1  Blood film in essential
ischaemia, gangrene or acrocyanosis. (>60 years) or with a very high platelet thrombocythaemia showing increased
numbers of platelets of varying size.
Thrombosis of large arteries is of even count (>1500) or a history of throm-
greater concern. Haemorrhagic prob- boembolic disease, the treatment of
lems are less common but include choice is hydroxycarbamide and low-
ecchymoses, epistaxis, menorrhagia and dose aspirin. Anagrelide or interferon
bleeding into the mouth and gut. alfa may be preferred where hydroxy-
Splenomegaly is unusual at least in part carbamide is not tolerated. The objective
because of splenic infarction, which can of treatment is to maintain the platelet
be painful. count in the normal range and prevent
thrombosis and haemorrhage. Low-
Diagnosis dose aspirin alone is a reasonable option
Platelet counts can be as high as in patients at lower risk of these compli-
2000 × 109/L and usually exceed 450 ×  cations. Interferon is the usual drug of
109/L (the normal range is 150–400 ×  choice in pregnancy.
109/L) (Fig 33.1). In practice, there is no
single test to specifically identify ET –
Myelofibrosis
diagnosis is often a process of exclusion.
As thrombocytosis may accompany a Primary myelofibrosis is a myeloprolif-
wide range of disorders including infec- erative neoplasm characterised by bone
tions, inflammatory conditions, malig- marrow fibrosis and splenomegaly. It
nancy and iron deficiency, a thorough may develop de novo or in the setting of
history and examination is mandatory. polycythaemia vera or essential throm-
The lack of a measurable ‘acute phase bocythaemia. Most patients are over 50
Fig 33.2  Blood film in myelofibrosis
response’ (i.e. normal erythrocyte sedi- years. showing a myelocyte and nucleated red cell
mentation rate, plasma viscosity and Myelofibrosis is a neoplastic clonal (i.e. leucoerythroblastic film) and tear-drop
fibrinogen) increases the likelihood of disorder originating in a single poikilocytes.
Essential thrombocythaemia and myelofibrosis 67

(a)
Fig 33.4  Massive splenomegaly in myelofibrosis.

Leukaemic transformation occurs in about 15% of patients.


Asymptomatic patients may require no treatment. For anaemia
a trial of a corticosteroid, androgen or erythropoietin is worth-
while but regular transfusion is usually needed. Oral chemo-
therapeutic agents such as hydroxycarbamide may improve
quality of life by reducing systemic upset and shrinking the
spleen. There is abnormal bone marrow angiogenesis in mye-
lofibrosis and the anti-angiogenic agents thalidomide and
lenalidomide can improve blood counts and reduce splenom-
(b)
egaly with some durable responses. The JAK2 inhibitor rux-
olitinib is a promising new agent. Its major benefits are
Fig 33.3  Bone marrow trephine biopsy in myelofibrosis. (a) Marked reduced splenomegaly and constitutional symptoms. Other
fibrosis and osteosclerosis. (b) Increased reticulin fibres (stained by silver
JAK2 inhibitors are under investigation.
impregnation).
Splenic irradiation can alleviate splenic pain. Splenectomy
must not be undertaken lightly as it is associated with con-
Clinical features siderable mortality (around 5–10%). However, it is considered
The disease is often insidious in onset with fatigue and weight for painful splenomegaly, unacceptable transfusion require-
loss. Splenomegaly is present in all cases and massive in 10% ments, life-threatening thrombocytopenia, profound cachexia
(Fig 33.4). Splenic pain is common and a bulky spleen may or complications of portal hypertension.
lead to portal hypertension, bleeding varices and ascites. Allogeneic stem cell transplantation is the only potentially
Hepatomegaly is seen in two-thirds of cases. curative procedure. Use of reduced intensity conditioning
regimens (see p. 57) may allow its wider application.
Diagnosis
Anaemia is almost universal and the blood film shows tear-
drop poikilocytes and a ‘leucoerythroblastic’ picture (Fig 33.2).
In the early stages, thrombocytosis and neutrophilia may
occur but in more advanced disease low counts are the rule. Essential thrombocythaemia
Bone marrow aspiration characteristically results in a ‘dry tap’ and myelofibrosis
(i.e. only peripheral blood aspirated), and a marrow trephine
■ ET is a chronic myeloproliferative neoplasm characterised by a
showing dense reticulin fibres on silver staining, fibrosis and
persistent increase in platelet count.
osteosclerosis is needed for diagnosis (Fig 33.3). There is
■ Patients
with ET may be asymptomatic or have either thrombotic
usually megakaryocytic hyperplasia. The JAK2 gene mutation
or haemorrhagic complications.
is present in approximately 50% of cases. X-rays often show
bone sclerosis. The major differential diagnosis is from other ■ Patientswith ET at high risk of complications are usually treated
with hydroxycarbamide and low-dose aspirin.
myeloproliferative disorders and myelodysplastic syndromes
which may be associated with a degree of marrow fibrosis. ■ Myelofibrosis
is a myeloproliferative neoplasm characterised by
Systemic causes of marrow fibrosis such as marrow infiltra- bone marrow fibrosis and splenomegaly.
tion by carcinoma or lymphoma and disseminated tubercu- ■ Common symptoms in myelofibrosis are fatigue, weight loss and
losis should also be considered. splenic pain.
■ Treatment of myelofibrosis is problematic. Regular transfusion is
Prognosis and management often needed for anaemia. Cautious chemotherapy, splenic
irradiation and splenectomy can relieve symptoms in some
Average survival is 4–7 years but this is very variable. Manage-
patients. JAK2 inhibitors show promise.
ment is increasingly guided by prognostic scoring systems.
68 7 HAEMOSTASIS AND THROMBOSIS

34 Thrombocytopenia
Thrombocytopenia can be simply defined as a blood platelet menorrhagia are all relatively common, with haematuria and
count of below 150 × 109/L. With the routine measurement of melaena less frequent. Intracranial bleeding is of serious
platelet number by automated cell counters it is a relatively import but, thankfully, is rare. Possible examination findings
common laboratory finding. Before initiating further investi- include purpura and more extensive petechial haemorrhages
gations it is important to confirm that a low platelet count is involving the skin and mucous membranes (Fig 34.2). The
genuine by careful inspection of the blood sample and film. retina should be routinely inspected for haemorrhages.
Either a small clot in the sample or platelet clumping (Fig
34.1) can cause artefactual thrombocytopenia.
Clinical syndromes
Immune thrombocytopenia (ITP)
Causes ITP is a disease characterised by immune thrombocytopenia
Major causes of thrombocytopenia are listed in Table 34.1. mediated by platelet antibodies that accelerate platelet destruc-
Many of the diseases and syndromes are discussed tion and inhibit their production. It is a heterogeneous
elsewhere. disorder but it is conventional to divide it into two discrete
In general terms there are four possible processes leading entities: acute ITP and chronic ITP (Table 34.2). This division
to thrombocytopenia: is convenient for discussion of pathogenesis and apt for most
patients, but in ‘real life’ there is overlap between the two
■ Failure of marrow production. The bone marrow failure of
syndromes.
haematological disease (e.g. aplastic anaemia, leukaemia)
usually causes pancytopenia. However, thrombocytopenia
Acute ITP
may be the only sign of intrinsic marrow disease or
The acute form of the disease is usually seen in childhood. It
marrow suppression associated with infection or
typically has an abrupt onset a week or so following a trivial
chemotherapy.
viral illness. It is likely that in post-viral cases IgG antibody
■ Shortened lifespan. Platelets can be destroyed in the
attaches to viral antigen absorbed onto the platelet surface.
circulation. The most common mechanism is an
The resultant sudden fall in platelet count (often to below
immunological reaction in clinical syndromes such as
20 × 109/L) can lead to all the symptoms and signs quoted
immune thrombocytopenia.
above. Despite this, serious complications such as intracranial
■ Sequestration. Splenomegaly can cause low platelet counts
bleeding are very rare and the disease is self-limiting in around
because of pooling in the enlarged organ. The spleen is
90% of cases. Often only observation is required, but where
not necessarily massively enlarged.
the bleeding tendency is unusually severe, oral corticosteroids
■ Dilution. Normal platelets are diluted by massive blood
or intravenous immunoglobulin can be given as in chronic
transfusion.
ITP (see below). A few children go on to develop chronic
thrombocytopenia, but even here the disease is relatively
Clinical presentation benign and may eventually spontaneously remit.

Patients with thrombocytopenia are particularly prone to


bleeding from mucous membranes. It should be emphasised Table 34.1  Causes of thrombocytopenia
that spontaneous bleeding is usually only seen with platelet Pathogenesis Disease examples
counts of less than 10–20 × 109/L, although patients with Failure of production Leukaemia, myelodysplasia, aplastic anaemia,
associated platelet dysfunction may bleed at higher counts. megaloblastic anaemia, myelofibrosis, malignant
Conjunctival haemorrhage, nose and gum bleeding and infiltration, infection, drugs1
Shortened lifespan
Immune ITP, drugs1, connective tissue disorders, antiphospholipid
antibody syndrome, infection, post-transfusion
purpura, neonatal alloimmune thrombocytopenia
Non-immune DIC, thrombotic thrombocytopenic purpura
Sequestration Hypersplenism, cardiopulmonary bypass surgery
Dilution Massive blood transfusion

1
See Table 34.3.
ITP, immune thrombocytopenia; DIC, disseminated intravascular coagulation.

Table 34.2  Comparison of classic acute and chronic ITP


Characteristic Acute ITP Chronic ITP
Age Childhood Adult life
Previous viral infection Frequent Unusual
Platelet count (µ 109/L) Often <20 Variable
Onset Sudden Insidious
Duration Few weeks Years/lifelong
Spontaneous remission Around 90% Rare
Fig 34.1  Blood film showing clumping of platelets. This phenomenon
causes an artefactual thrombocytopenia in the automated blood count. ITP, immune thrombocytopenia.
Thrombocytopenia 69

Table 34.3  Some drugs associated new agents are introduced. The mono-
with thrombocytopenia clonal antibody rituximab may give
Heparin Penicillin durable responses. Second-generation
Quinine/quinidine Diazepam thrombopoietin receptor (TPO-R) ago-
Gold salts Tolbutamide nists stimulate platelet production via
Sulphonamides Aspirin megakaryocyte proliferation and matu-
Thiazides Cephalosporins ration. Two TPO-R agonists in current
Rifampicin Ranitidine use are romiplostim and eltrombopag.
There remains a need for other
approaches including relatively non-
toxic doses of corticosteroids (e.g. pred-
Fig 34.2  Purpuric rash in a patient with other causes of isolated thrombocytope- nisolone 10 mg), pulsed high dose
acute ITP. nia such as connective tissue disorders corticosteroids, intermittent IVIg,
and antiphospholipid antibody syn- danazol, vinca alkaloids, ciclosporin, aza-
drome. Apparent ‘primary’ ITP may be thioprine and mycophenolate. All give
secondary to subclinical viral infections some responses reflecting the heteroge-
such as hepatitis C, cytomegalovirus, neity of the disease.
HIV and Helicobacter pylori. In younger
patients congenital thrombocytopenias Drug-induced thrombocytopenia
may be confused with ITP. A thorough Many drugs have been linked with iso-
drug history is essential. lated thrombocytopenia (Table 34.3).
Patients with asymptomatic mild The mechanism is usually the forma-
thrombocytopenia can be merely tion of antiplatelet antibodies. General
observed. It is difficult to state a platelet management is withdrawal of the
count below which treatment is manda- offending drug and platelet transfusion
tory. In practice, serious bleeding is rare for significant bleeding.
even at lower platelet counts and drug Heparin-induced thrombocytopenia
side-effects are common so treatment (HIT) is an immune-mediated disorder
should generally be reserved for patients caused by the development of antibod-
who have symptoms or signs. The ies to platelet factor 4 and heparin.
Fig 34.3  Blood film in ITP. The platelets are normal first-line treatment is pred- The thrombocytopenia is typically
reduced in number and increased in size.
nisolone (1 mg/kg body weight). About non-severe and occurs 5–10 days after
two-thirds of patients have a significant starting heparin. Unlike other drug-
Chronic ITP increase in platelet count within weeks induced thrombocytopenias, HIT leads
There has been recent change in our but subsequent dose reduction often to increased risk of thromboembolism.
understanding of the pathophysiology leads to relapse. Where there is no Heparin should be stopped and an alter-
of chronic ITP. Antibodies that mediate response to steroids, immunoglobulin native anticoagulant substituted.
platelet destruction also impact platelet (IVIg) can be efficacious. Platelet trans-
production by damaging megakaryo- fusions are seldom indicated as the Post-transfusion purpura
cytes and/or blocking the release of platelets are rapidly destroyed but In this very rare syndrome severe throm-
proplatelets. A few cases may not they may be considered in severe bocytopenia develops approximately 1
be antibody-mediated and will not haemorrhage. week after a blood transfusion. In most
respond to standard immunosuppres- If the platelet count cannot be ade- cases the patient’s platelets are negative
sive therapies. quately maintained on non-toxic doses for the platelet antigen HPA-1a and the
Chronic ITP is most common in adult of corticosteroid then splenectomy is transfused platelets are HPA-1a positive.
life. Patients may be asymptomatic or considered. About two-thirds of patients In a way incompletely understood an
have insidious onset of bleeding prob- have a good response. The management anti-HPA-1a isoantibody destroys the
lems. Serious spontaneous bleeding is of severe/symptomatic thrombocytope- patient’s own platelets. Bleeding may be
generally limited to platelet counts nia post-splenectomy is improving as severe. IVIg is an effective treatment.
below 10 × 109/L and even then it is
unusual. Fatigue is common. Paradoxi-
cally, it appears that the disorder may
have a pro-thrombotic element where Thrombocytopenia
platelet counts are restored to normal.
■ Thrombocytopenia (a low platelet count) is a relatively common laboratory finding. It is
A palpable spleen suggests a diagnosis
important that it is confirmed by inspection of a blood film.
other than ITP.
■ In general thrombocytopenia can be caused by failure of marrow production, shortened
The blood film confirms thrombocy-
platelet lifespan, sequestration in the spleen and dilution by massive blood transfusion.
topenia; often the platelets are increased
in size (Fig 34.3). There is no single spe- ■ Immune thrombocytopenia (ITP) is a heterogeneous disease characterised by platelet
antibodies which accelerate platelet destruction and inhibit their production.
cific test for ITP. A bone marrow aspirate
and trephine biopsy will show increased ■ Acute ITP is usually seen in childhood and is typically self-limiting. Chronic ITP classically
megakaryocytes but it is often not neces- occurs in adult life. There is often an initial response to steroid treatment but splenectomy
may ultimately be required. Newer agents include rituximab and thrombopoietin receptor
sary if other features are typical. Further agonists.
investigations are designed to exclude
70 7 HAEMOSTASIS AND THROMBOSIS

35 Disorders of platelet function and


vascular purpuras

Platelet dysfunction should be consid- Addition


ered wherever there are the clinical of agonist
ADP Collagen Adrenaline
symptoms and signs of thrombocytope-
nia (p. 14) in the presence of a normal
or only moderately reduced platelet

Increasing light transmission


count. Disorders of platelet function can

Optical density
be divided into inherited disorders
which are rare but well characterised in
the laboratory, and acquired disorders
which are much more common but
often of obscure aetiology. Bleeding
problems may also arise in a number of
inherited and acquired disorders of the
vasculature and its supporting connec-
Normal platelets Glanzmann's thrombasthenia
tive tissue – the vascular purpuras.
Fig 35.1  Platelet aggregation studies. When compared with the normal control it can be seen
that in Glanzmann’s thrombasthenia there is loss of aggregation with all the agonists used.

Laboratory testing of
agonists except ristocetin (see Fig 35.1).
platelet function Inherited disorders of Clinical manifestations are variable but
Ideally, blood samples for testing of
platelet function there is typically onset in the neonatal
platelet function should be taken from The commonest inherited platelet func- period and subsequent cutaneous and
fasting and resting subjects who have tion and coagulation disorder, von Wil- gastrointestinal bleeding, and menor-
not smoked, ingested caffeine or drugs lebrand disease, is described on page 74. rhagia. Platelet transfusions are indi-
known to affect platelet function. A cated where local haemostatic measures
blood count and blood film are rou- Bernard–Soulier syndrome fail. If there is platelet refractoriness,
tinely performed. The bleeding time, This is a rare autosomal recessive bleed- recombinant factor VIIa can be used.
where a small incision is made in the ing disorder. There is a combination of
forearm skin and the time to cessation platelet dysfunction, thrombocytopenia Other disorders
of bleeding recorded, is now less used and abnormal platelet morphology. Hereditary diseases of platelet function
as it is subjective and has poor reproduc- The mild thrombocytopenia is probably may also result from deficiency of plate-
ibility. A number of dedicated platelet caused by reduced platelet survival. The let storage organelles (storage pool dis-
function instruments (e.g. PFA-100) functional platelet defect arises from orders) or release defects where there is
allow screening tests but the results mutation in the polypeptides of the failure to successfully release granule
must be interpreted with caution as they glycoprotein (GP) Ib/IX/V complex. This contents upon platelet activation. These
are not diagnostic or sensitive for mild complex is crucial for the initial adhe- disorders usually cause only mild bleed-
platelet disorders. sion of platelets to exposed subendothe- ing problems.
Platelet aggregation studies assess lium at high shear flow and for binding
the ability of platelets to aggregate in of platelets to von Willebrand factor.
response to the addition of a variety of
Acquired disorders of
In platelet aggregation studies there is
agonists (e.g. ADP, adrenaline (epine-
platelet function
failure to aggregate with ristocetin.
phrine), collagen, arachidonic acid, ris- Bleeding can be severe and particularly These disorders are common. Causes
tocetin). The tracings produced (Fig complicates other predisposing events include foods, drugs, systemic disorders
35.1) require expert interpretation. The such as peptic ulcers and pregnancy. and diseases of the blood (Table 35.1).
methodology remains the gold standard Patients require platelet transfusion for
with the response to agonists giving severe bleeding and prior to surgery. Aspirin
characteristic patterns in inherited disor- Antifibrinolytic agents and DDAVP (see Many drugs can affect platelet function
ders. Other tests of platelet function p. 73) are useful in some cases. but aspirin is the best documented and
include flow cytometry for the quantita- the most frequently prescribed. At lower
tion of glycoprotein receptor density, Glanzmann’s thrombasthenia doses aspirin selectively acetylates and
and the measurement of total and/or This rare autosomal recessive disease is irreversibly inactivates the enzyme
released adenine nucleotides. The latter also caused by loss or dysfunction of a cyclooxygenase-1 (COX-1), preventing
tests may confirm the findings from platelet glycoprotein complex – GP IIb/ the production of thromboxane A2 from
platelet aggregation studies (e.g. in IIIa. This normally acts as a receptor for arachidonic acid and inhibiting aggrega-
Bernard–Soulier syndrome) or reveal adhesive proteins such as fibrinogen tion for the remainder of the platelet’s
abnormalities where aggregation studies and von Willebrand factor. Platelet lifespan. Responses are variable but
are normal or equivocal (e.g. in a storage numbers and morphology are normal aspirin can dramatically prolong the
pool disease or release defect). but the platelets fail to aggregate with all bleeding time and cause haemorrhage
Disorders of platelet function and vascular purpuras 71

in patients with thrombocytopenia or Table 35.1  Causes of abnormal


other coexistent bleeding problems. platelet function
Inherited Bernard–Soulier syndrome
Chronic renal failure Glanzmann’s thrombasthenia
Uraemia can lead to multiple platelet Storage pool disorders
Release defects
defects. Elevated levels of nitric oxide
Other (e.g. von Willebrand disease)
may inhibit platelet adhesion, activation
Acquired Drugs (e.g. aspirin)
and aggregation. Anaemia contributes Foods (e.g. garlic)
to uraemic bleeding as fewer red cells Chronic renal failure
mean that fewer platelets are displaced Cirrhosis
towards an injured vessel wall. Dialysis Cardiopulmonary bypass surgery
Blood diseases: acute myeloid
reduces haemorrhagic symptoms. This
leukaemia, myelodysplastic syndromes,
can be supplemented by correction of myeloproliferative disorders, myeloma
severe anaemia, DDAVP and platelet Various systemic disorders1
transfusions. 1
These include disseminated intravascular coagulation (DIC) and
thrombotic thrombocytopenic purpura (TTP).
Cardiopulmonary bypass
Platelets are activated and degranulated Table 35.2  The vascular purpuras
in the extracorporeal circuit, impairing Inherited Hereditary haemorrhagic telangiectasia1
their effectiveness in vivo. This may be Connective tissue diseases Fig 35.2  Telangiectasia in hereditary
exacerbated by hypothermia and large Ehlers–Danlos syndrome haemorrhagic telangiectasia.
doses of heparin. Excessive bleeding is Pseudoxanthoma elasticum
Marfan syndrome
uncommon but where this happens
Acquired Henoch–Schönlein purpura
platelet transfusion is efficacious.
Various infections
Drug reactions (allergic purpuras)
Haematological diseases Senility
Platelet function is impaired in a number Prolonged corticosteroid treatment
of blood diseases, including acute Scurvy
Mechanical
myeloid leukaemia, myelodysplastic
syndromes, myeloproliferative disorders 1
Sometimes known as Rendu–Osler–Weber disease.
and myeloma.
Inherited diseases of
connective tissue
Vascular purpuras Several rare inherited disorders of con-
A bleeding tendency caused by a local nective tissue predispose to bleeding.
or general vascular abnormality is The mechanism is either a general
referred to as a vascular purpura (Table failure of support of blood vessels or
35.2). Diagnosis of these diseases is defective interaction between platelets
made mainly on clinical grounds with and abnormal collagen. Specific diseases
laboratory exclusion of other haemo- include Ehlers–Danlos syndrome, pseu-
static defects. doxanthoma elasticum and Marfan
syndrome. Fig 35.3  Senile purpura.
Inherited disorders
Hereditary haemorrhagic Acquired disorders the rule but renal failure may result.
telangiectasia (HHT) This is a very heterogeneous group. Other causes of acquired purpuric
The hallmark of this rare autosomal Henoch–Schönlein purpura is a syn- rashes include infections, drug reac-
dominant disease is the development of drome usually seen in childhood where tions, scurvy, trauma, prolonged steroid
multiple ateriovenous malformations an itchy purpuric rash typically follows therapy and simple old age (senile
(AVMs). Small AVMs are referred to as an infection. Spontaneous remission is purpura, Fig 35.3).
telangiectasia. Close to the surface of the
skin and mucous membranes, they
often rupture and bleed. Two mutated Disorders of platelet function
genes, endoglin and ALK1, have been and vascular purpuras
implicated. Clinical problems include
■ Platelet
dysfunction should be considered where there are the clinical features of
recurrent epistaxes (90% of cases), gas-
thrombocytopenia in the presence of a normal or only moderately reduced platelet count.
trointestinal haemorrhage, haematuria
■ Laboratory testing of platelet function normally includes a blood count, a blood film and
and larger pulmonary arteriovenous
platelet aggregation studies. Careful collection of the sample is crucial.
malformations (PAVMs). Chronic bleed-
■ Inheriteddisorders of platelet function are generally well characterised but rare (e.g.
ing from the gut causes iron deficiency
Bernard–Soulier syndrome), whereas acquired disorders are more frequent but often of
anaemia. On examination there are the obscure aetiology.
characteristic telangiectasia (Fig 35.2).
■ Aspirin is a common cause of acquired platelet dysfunction.
Management includes local control of
■ A ‘vascular purpura’ is a disorder with a bleeding tendency caused by a local or general
bleeding (e.g. laser treatment of tel-
vascular abnormality. Diseases may be inherited (e.g. hereditary haemorrhagic
angiectasia), iron supplements and
telangiectasia) or acquired (e.g. Henoch–Schönlein purpura).
embolisation of PAVMs.
72 7 HAEMOSTASIS AND THROMBOSIS

36 Haemophilia
Haemophilia is an inherited disorder of Table 36.1  Factor VIII level and clinical
coagulation. The general term haemo- severity of haemophilia
philia is usually taken to mean haemo- Factor VIII level Clinical severity
philia A, a deficiency of factor VIII, but Less than 2 units/dL Severe: frequent spontaneous
a smaller number of cases are caused by bleeds into joints and muscles
a deficiency of factor IX (haemophilia 2–5 units/dL Moderate: some spontaneous
bleeds, bleeding after minor
B).
trauma
5–45 units/dL Mild: bleeding only after
Haemophilia A significant trauma or surgery

Haemophilia A is transmitted as an can compress adjacent nerves and


X-linked recessive disorder. Thus, all vessels with serious consequences (Fig
males with the defective gene have hae- 36.2). Haematuria is not unusual and,
mophilia, all sons of haemophiliac men until recently, intracranial bleeding was
Fig 36.2  Psoas muscle bleed in haemophilia
are normal, all daughters are obligatory the most common cause of death in A. There was sensory loss in the outlined area
carriers and daughters of carriers have a haemophilia. caused by pressure on the femoral nerve.
50% chance of also being carriers. The
disease prevalence is 1 in 10 000 people. Complications of treatment performed on umbilical cord blood. In
The gene for factor VIII is situated at the In affluent countries, factor VIII replace- the absence of a family history the
tip of the long arm of the X chromo- ment treatment as described below has disease may present in a young child
some. A wide variety of mutations of the been enormously beneficial in allowing with bruising and a swollen joint and be
gene can lead to underproduction of early control of bleeding and the avoid- mistakenly regarded as non-accidental
factor VIII and the clinical syndrome of ance of chronic joint damage. Unfortu- injury. Mild haemophilia may only
haemophilia. In about half of haemo- nately, most haemophiliac patients cause problems after trauma or surgery.
philia families an unusual molecular treated before 1985 became infected All patients with bleeding or bruising of
genetic abnormality involving inversion with pathogenic viruses contaminating a severity disproportionate to the trauma
of the factor VIII gene at intron 22 has factor VIII concentrate, notably HIV sustained should be investigated to
been found. A family history is not inevi- and hepatitis C. There are now improv- exclude a bleeding disorder.
tably present, as up to 30% of all new ing therapies for both HIV and hepatitis
cases of haemophilia are due to recent C infection and younger patients receiv- Management
sporadic mutations. ing only virus-free factor products have Treatment of haemophilia is complex,
avoided these complications. Approxi- and severe disease is best managed in
Clinical features mately 25% of patients with severe hae- haemophilia centres where an experi-
As factor VIII is a critical component of mophilia will develop antibodies to enced team of doctors, nurses, physio-
the blood coagulation pathway (see p. factor VIII (‘inhibitors’). They tend to therapists and social workers can help
12), low levels predispose to recurrent appear in childhood but may occur after patients and their families to lead a rela-
bleeding. The likelihood of bleeding can years of treatment. tively normal life.
be roughly predicted from the factor
VIII level, which may be expressed as Diagnosis Treatment of bleeding
units/dL or as percentage activity (Table Haemophilia is associated with a pro- Most haemophiliac patients require
36.1). longed activated partial thromboplastin replacement therapy with factor VIII
time (APTT) in the routine clotting concentrate and this is often self-
Bleeding in haemophilia screen. The diagnosis is confirmed by a administered at home when a bleed
The disease usually becomes apparent factor VIII assay. In the presence of a occurs (‘on demand’ treatment). The
when the child begins to crawl. Severely family history there are usually few
affected patients not receiving prophylac- problems in diagnosis. Tests can be
tic treatment experience 30–50 bleeding
episodes each year. The most common
problems are spontaneous bleeds into
joints, often elbows or knees, although
any joint can be involved. Patients may
develop particular target joints which
bleed frequently. They often have an
innate feeling that a bleed has started
prior to any objective signs. Recurrent or
inadequately managed joint bleeds lead
to chronic deformity of the joint with
swelling and pain (Fig 36.1).
Bleeding may also afflict deep-seated (a) (b)
muscles, often the flexor muscle groups. Fig 36.1  Chronic knee damage in severe haemophilia A. (b) Shows bilateral osteoporosis,
If ignored, the enlarging haematoma narrowing of the joint space and joint deformity.
Haemophilia 73

Plasma factor VIII


level (%)
100

80

60

40
Fig 36.4  Southern blotting illustrating the
factor VIII gene inversion. Lane 1, normal male;
20 Lane 2, female heterozygous for proximal
inversion; Lane 3, male with distal inversion; Lane
4, female heterozygous for distal inversion; Lane
0 5, normal female.
0 12 24 Hours

3500 u 1750 u levels of factor VIII (10–30%) to cause


Factor VIII Factor VIII excessive bleeding after trauma. In fami-
Fig 36.3  Typical response to factor VIII infusion in a patient with severe haemophilia. lies with inversion of the factor VIII
An infusion of 3500 units will increase the level to around 100% in a 70 kg man. As factor VIII has a gene (see above), first-generation molec-
half-life of 12 hours, the level falls to 50% at this time – an infusion of 1750 units increases the level ular biology methods have been used in
from 50 to 100%.
carrier and prenatal diagnosis (Fig 36.4).
dose and duration of treatment depends Likely advances in haemophilia drug The more recent development of
on the patient’s size and the locality and therapy are pegylated recombinant polymerase chain reaction (PCR)-based
magnitude of the bleed. One unit of factor VIII allowing less frequent admin- screening and sequencing technology
factor VIII is the amount contained in istration and novel approaches includ- has allowed identification of the muta-
1 mL of normal plasma. For spontane- ing non-peptide haemostatic agents tion in nearly all patients with haemo-
ous haemarthroses it is sufficient to which reduce reliance on replacement philia A. Large databases of the known
raise the factor VIII level to 30% of coagulation factor. mutations are freely available.
normal; in a 70 kg man this entails a
Treatment of viral infection
dose of around 1000 units. More serious Haemophilia B
bleeding or surgery requires levels of Haemophiliac patients with HIV infec-
70–100% maintained until the risk sub- tion require state-of-the-art manage- Haemophilia B is an X-linked recessive
sides (Fig 36.3). Factor VIII products ment of the physical and social problems bleeding disorder in which there is a
undergo processing to maximise quality, which can arise. Hepatitis C infection deficiency of factor IX. There are many
purity and viral safety. Plasma-derived carries a long-term risk of cirrhosis (20– clinical similarities to haemophilia A –
factor VIII is being increasingly replaced 30%) and hepatocellular cancer. The severely affected patients suffer recur-
by recombinant factor VIII. Third- combination of pegylated interferon rent spontaneous joint bleeds. However,
generation recombinant factor VIII is and ribavirin eradicates the virus in 50– inhibitors (antibodies to factor IX) are
free of any animal or human protein. 80% of cases dependent on the viral less common than in haemophilia A.
Prophylactic (alternate day) recom- genotype. Earlier factor IX concentrates were asso-
binant factor VIII treatment in children ciated with thromboembolic complica-
Gene therapy tions but safer high purity preparations
eradicates bleeding and improves quality
Gene therapy is a potentially curative and recombinant products are now
of life. Periods of ‘secondary prophylaxis’
treatment for haemophilia and is dis- available for treatment. The half-life of
may be considered in older patients
cussed on page 103. infused factor IX is around 18 hours and
with problematic joint bleeds.
Treatment of inhibitors is highly spe- thus it can often be given just once daily
The carrier state and
cialised. Acute bleeds can be treated with to maintain levels after spontaneous
genetic counselling
recombinant factor VIIa or a concentrate bleeding or surgery. Prophylactic treat-
Female carriers are generally asympto-
of activated vitamin K derived clotting ment can be given once or twice weekly.
matic but some will have low enough
factors (FEIBA). Eradication of the inhib-
itor may be achieved by immune toler-
Haemophilia
ance regimens where factor VIII is given
regularly in high doses with or without ■ Haemophilia A is an X-linked recessive disorder characterised by deficiency of factor VIII.
immunomodulatory agents. ■ Severely affected patients suffer recurrent spontaneous bleeds, most often into joints.
In patients with mild disease, 1-amino- ■ Replacement therapy with factor VIII concentrate is needed in
8-D-arginine vasopressin (DDAVP), all but mild cases; previous contamination of plasma-derived concentrates has led to HIV
given intravenously or by nasal spray, and hepatitis C infection.
mobilises factor VIII from stores and ■ DDAVP and tranexamic acid can help control bleeding in mild disease.
may avoid the need for concentrate. The
■ Themanagement of choice in severely affected children is prophylactic treatment with
antifibrinolytic agent tranexamic acid genetically engineered recombinant factor VIII.
can also be used to reduce bleeding – it
■ Haemophilia B is characterised by deficiency of factor IX; inheritance and clinical features
should, however, be avoided in haema- are similar to haemophilia A.
turia where it can induce clot colic.
74 7 HAEMOSTASIS AND THROMBOSIS

37 Von Willebrand disease and other inherited


coagulation disorders
Suspected VWD
Von Willebrand disease
e.g. abnormal coagulation screen,
Von Willebrand disease (vWD) is the most common inherited family history of VWD,
bleeding symptoms
bleeding disorder. The prevalence of symptomatic vWD is
approximately 0.01%. Identification of milder forms is com-
plicated by the broad range of von Willebrand factor (vWF)
levels in the normal population; it is important to recognize Bleeding score normal
that the diagnosis of vWD requires the presence of bleeding Patient evaluation and
Not VWD1
bleeding score
symptoms (see Fig 37.1).
All vWD is caused by mutations in the gene for vWF. vWF Bleeding score
abnormal
is an adhesive glycoprotein secreted by endothelium and meg-
akaryocytes (see also p. 12). It is a multimeric protein with a Normal
Evaluate for other
Measurement of
characteristic normal distribution of multimer sizes in plasma. VWF levels bleeding disorders
vWF has two key functions: promotion of platelet adhesion
to damaged endothelium and other platelets (Fig 37.2) and the Low

transport and stabilisation of factor VIII. Thus, the clinical


disorder of vWD is associated with excessive bleeding due to Probable VWD

abnormal platelet function and low factor VIII activity. The further testing: e.g. FVIIIc, RIPA,
relationship between the risk of bleeding and vWF level is not multimers, DDAVP infusion test
strong until the level is very low. The clinical and laboratory
heterogeneity of vWD necessitates the definition of several Fig 37.1  One approach to vWD diagnosis. In practice, this has to be
subtypes. individualised (see text). vWD, von Willebrand disease; vWF, von Willebrand
factor; FVIIIc, factor VIII; RIPA, ristocetin-induced platelet aggregation.
1
In children should have lower threshold for measuring vWF levels.
Classification (Table 37.1 and Fig 37.3)
Platelet GP
The current classification of vWD depends on electrophoretic IIIa
GP
analysis of vWF multimers. In type 1 vWD, the multimers GP IIb
Membrane Ib
appear to be normal in structure and function but decreased
in concentration. In type 2 vWD there is a qualitative defi-
ciency of vWF divisible into four subtypes. In type 2A there
is an absence of high molecular weight vWF multimers and vWF
Subendothelial
markedly reduced vWF binding to platelets. 2B refers to a microfibrils
variant where defective platelet adhesion results, paradoxi-
cally, from increased binding of vWF to platelets. In 2M there Vessel
is decreased platelet-dependent vWF function despite a rela- wall
tively normal multimer pattern while 2N is characterised by Collagen
failure of vWF to bind factor VIII. In the rare type 3 form, Fig 37.2  The role of von Willebrand factor in platelet adhesion.
there is an almost complete deficiency of vWF and the factor Following vessel wall injury, large multimers of vWF bind to subendothelial
VIII level is markedly decreased. microfibrils and also to glycoprotein (Gp) Ib on the platelet membrane thus
There is correlation between the subtype and the mode of mediating platelet adhesion. A secondary binding site with platelet Gp IIb/
inheritance. Type 1 vWD is the most common form of the IIIa promotes further adhesion.
disease (80% of cases) and inheritance is often autosomal
dominant. Type 2 vWD (15% of cases) may be dominant or
recessive and the type 3 variant is recessive. Because inherited
deficiencies of vWF function are common the accidental
co-inheritance of otherwise recessive vWD alleles may occur
(‘compound heterozygosity’). There is currently no genotypic Large
multimers
classification of vWD. More than 250 mutations of all Intermediate
types have been identified. These include large and small multimers
deletions, nonsense and missense mutations and splicing Small
abnormalities. multimers

Normal Type 1 Type 2A Type 2B Type 3


Clinical features Fig 37.3  SDS–PAGE multimer analysis of von Willebrand factor.
Severe vWD is characterised by spontaneous bleeding, par- Typical multimer patterns in normal plasma and types 1, 2A, 2B and 3 vWD
are shown diagrammatically.
ticularly epistaxes, gum bleeding and menorrhagia. Easy
bruising is also common but (with the exception of type 3) missed. A thorough history is crucial and must include assess-
haemarthroses and muscle haematomas are rare. Milder ment of the severity of recent bleeding, the existence of previ-
disease often presents with excessive bleeding following ous bleeding problems (particularly after surgery, dental
trauma or surgical procedures and the diagnosis can easily be extractions and childbirth) and the presence of a family
Von Willebrand disease and other inherited coagulation disorders 75

Table 37.1  Summary of classification now little used. The PFA-100 (see p. bleeding include factor XI concentrate
of vWD 70) is a useful screening test but is and recombinant factor VIIa.
■ Type 1 vWD is a partial quantitative deficiency of also abnormal in other platelet
vWF disorders. Factor VII deficiency
■ Type 2 vWD is a qualitative deficiency of vWF This is inherited as an autosomal reces-
■ Type 3 vWD is a virtually complete deficiency of Management sive disorder. The bleeding tendency is
vWF Very mild bleeding problems may very variable with central nervous system
■ Type 2A vWD is a qualitative variant with an require little intervention, perhaps just
absence of high molecular weight vWF multimers
haematoma a real risk in severe cases.
local measures and the prescription of The diagnosis is confirmed by factor VII
■ Type 2B vWD is a qualitative variant with increased
affinity of vWF for platelet glycoprotein lb (reduced an antifibrinolytic drug such as tran- assay and recombinant factor VII con-
in other types) examic acid. More significant bleeding centrate is available for treatment.
■ Type 2M vWD is a qualitative variant not caused by generally responds to an infusion or
absence of high molecular weight multimers intranasal spray of DDAVP (desmo- Factor V deficiency
■ Type 2N vWD is a qualitative variant with reduced
pressin) which stimulates release of This is a very rare autosomal recessive
affinity of vWF for factor VIII
vWF from stores. DDAVP is predictably condition. Bleeding episodes are treated
Note: Mixed phenotypes may be caused by compound most effective in patients with a partial with virally inactivated fresh frozen
heterozygosity.
quantitative impairment of vWF (type plasma.
1). It is less effective in most type 2 vari-
history of easy bleeding. A standard ants and is generally contraindicated in Factor XIII deficiency
questionnaire can be used to generate a type 2B. Patients with type 3 disease do Another rare autosomal recessive disor-
quantitative ‘bleeding score’. Death from not respond to DDAVP as they lack any der, factor XIII deficiency causes a
bleeding is rare but it may follow capacity to secrete vWF. Where DDAVP severe haemorrhagic tendency and poor
massive gastrointestinal haemorrhage. is ineffective or contraindicated, then wound healing. Most sufferers present
selected plasma-derived factor VIII con- early in life, often with profuse bleeding
Laboratory diagnosis centrates containing sufficient vWF are from the umbilical cord, and death may
Diagnosis can be complicated and tests used. An unusually sustained rise in result from intracranial haemorrhage.
often have to be repeated. It is not clear factor VIII levels can be obtained as the Screening coagulation tests are normal.
which laboratory measurement best vWF in the concentrate prolongs sur- Diagnosis requires the laboratory dem-
correlates with the severity of bleeding. vival of the patient’s own factor VIII. onstration of solubility of patient plasma
Recombinant vWF is under develop- clots in urea (there is defective cross-
1. Blood count. The platelet count is linking of fibrin). Factor XIII concen-
ment. Patients with vWD normally
normal except for a moderate trate is available for treatment.
require treatment with either DDAVP or
reduction in some cases of type 2B
factor VIII concentrate prior to surgery.
disease. Abnormalities of fibrinogen
Effective genetic counselling in
2. Activated partial thromboplastin Inherited disorders of fibrinogen are
vWD demands a full understanding
time (APTT). Usually prolonged broadly divisible into quantitative
of the disease subtype and mode of
due to low factor VIII : C levels. The deficiencies (afibrinogenaemia and
inheritance.
prothrombin time (PT) is normal. hypofibrinogenaemia) and qualitative
3. Quantitative immunoassay for abnormalities (dysfibrinogenaemia).
vWF antigen. Other inherited Afibrinogenaemia is an autosomal
4. Functional assay of vWF. The coagulation disorders recessive disease in which blood fails
commonest methodology is the to clot in all coagulation screening
Factor deficiencies
‘ristocetin cofactor assay’. Collagen tests and plasma fibrinogen is barely
Factor VIII and factor IX deficiencies
binding assays are also used. detectable by radioimmunoassay. The
See section on haemophilia (pp. 72–73).
5. Factor VIII : C assay. Often low. bleeding tendency can be severe with
May be borderline or normal in Factor XI deficiency spontaneous haemorrhage and exces-
mild type 1 disease. This bleeding disorder occurs most sive blood loss after surgery. Fibrinogen
6. Multimer analysis. The multimer commonly in Ashkenazi Jews. There is concentrate is the treatment of choice.
composition of circulating VWF is a variable relationship between the Many patients with hypofibrinogenae-
assessed by either crossed factor XI level and the bleeding ten- mia and dysfibrinogenaemia are
immunoelectrophoresis or sodium dency. Treatment options for significant asymptomatic.
dodecyl sulphate electrophoresis
(see Fig 37.3). Von Willebrand disease and
7. Platelet aggregation studies. other inherited coagulation
Ristocetin (an obsolete antibiotic) disorders
induces platelet aggregation in
■ vWD is a relatively common and very heterogeneous inherited bleeding disorder.
normal plasma but not in severe Deficiency of von Willebrand factor (vWF) causes abnormal platelet function and low factor
vWD. An exception is the type 2B VIII activity.
variant where platelets aggregate at ■ Classification of vWD relies on electrophoretic analysis of vWF multimers.
unusually low concentrations of
■ Mild bleeding problems in vWD require little intervention. More significant bleeding is
ristocetin. treated with either DDAVP or factor VIII concentrates containing vWF.
8. Blood group. Normal plasma vWF
■ Thereare various other inherited coagulation factor deficiencies. In most there are specific
levels tend to be lower in group O
concentrates available for treatment.
individuals.
■ Inherited disorders of fibrinogen include quantitative deficiencies (afibrinogenaemia and
9. General tests of primary
hypofibrinogenaemia) and qualitative abnormalities (dysfibrinogenaemia).
haemostasis. The bleeding time is
76 7 HAEMOSTASIS AND THROMBOSIS

38 Acquired disorders of coagulation

Disseminated intravascular
coagulation (DIC) Trigger

DIC is a complex clinical syndrome which complicates many


serious illnesses (Table 38.1). It is characterised by intravascular
deposition of fibrin and accelerated degradation of fibrin and
fibrinogen caused by excess activity of proteases, notably
thrombin and plasmin, in the blood (Fig 38.1). DIC is hetero- Tissue factor Cytokine Plasminogen
geneous both in its pathophysiology and clinical manifestations. release release activator release
In most cases it probably begins when circulating blood is
exposed to tissue factor released from damaged tissues, malig-
Thrombin Plasmin
nant cells or injured endothelium. This in turn leads to genera- generation generation
tion of thrombin which causes formation of soluble fibrin,
activation of circulating platelets, and secondary fibrinolysis. Thrombocytopenia
DIC can cause bleeding, large vessel thrombosis and haem-
orrhagic tissue necrosis (Fig 38.2). The coagulation defect Platelet Fibrinolysis
activation Fibrin
arises from consumption of coagulation factors and platelets
and increased fibrinolytic activity. In clinical practice acute
DIC usually presents as widespread bleeding in an ill patient.
Oozing of blood from cannulation sites is characteristic. Thrombosis Bleeding
Microthrombus formation can lead to irreversible organ
Tissue
damage; the kidney, lungs and brain are frequent targets. DIC necrosis
is not necessarily a fulminant syndrome; more chronic forms
may be seen particularly in association with malignancy (e.g. Fig 38.1  Pathophysiology of DIC. A simplification of the complex
interactions.
prostatic carcinoma).
Diagnosis depends on the laboratory
demonstration of accelerated fibrinoly-
sis accompanied by falling levels of
coagulation factors in an ill patient. The
following combination of laboratory test
abnormalities is typical:
■ reduced platelet count
■ prothrombin time (PT) prolonged
and activated partial thromboplastin
time (APTT) usually prolonged
■ thrombin time prolonged
■ fibrinogen level reduced
■ high levels of fibrin(ogen) Fig 38.3  Spontaneous bruising in acquired
haemophilia.
degradation products (FDPs) and
cross-linked fibrin degradation
Table 38.1  Common causes of DIC
products (‘D-dimers’).
■ Infections – particularly septicaemia
The cornerstone of management of DIC ■ Malignancy – disseminated carcinoma or acute
leukaemia
is the treatment of the underlying
■ Obstetric emergencies – septic abortion, abruptio
disease. Patients are more likely to die placentae, etc. (see p. 89)
from this than from thrombosis or ■ Shock – surgical trauma, burns
bleeding. However, specific treatment of Fig 38.2  Haemorrhagic bullae and ■ Severe haemolytic transfusion reaction
DIC may be life-saving and if bleeding gangrene in severe DIC. ■ Liver disease

occurs support with blood products is


indicated. Platelets, fresh frozen plasma can also increase the haemorrhagic Once absorbed it is stored in the liver
(FFP – a source of coagulation factors) risk by its anticoagulant action. Its use and following further metabolism it acts
and cryoprecipitate (a source of fibrino- should be considered where thrombosis as a cofactor for γ-glutamyl carboxyla-
gen) may all be used. Wherever possible predominates. Recombinant activated tion of coagulation factors II, VII, IX and
the choice of blood products should be protein C may be beneficial in patients X and proteins C and S. Vitamin K defi-
guided by the platelet count and coagu- with severe sepsis and DIC. ciency is probably the most common
lation tests. More controversial is the acquired coagulation disorder encoun-
use of pharmacological inhibitors of tered in hospital patients. The vitamin K
Vitamin K deficiency
coagulation and fibrinolysis. Although antagonist effect of warfarin is discussed
heparin can reduce clotting factor con- Vitamin K in the body is derived from on page 80 and the vitamin K deficiency
sumption and secondary fibrinolysis, it dietary vegetables and intestinal flora. of liver disease later in this section.
Acquired disorders of coagulation 77

Dietary deficiency low factor VIII level. Laboratory assay of the inhibitor is based
Normal dietary requirements for vitamin K are low (0.1–0.5 g/ on the ability of the patient’s plasma to neutralise the activity
kg) and thus patients must be considerably malnourished of a known amount of factor VIII.
before overt deficiency occurs. This most commonly occurs Management is complex and controversial but can be
in patients receiving intensive medical care, particularly where divided into the treatment of the acute bleeding episode and
broad-spectrum antibiotics are used. Deficiency is suggested subsequent attempts to eliminate the autoantibody by immu-
clinically by excessive bleeding and in the laboratory by a nosuppressive treatment. Possible approaches to the acute
prolonged prothrombin time. Supplemental vitamin K should episode include activated prothrombin complex concentrate
ideally be given before bleeding problems occur. (such as FEIBA, which contains activated factors VII, IX, and
X) and recombinant factor VIIa. Immunosuppressive strate-
Malabsorption gies include intravenous immunoglobulin and plasmapher-
Malabsorptive conditions such as coeliac disease and tropical esis in the acute episode and longer-term steroids or
sprue may lead to vitamin K deficiency. Vitamin K can also cyclophosphamide. The usual approach is summarised in
be lost in chronic biliary obstruction due to failure of bile salts Figure 38.4.
necessary for fat absorption to reach the bowel.

In the newborn
Vitamin K deficiency may arise in the first weeks or months
of life, most commonly in breast-fed, full-term and otherwise
healthy babies. Contributory factors include low placental Bleeding patient
transfer of vitamin K, low concentrations of vitamin K in
breast milk, low intake of milk and a sterile gut. The time of
onset is variable but haemorrhage most commonly occurs on
the 2nd to 4th day. A coagulation screen is abnormal with the
prothrombin time and APTT both prolonged. In most coun-
tries prophylactic vitamin K (1 mg intramuscular injection) is
given to newborn babies. Affected babies respond to parenteral Acute treatment
Immunosuppression
of bleeding
vitamin K but fresh frozen plasma or prothrombin complex
concentrate may be needed for severe haemorrhage.

Liver disease
Intravenous Ig
The liver is vital to normal haemostasis. It produces all the Plasmapheresis Short term
factors of the intrinsic and extrinsic coagulation pathway and
clears potentially damaging products of coagulation such as
fibrin degradation products and activated clotting factors. In
advanced liver disease there are often multiple haemostatic
abnormalities including reduced synthesis of clotting factors,
APCC or Steroids
increased consumption of clotting factors (DIC), qualitative recombinant factor VIIa Cyclophosphamide Longer term1
and quantitative platelet abnormalities, qualitative fibrinogen
abnormalities and accelerated clot lysis. Where bleeding Fig 38.4  The management of acquired haemophilia. APCC, activated
occurs, therapy is guided by the dominant haemostatic prob- prothrombin complex concentrate; Ig, immunoglobulin; a, activated.
lems. Possible interventions include parenteral vitamin K, 1
In refractory cases other immunosuppressive agents such as ciclosporin or
fresh frozen plasma, cryoprecipitate and platelet infusions. rituximab (anti-CD20) may be considered.

Acquired haemophilia
Antibodies (‘inhibitors’) that block the action of coagulation
factors may appear in patients who have no hereditary disor-
Acquired disorders
der of coagulation. Such autoantibodies most commonly
of coagulation
target factor VIII and the clinical syndrome is termed ‘acquired
haemophilia’. Acquired haemophilia may be associated with ■ Disseminated intravascular coagulation (DIC) is a complex clinical
a number of conditions including rheumatoid arthritis and syndrome which complicates serious illness. It causes both
haemorrhage and thrombosis. Laboratory tests are needed to
other autoimmune disorders, skin disorders, malignancy,
confirm the diagnosis.
drug therapy (particularly penicillin) and pregnancy. However,
■ Treatment of DIC is essentially that of the underlying cause. Blood
the most common presentation is in an elderly patient with
products are often indicated where bleeding occurs.
no associated condition. Possible clinical problems include
■ Vitamin K deficiency is a common acquired coagulation disorder.
haemorrhage into soft tissues and muscles (Fig 38.3), haema-
turia, haematemesis and prolonged bleeding postpartum or ■ Advanced liver disease can cause multiple haemostatic
postoperatively. Bleeding can be difficult to control and death abnormalities.
occurs in approximately 10% of cases. ■ Acquired haemophilia is generally caused by an autoantibody
In the laboratory, the diagnosis of acquired haemophilia is targeted against factor VIII. It may be idiopathic or associated with
other autoimmune diseases, malignancy, pregnancy or drug
suggested by a prolonged APTT worsening with incubation
treatment.
and not corrected by the addition of normal plasma, and a
78 7 HAEMOSTASIS AND THROMBOSIS

39 Thrombophilia
Patients who are predisposed to thrombosis generally either early age or to develop recurrent thrombotic problems.
have a disorder of the blood or an abnormality of the vessel Venous thrombosis predominates with the chance of throm-
wall. Where enhanced coagulation is the major mechanism, bosis increased by the coexistence of other risk factors.
the disorder is referred to as ‘thrombophilia’. Patients with Thrombophilia can be inherited or acquired.
thrombophilia either tend to have thrombosis at an unusually

Which patients should be investigated for thrombophilia?


Testing for heritable thrombophilia is not indicted in unse- Table 39.1  Characteristics suggesting possibility of thrombophilia
lected patients presenting with venous thrombosis. Table 39.1 ■ Venous thrombosis in patient less than 40 years old
summarises factors which should prompt consideration of ■ Recurrent venous thrombosis or thrombophlebitis
thrombophilia. Accurate history taking is essential; particular ■ Venous thrombosis in unusual site
attention should be given to the nature of the recent throm- ■ Arterial thrombosis in patient less than 30 years old
■ Strong family history of venous thrombosis
botic event, the presence of known risk factors (Table 39.2), a
■ Recurrent fetal loss
previous history of thrombosis and the family history. Defini- ■ Skin necrosis in patient receiving warfarin
tion of a ‘positive’ family history of thrombosis is problematic. ■ Purpura fulminans
If we use the simple definition of a history of deep vein
thrombosis (DVT) or pulmonary embolus (PE) in a first or
Table 39.2  Major risk factors for thrombosis
second degree relative, then approximately 25% of all patients
Venous Arterial
will have a positive family history. Even among those with a
Increasing age Increasing age
strong family history only a small minority will have a cause
Immobility Smoking
of inherited thrombophilia identified.
Obesity Male sex
Basic investigations of thrombophilia should include a
Oral contraceptive pill Hypertension
blood count (to exclude polycythaemia and other myelopro- Trauma/surgery Strong family history
liferative disorders) and a coagulation screen. Further labora- Thrombophilia (see text) Hyperlipidaemia
tory testing is dictated by the possible causes of familial and Pregnancy Diabetes mellitus
acquired thrombophilia detailed below. Testing for throm- Malignancy Raised fibrinogen
bophilia should not be undertaken during an acute episode
of venous thromboembolism when low levels of coagulation
inhibitors are routinely found. Systemic disorders such as Thrombin Protein C Protein S
Active
liver disease or disseminated intravascular coagulation (DIC) factors
can depress the levels of coagulation inhibitors and thus V or VIII
simulate the laboratory abnormalities found in familial 1 2 3
thrombophilia. Plasma 4
Inactive

Familial thrombophilia C C
Thrombin Thrombin Thrombin S
In theory, familial thrombophilia could be caused by any
genetically determined defect of the coagulation or fibrinolytic TM TM TM PL
systems that causes accelerated thrombin formation or Endothelium
impaired fibrin dissolution. In practice, the well-defined causes
are associated with accelerated thrombin formation either due Fig 39.1  Actions of proteins C and S. Thrombin and protein C bind to
to a shortage or failure of activation of one of a number of thrombomodulin (TM), an endothelial membrane protein (steps 1 and 2).
Protein S then binds to this complex and also endothelial phospholipid (PL)
circulating inhibitors of coagulation (Fig 39.1). Inherited
(step 3). The resulting complex proteolytically degrades activated factors V
thrombophilia defects are only important in venous
and VIII (step 4). Protein C is activated by proteolytic cleavage by thrombin.
thrombosis. In APCR, factor V is relatively resistant to inactivation by the protein C
complex.
Factor V Leiden (FVR506Q)
The anticoagulant property of activated protein C (APC) lies unselected cases of venous thrombosis. The risk of venous
in its capacity to inactivate the activated cofactors Va and VIIIa thrombosis is highest in patients homozygous for the muta-
by limited proteolysis. Inherited resistance to the anticoagu- tion or in heterozygotes with other risk factors.
lant action of APC (APCR) is an important cause of throm-
Prothrombin G20210A
bophilia. In most cases resistance is caused by a single point
This prothrombin gene polymorphism is the second most
mutation in the factor V gene (factor V Leiden) with replace-
common known cause of familial thrombophilia. It is found
ment of Arg506 with Gln. Arg506 is located at one of the APC
in approximately 4% of unselected patients with DVT. Both
cleavage sites in factor Va and the mutated Va is less sensitive
APCR and the prothrombin gene polymorphism are associ-
than normal Va to APC-mediated inactivation.
ated with a small increased risk of recurrent fetal loss.
APCR has an autosomal dominant mode of inheritance and
is the most common known cause of familial thrombophilia. Protein C and S deficiencies
The increased risk of venous thrombosis in APCR has been Hereditary deficiency of protein C is an autosomal dominant
estimated as 4–8-fold in heterozygotes and 50–100-fold in disorder found in 2–5% of patients with thromboembolic
homozygotes. The prevalence of the disorder in Western disease. An acquired deficiency of protein C can occur in liver
Europe is 3–7% with an incidence of around 15% in disease, DIC and warfarin treatment. Familial protein C
Thrombophilia 79

deficiency manifests as an increased leading to a hypercoagulable state and Table 39.3  Clinical and laboratory criteria
incidence of venous thromboembolism. thrombosis in the subcutaneous circu- for a diagnosis of antiphospholipid
Thrombotic events vary from a superfi- lation. The risk can be minimised by antibody syndrome (at least one clinical
cial thrombophlebitis to DVT and PE. ensuring full heparinisation before and one laboratory feature must
be present)
They may be spontaneous or triggered introducing warfarin. Protein C concen-
Clinical features
by other factors such as surgery or preg- trates have been given to treat purpura
Vascular thrombosis
nancy. In the rare homozygous form, the fulminans in homozygous disease. Pregnancy morbidity (e.g. unexplained late fetal death,
infant can be born with undetectable prematurity due to placental insufficiency/eclampsia,
Other situations
levels of protein C and quickly develop recurrent first trimester spontaneous abortion)
Case finding of asymptomatic relatives
DIC and skin necrosis due to microv- Laboratory tests
with low risk thrombophilia (e.g. factor Antiphospholipid lupus anticoagulant and/or
ascular thrombosis of subcutaneous
V Leiden) is not indicated. Asympto- antibodies: anticardiolipin/β2-
vessels (purpura fulminans). Protein S is
matic patients with familial throm- glycoprotein 1 antibodies
the non-enzymatic cofactor of protein C.
bophilia detected on laboratory tests
Hereditary deficiency has similar clinical
do not usually need anticoagulation. the patient also has systemic lupus ery-
features to protein C deficiency.
Patients with recurrent thrombosis or a thematosus (SLE) or a lupus-like disease.
Antithrombin deficiency single thrombosis with a high risk of About half of all patients have the
Antithrombin (AT) is the major physi- recurrence (e.g. multiple thrombophilic primary form of the disorder. Up to 2%
ological inhibitor of thrombin and clot- defects) should be considered for long- of the general population have detecta-
ting factors IXa, Xa, XIa and XIIa. term anticoagulation. Management of ble antiphospholipid antibodies – the
Deficiency can be inherited in an auto- thrombophilia in pregnancy is complex. probability of clinical problems is great-
somal dominant manner. Its prevalence Warfarin is potentially teratogenic and est where the antibody titre is high.
is unclear but AT deficiency probably subcutaneous low molecular weight The cause of thrombophilia in
contributes to venous thrombosis in heparin is given where anticoagulation antiphospholipid antibody syndrome is
around 2–5% of younger patients. is necessary. not entirely understood. Antiphospholi-
The risk of thrombosis varies between Counselling pid antibodies have been shown to play
disease subtypes, being greater for an Counselling is frequently not straight- a direct role in the development of
abnormality affecting the reactive forward. Any doubts relating to diagno- thrombosis in experimental animal
(thrombin binding) site than for an sis and the probability of thrombosis in models. Management must be individu-
abnormality affecting the heparin asymptomatic family members must be alised. Where there has been an episode
binding site. Overall, it seems that the acknowledged. Known acquired risk of major thrombosis, warfarin appears
risk of venous thrombosis is larger in factors such as immobility, obesity and to offer the best protection against
heterozygotes for AT deficiency than for the oestrogen-containing oral contra- recurrent thrombosis. Aspirin may give
those with APCR, protein C or protein ceptive should be avoided wherever additional benefit in arterial thrombosis.
S deficiency. The risk increases with age, possible. There is a two to four times Women with a history of morbidity in
with up to 80% developing venous increased risk of venous thromboembo- pregnancy are best treated in future
thrombosis by 55 years. lism in women receiving hormone pregnancies with a combination of
replacement therapy (HRT). aspirin and heparin.
Other forms of
familial thrombophilia Other acquired forms
High levels of the amino acid homo- Acquired forms of thrombophilia
cysteine are associated with atheroscle- of thrombophilia Myeloproliferative disorders are dis-
rosis and venous thrombosis and high cussed elsewhere (pp. 64–67). Increased
Antiphospholipid
factor VIII concentrations have been levels of plasma fibrinogen, and
antibody syndrome
linked with an increased risk of venous D-dimers may be predictors for coro-
Diagnosis of this syndrome requires
thrombosis. The mechanisms involved nary artery disease. Whether these and
either venous and/or arterial throm-
and the degree to which they are geneti- other haemostatic abnormalities are
boembolism or adverse outcomes in
cally determined is unclear. Other can- constitutional changes predisposing to
pregnancy in the presence of a persist-
didates for familial thrombophilia status coronary atherosclerosis and thrombo-
ing antiphospholipid antibody (Table
include the dysfibrinogenaemias (p. 75) sis or whether they are markers of pre-
39.3). The syndrome can be ‘primary’,
and factor XII deficiency. existing inflammation and endothelial
where the patient has no obvious
autoimmune disease, or ‘secondary’ if dysfunction is unclear.
Management of
familial thrombophilia
The precise role of laboratory throm- Thrombophilia
bophilia testing in clinical decision ■ The term ‘thrombophilia’ describes a predisposition to thrombosis caused by abnormally
making remains unclear. enhanced coagulation. Patients often have venous thrombosis at an early age or develop
recurrent thrombotic problems.
Acute venous thrombosis ■ Classicfamilial thrombophilia disorders are deficiencies of the naturally occurring inhibitors
This should be treated with heparin of coagulation, protein C, protein S and antithrombin.
and warfarin as in patients without her-
■ FactorV Leiden is a thrombophilia disorder caused by an inherited mutation in the factor V
itable thrombophilia (p. 80). Patients gene. Heterozygosity is common (3–7% in Western European population).
with protein C (and occasionally protein
■ The clinical role of laboratory thrombophilia testing is not well defined.
S) deficiency can rarely develop warfarin-
■ Antiphospholipid antibody syndrome is an acquired disorder characterised by laboratory
associated skin necrosis; this may be
identification of antiphospholipid antibodies and clinical features including thrombophilia
caused by an initial rapid fall in protein and morbidity in pregnancy.
C levels after warfarin commencement
80 7 HAEMOSTASIS AND THROMBOSIS

40 Anticoagulation and thrombolytic therapy


Two major classes of drugs are commonly used in the man- prophylaxis. They appear to have superior efficacy, a better
agement of thromboembolic disease. The anticoagulants safety profile and to be more cost-effective than unfraction-
heparin and warfarin are used to prevent thrombosis and ated heparin. LMW heparins cause less bleeding and a lower
limit the extension of an established clot, while thrombolytic incidence of thrombocytopenia and osteoporosis. The more
agents such as streptokinase are used to dissolve thrombus. predictable dose response precludes the need for routine
monitoring and the long half-life allows once or twice daily
subcutaneous administration. Where monitoring is indicated
Anticoagulation
(e.g. in renal failure), the anti-Xa effect is measured. LMW
Heparin heparin treatment allows outpatient management of uncom-
Unfractionated heparin is a naturally occurring gly- plicated DVT.
cosaminoglycan produced by mast cells. Low molecular Fondaparinux is a synthetic form of the heparin pentasac-
weight (LMW) heparin is prepared by controlled depolymeri- charide molecule with a specific anti-Xa effect. It has similar
sation of the unfractionated form. Both unfractionated and indications to LMW heparin.
LMW heparin exert their anticoagulant properties by binding
to antithrombin (AT) and potentiating its activity. AT is a Warfarin
normal circulating anticoagulant which inhibits the actions of Oral anticoagulant drugs are derived from 4-hydroxycoumarin
factor Xa and thrombin. LMW heparin differs from unfrac- and the standard agent is warfarin. Warfarin works by antago-
tionated heparin in having a relatively greater anti-Xa than nising vitamin K, which is needed for the gamma carboxyla-
antithrombin activity. tion of certain glutamic acid residues that facilitate calcium
binding of coagulation factors II, VII, IX and X (Fig 40.2).
Unfractionated heparin Some indications for warfarin are shown in Table 40.1.
Standard unfractionated heparin may be used therapeutically Where rapid anticoagulation is required a reasonable starting
to treat established thrombosis (usually intravenously at regimen is a single 10 mg dose and then protocol-guided
higher dosage) or prophylactically to prevent thrombosis adjustment according to the international normalised ratio
(usually subcutaneously at lower dosage). Most common (INR). A coagulation screen should always be checked before
indications for therapeutic use are deep vein thrombosis
(DVT) and pulmonary embolism (PE) (Fig 40.1). A typical
regimen is an intravenous loading dose of 5000 units followed
by an infusion of 1000–2000 units/hour. The anticoagulant
response varies as the drug binds non-specifically to plasma
and cellular proteins. Laboratory monitoring using the APTT
(see p. 20) is required; the therapeutic range is usually 1.5–2.5,
these values being the ratio of the patient’s APTT to a control
sample. As the half-life is short, high APTTs are managed by
stopping the heparin but in the event of bleeding (in up to
7% of cases) the antidote protamine can be given. When the
APTT is too low the heparin dose should be promptly
increased. Heparin is normally continued until oral anticoagu-
lation is therapeutic.
Prophylactic heparin is most commonly given to prevent
DVT and PE in patients undergoing surgery. It is particularly
indicated in patients with known risk factors for venous Fig 40.1  Large pulmonary embolus at the bifurcation of the main
pulmonary trunk.
thrombosis (see p. 78) and in major procedures. A typical
prophylactic regimen is 5000 units sub-
cutaneously preoperatively and 5000 Inactive coagulation g-carboxylated (active)
factors II, VII, IX, X coagulation factors
units 8- to 12-hourly after surgery, for 7 II, VII, IX, X
days or until the patient is mobile. No g - glutamyl
carboxylase
laboratory monitoring is necessary in
routine cases – where required anti-Xa
assays are used.
Apart from haemorrhage, patients on O2
heparin may develop thrombocytopenia Dietary Vitamin Vitamin K-2,3
(see p. 69) and prolonged use can cause vitamin K K-H2 epoxide
osteoporosis. It should be prescribed
cautiously where there is any bleeding
Vitamin K Vitamin K
tendency. epoxide
reductase Vitamin K
-quinone reductase
LMW heparin Fig 40.2  The vitamin K cycle and the action of warfarin. The major site of warfarin action is not a
LMW heparins are now preferred direct effect on the carboxylation step needed for coagulation factor activation but on steps needed for
for treatment of DVT and PE and resynthesis of active vitamin K from its epoxide form.
Anticoagulation and thrombolytic therapy 81

Table 40.1  Warfarin: common indications permits reporting as an INR such that clinics where control is audited and
and recommended INRs INR = (prothrombin time)ISI. technologies such as computerisation
Indication Target INR1 As it takes several days for warfarin to exploited.
Deep vein thrombosis 2.5 become therapeutic, the conventional
Pulmonary embolus 2.5 treatment of established thrombosis is New oral anticoagulant agents
Atrial fibrillation 2.5 to start heparin and warfarin simultane- There is a need for oral anticoagulant
Mural thrombosis 2.5 ously and only to stop heparin when the agents with more predictable pharma-
Cardioversion 2.5 desired INR has been achieved. Warfa- cokinetics than warfarin. A number of
Mechanical prosthetic heart valves 2.5–3.52 rin should be used with caution in new drugs have been tested in clinical
Recurrent venous thromboembolism 3.5 patients with a bleeding tendency. The trials and are now entering clinical prac-
on warfarin therapy
most common side-effect is haemor- tice for both prophylaxis and treatment
Thrombosis in antiphospholipid 2.5
rhage, the risk of serious bleeding cor- of thromboembolic disease. Examples
antibody syndrome
relating with the height of the INR. Poor include the direct thrombin inhibitor
1
An INR within 0.5 units of the target is usually satisfactory. control of anticoagulation and bleeding dabigatran exilate and the factor X
2
Depending on valve.
may arise from poor prescribing or inhibitor rivaroxaban. This is a fast
compliance, intercurrent illness and moving area of medicine and new anti-
interaction with a potentiating drug coagulants are further discussed on
Table 40.2  Drugs interacting
(Table 40.2). A prolonged INR in a non- page 102.
with warfarin1
haemorrhagic patient may only require
Potentiating Antagonising
withdrawal of the drug for a few days.
Alcohol Oral contraceptives Thrombolytic therapy
Where there is haemorrhage, warfarin
Cimetidine Spironolactone
can be reversed within hours by oral/ Thrombolytic agents dissolve fresh clots
Allopurinol Antihistamines
Quinine Barbiturates
intravenous vitamin K (0.5–5 mg) and and therefore restore vascular patency
Amiodarone Rifampicin
instantly by infusion of a concentrate of more quickly than anticoagulants. The
Co-trimoxazole Sucralfate prothrombin complex. Guidelines are commonly used agents – streptokinase,
Metronidazole Anti-epileptics complex and significant warfarin over- urokinase and tissue plasminogen acti-
Tricyclics dosage should be discussed with a hae- vator (alteplase) – work by activating the
Aspirin and salicylates matologist. The duration of warfarin fibrinolytic system (see p. 13 and Fig
Anabolic steroids treatment depends on the indication. 40.3). They convert plasminogen, the
Thyroxine Anticoagulation may be needed for only inactive proenzyme of the system, to the
Sulfinpyrazone 3 months in a patient with a limited proteolytic enzyme plasmin. Thrombo-
1
DVT and reversible risk factors (e.g. lytic drugs are indicated in any patient
These are some commonly implicated agents – this is not a
comprehensive list. post-surgery). Longer periods are indi- with acute myocardial infarction in
cated in idiopathic venous thrombosis, which the benefit is likely to outweigh
and lifelong warfarin treatment may be the risk of treatment. Early administra-
warfarin is prescribed. The maintenance justified following recurrent episodes of tion gives the best results. Other possi-
dose is usually between 3 and 9 mg. venous thrombosis or where there is a ble uses for this class of drugs include
Laboratory monitoring depends on the known ongoing thrombotic risk such as treatment of complicated venous throm-
prothrombin time (see p. 20). As throm- a prosthetic heart valve, atrial fibrillation bosis, acute ischaemic stroke and the
boplastin reagents used in this test vary, or a thrombophilic state. unblocking of occluded venous cathe-
their sensitivity is labelled with an inter- Community and outpatient warfarin ters. The major side-effect is excessive
national sensitivity index (ISI) which treatment is best monitored in specialist bleeding.

Extrinsic activation Intrinsic activation


(e.g. tPA) from vessel wall Anticoagulation and
thrombolytic therapy
■ The anticoagulant drugs heparin and warfarin are used to prevent
Fibrin thrombosis and limit the extension of an established clot.
Plasminogen Plasmin
degradation ■ Unfractionated heparin is given intravenously and low molecular
weight heparin, now generally preferred, is given subcutaneously.
■ Warfarin is given orally and acts by inhibiting vitamin K.
Thrombolytic ■ Therapeutic treatment with both unfractionated heparin and
agents warfarin requires careful laboratory monitoring.
Fig 40.3  Action of thrombolytic agents. ■ New oral anticoagulant drugs (e.g. dabigatran, rivaroxaban) are
entering clinical practice.
■ Thrombolytic agents are used to dissolve thrombus. They act by
converting plasminogen to the proteolytic enzyme plasmin.
82 8 BLOOD TRANSFUSION

41 Blood groups and blood testing

The blood groups Table 41.1  The occurrence of ABO Testing before transfusion
antigens and antibodies Most incompatible transfusions are
Blood group antigens exist on the ABO blood group caused not by errors in the transfusion
surface of the red cell membrane (see O A B AB laboratory but by giving blood to the
also p. 4). There are numerous blood Antigens on red cells None A B A+B ‘wrong’ patient (i.e. not the patient
group systems encoded by genes on dif- Antibody in serum Anti-A+B Anti-B Anti-A None whose serum was tested prior to the
ferent chromosomes. They are highly Frequency (%)1 47 42 8 3 transfusion). The source of such mis-
variable in their polymorphism and 1 takes is usually inaccurate documenta-
In the United Kingdom.
clinical significance. Incidences vary greatly in different populations. tion on forms and specimens or
The most important blood group is inadequate procedures for identifying
Table 41. 2  Routine testing of
the ABO system. The genes encoding patients prior to transfusion (see also
donated blood
the ABO antigens are located on chro- p. 84).
ABO group
mosome 9 and are inherited in an auto- Rhesus group If tests on donor and recipient blood
somal dominant fashion. Each antigen Red cell antibody screen confirm matching for ABO and Rhesus
is a sugar residue made by a specific Hepatitis B surface antigen, HBV DNA groups, the transfusion will be compat-
glycosyl transferase. The ABO system is Antibody to syphilis
ible in around 98% of cases. The
Anti-HIV-1 and anti-HIV-2, HIV-1 antigen, HIV RNA
crucial in clinical blood transfusion as sequence of tests prior to transfusion
Anti-Hepatitis C, HCV RNA
there are naturally occurring IgM anti- Anti-HTLV
bodies in the serum targeted against the
Most incompatible blood
non-present ABO antigens (Table 41.1). disease (CJD) infection are not suitable
transfusions arise from clerical
These antibodies necessitate the use donors.
errors and mistaken patient
of ABO ‘compatible’ blood for transfu- The objective of routine testing of
identity.
sion. For example, the administration of donated blood is to provide blood which
incompatible group A blood to a group can be selected for likely compatibility
B patient would engender a potentially with a patient and which contains no includes antibody screening of the
fatal haemolytic transfusion reaction identifiable infectious agent (Table 41.2). patient’s serum and crossmatching to
due to the destruction of the donor’s Antibody testing (e.g. for HIV and hepa- ensure compatibility in the remaining
group A red cells by the recipient’s titis C) is now supplemented by molecu- 2%.
anti-A antibody. lar techniques sensitive enough to trace
In other blood group systems ‘natu- the virus in the blood before the devel- Blood grouping
rally occurring’ antibodies are rare. opment of antibodies (i.e. during the The recipient’s red cells are tested for
However, ‘immune antibodies’, usually ‘window period’). ABO and Rhesus antigens and the
of IgG type, may be induced by transfu-
sion of blood expressing different blood
group antigens or maternal exposure to
fetal red cell antigens. Where such
immune antibodies are present, trans-
fused blood must be matched for the
relevant blood group system in addition
to ABO. Maternal formation of immune
antibodies against antigens of the
Rhesus (Rh) blood group system, par-
ticularly the strongest antigen D,
accounts for most cases of haemolytic
disease of the newborn (p. 90).

Fig 41.1  ABO blood grouping on a microplate.


The testing of blood
Donor blood
The safety of blood transfusion is max-
imised by careful selection of donors. All
donors should be in good health and,
wherever possible, unpaid volunteers.
Particular care is taken to exclude poten-
tial donors who may harbour infective
diseases which are transmissible by
blood transfusion – thus people with
recent jaundice (? hepatitis), a history of (a) (b)
recent travel to malarial areas or risk Fig 41.2  Blood grouping using a gel system. (a) ABO and RhD grouping. (b) Rh and Kell grouping.
factors for HIV or Creutzfeldt–Jakob Unagglutinated red cells pass through the gel (after centrifugation) whereas agglutinated cells do not.
Blood groups and blood testing 83

Washing to Addition of
remove unbound anti-human
antibodies globulin
Agglutination
Positive test

Sensitised red cells

Red cell antigen Red cell antibody Anti-human globulin

Fig 41.3  The antiglobulin test. In the direct test red cells are sensitised in vivo, in the indirect test they are sensitised in vitro.
serum tested for naturally occurring a combination of repeat group and anti- Table 43.3  Possible guidelines for blood
antibodies to confirm the ABO group. body testing, specialised software and ordering in a few common operations
Blood grouping tests traditionally rely stringent standard operating procedures Protocols vary between hospitals and should be based
on the visual identification of agglutina- to detect any incompatibility. on previous blood usage and local transfusion
tion of red cells induced by the presence laboratory practice

of antibodies against antigens present Procedure Recommendation1


Practicalities of Cholecystectomy Group and save
on the cell surface (Fig 41.1). Gel technol-
blood ordering Colectomy/hemicolectomy 2
ogy (see Fig 41.2) is widely used and
Breast biopsy Group and save
increasingly automated. DNA-based Where blood transfusion is required
Heart valve replacement 8
tests may be employed as an adjunct to and adequate time is available, tests
Resection of abdominal aortic 8
haemagglutination but they are not cur- proceed as above and compatible units
aneurysm
rently indicated for routine ABO and Rh are issued. In emergencies, blood is Abdominal hysterectomy Group and save
group testing. sometimes needed more quickly than Total hip replacement 3
this routine testing allows. Normal pro- Transurethral resection of Group and save
Antibody screening cedures may be adapted to speed up prostate
The patient’s serum is tested against issue of group specific blood. If there is
1
Figures refer to the number of units of red cells crossmatched prior
three standard sets of screening red cells insufficient time to determine the to surgery.
of known antigenic type. This is to patient’s ABO group, then group O
detect immune or ‘atypical’ antibodies Rhesus-negative blood may be used. Most hospitals have implemented a
(i.e. non-ABO) which might destroy The bulk of blood is crossmatched for formal surgical blood order schedule
donor red cells. Clinically relevant anti- use in elective surgical procedures. with guidelines for common operations
bodies are generally reactive at 37°C. A Where there is only a small chance (less (Table 43.3). Such guidelines are gener-
sensitive indirect antiglobulin test tech- than 10%) that transfusion will be alisations and special provision is made
nique is used (see Fig 41.3). If an anti- required it is reasonable to limit wastage for unusually difficult procedures or
body is detected by this screen then a by adopting a ‘group and save’ policy. patients who are judged to be at a higher
larger panel of red cells is tested to The patient’s blood group is determined than average risk of haemorrhage. Elec-
indentify it. The blood selected for the and the serum screened for atypical tronic crossmatching is very rapid and
patient must be negative for the relevant antibodies. Provided the screen is nega- its introduction potentially reduces the
antigen. tive, blood is not routinely crossmatched. number of operations for which blood
is issued in advance.
Crossmatching
The final compatibility check is to mix
the patient’s serum with red cells from Blood groups and blood testing
each donor unit. The aim is to highlight
■ The blood group antigens exist on the surface of the red cell membrane. Blood groups are
any earlier errors in grouping or anti- highly variable in their polymorphism and clinical significance.
body screening and to identify the pres-
■ The ABO system is crucial in blood transfusion as there are naturally occurring IgM
ence of antibodies against rare antigens
antibodies in the serum targeted against non-present ABO antigens – this necessitates the
not present on the screening cells. The use of ABO ‘compatible’ blood.
minor crossmatch – mixing of donor
■ Blood donors are carefully selected and donor blood tested to exclude transmissible
serum with patient red cells – is not infections.
routinely performed. With the integra-
■ Testingof donor and recipient blood for ABO and Rhesus groups, antibody screening and
tion of sophisticated computer systems crossmatching are routinely performed before transfusion to ensure compatibility.
into laboratories, the serological cross-
■ Most incompatible blood transfusions arise from clerical errors and mistaken patient
match is being selectively replaced by identity.
‘electronic crossmatching’. This relies on
84 8 BLOOD TRANSFUSION

42 Clinical practice
(is another patient about to get a ‘wrong’
Red cell transfusion unit due to a mix up?) and the blood
Two questions need to be answered bank informed. Initial investigations
before transfusion of red cells is must include blood samples from the
undertaken: patient for a blood count and film,
blood group, antibody screen and direct
1. Is it indicated?
antiglobulin test. The blood bank will
2. If it is indicated, which red cell
also repeat tests on the donated unit.
preparation should be used?
Management of complications will
Some general indications for red cell require senior advice and often intensive
transfusion are listed in Table 42.1. care. The overall mortality of ABO
Whole blood is now rarely used. incompatible transfusion is approxi-
Haemorrhage requires transfusion of mately 10%.
fluids to maintain blood volume and red Non-haemolytic transfusion reac-
cells to raise the haemoglobin level. For tions. The majority of adverse reactions
correction of anaemia not responsive to to blood are ‘febrile reactions’ caused by
other measures red cells stored in Fig 42.1  Unit of red cells. antileucocyte antibodies in the patient.
‘optimal additive solution’ are used. Uncomplicated febrile reactions are
There are few indications for red cells IBCT (35%) simply managed by slowing the transfu-
stored in plasma. sion and giving paracetamol. Routine
leucodepletion of red cells reduces such
TACO (1%) HTR (6%) (6%)
Practicalities of red I and U reactions. Occasionally patients develop
TTI (1%)
cell transfusion TRALI (3%) HSE (12%) allergic reactions with urticaria, wheez-
All those involved in the prescription ing and (rarely) anaphylaxis.
Other (3%)
and administration of blood should ATR (21%) Anti-D Transfusion associated circula-
follow local guidelines with respect to (12%) tory overload (TACO). Care must
patient identification and the checking be taken not to transfuse too rapidly,
of the compatibility and viability of the Fig 42.2  Serious hazards of transfusion, especially in elderly patients with heart
transfused units. Critical information is United Kingdom 1992–2010 (data with disease.
permission of SHOT). IBCT, incorrect blood
contained on the blood bag and the Transfusion-related acute lung
transfused; ATR, acute transfusion reactions; HSE,
attached compatibility label (Fig 42.1). injury (TRALI). This is an acute syn-
handling and storage errors; I and U,
No discrepancies are permissible. Most inappropriate/unnecessary/delayed transfusions; drome occurring within 6 hours of
serious adverse transfusion reactions HTR, haemolytic transfusion reactions; TRALI, transfusion and characterised by respi-
are due to transfusion of the wrong transfusion-related acute lung injury; TACO, ratory distress, hypoxia, bilateral pulmo-
blood to the patient (Fig 42.2). Errors transfusion-associated circulation overload; TTI, nary infiltrates and a fever. Donor
can be reduced by newer technologies transfusion-transmitted infections. antibodies to HLA class I and II antigens
such as bar coding and radiofrequency haemoglobin can be expected from each and/or granulocyte-specific antigens
chips – these generally rely on machine unit. Red cells are infused via specially have been implicated in pathogenesis.
readable data on patient wristbands. designed sterile ‘giving sets’ which Mortality is around 10%.
In shocked patients blood is trans- contain 170 µm filters. Careful monitor-
fused rapidly, the precise rate dependent ing is particularly important during the Delayed
on the monitoring of vital signs such as first 10 minutes of each unit. Infection. Bacteria, viruses and para-
pulse, blood pressure and urine output. sites may all be transmitted via blood
Transfusion for correction of anaemia Complications of red transfusion. Blood is screened for the
is usually a more elective process. Units cell transfusion relevant agents and in practice the great-
of red cells are typically given over 2–4 Immediate est risk is of bacterial contamination. To
hours and a rise of around 10 g/L of Haemolytic transfusion reactions. help reduce the chance of transmission
These potentially fatal reactions arise of the abnormal prion associated with
Table 42.1  Major indications for red from the transfusion of incompatible variant Creutzfeldt–Jakob disease (vCJD)
cell transfusion
blood (usually for ABO). Symptoms red cell donations are leucodepleted and
To replace blood loss
often occur within minutes and may plasma is increasingly imported from
Trauma
Surgery include chest, abdominal and loin pain, countries with no bovine spongiform
Other haemorrhage (e.g. gastrointestinal bleed) vomiting, a ‘burning’ skin, dyspnoea and encephalopathy (BSE). The significance
To correct anaemia headache. Common signs are fever, of transmission of infection from blood
Marrow failure (e.g. aplastic anaemia, leukaemia) can depend on the status of the recipi-
tachycardia and hypotension. Renal
Haemoglobinopathies (e.g. thalassaemia, sickle cell
failure and disseminated intravascular ent. Thus, cytomegalovirus (CMV) is of
disease)
Chronic disorders (e.g. renal failure, malignancy) coagulation (DIC) can follow. Once a little relevance in healthy adults but
Severe haemolysis (e.g. haemolytic disease of the haemolytic reaction is suspected, the potentially life-threatening in a patient
newborn) transfusion should be stopped and the receiving an allogeneic stem cell trans-
The final decision to transfuse requires consideration of the patient’s
venous access used to give crystalloid. plant or in a low birthweight premature
age, clinical state, and haemoglobin concentration. The transfused unit should be checked infant.
Clinical practice 85

Delayed transfusion reactions.


These occur approximately 5–10 days
Transfusion of platelets Table 42.2  Possible indications for use of
fresh frozen plasma
after transfusion and are caused by a
and granulocytes
Disseminated intravascular coagulation (DIC)
previously undetected antibody being Platelet transfusion Severe liver disease (e.g. prior to liver biopsy)
boosted by transfusion of incompatible This is used to treat or prevent haemor- Coagulopathy of massive blood transfusion
Replacement therapy of some rare congenital factor
cells. Characteristic features include rhage in patients with significant throm-
deficiencies
fever, jaundice and a falling haemo- bocytopenia. It is more useful where Bleeding in haemorrhagic disease of newborn/
globin. They are only rarely fatal. platelets are low due to underproduc- malabsorption vitamin K
Iron overload. A unit of blood con- tion (i.e. marrow failure) or dilution Thrombotic thrombocytopenic purpura (with plasma
tains around 250 mg of iron. Iron is than where thrombocytopenia is due to exchange)
Depletion of coagulation factors following
only lost from the body in small immune destruction as in immune
thrombolysis
amounts and repeated transfusion can thrombocytopenia (ITP). Platelets are
lead to accumulation and toxic effects collected either from routine blood
identical to those seen in haemochro- donations or from a single donor by adult is one litre. FFP can transmit
matosis. Where repeated transfusion is plasmapheresis. They should ideally be infection and cause immunological
predictable in a younger person (e.g. in matched with the patient for ABO and reactions – it is not suitable for
thalassaemia), chelation of iron limits Rhesus. The standard dose for an adult volume expansion alone.
overload and prolongs life. is either a single plasmapheresis dona- ■ Cryoprecipitate. This is prepared from

tion or 4–6 pooled standard donations. FFP by slow thawing and separation
Massive blood transfusion
Where repeated platelet transfusions of the resultant precipitate. It is rich
Massive transfusion is defined as replace-
are given, patients can become sensi- in fibrinogen and may be useful in
ment of the patient’s whole blood
tised against class I HLA antigens the treatment of DIC and
volume by stored allogeneic blood in
absorbed onto the platelet surface with management of massive blood
less than 24 hours. There have been
the result that they derive a lower incre- transfusion.
recent changes in practice driven by the
ment in platelet count than would be ■ Factor VIII and IX concentrate. See
military experience of trauma with
predicted (‘platelet refractoriness’). In pages 72–73.
increased early use of plasma and plate-
these cases, platelet donors matched ■ Albumin. This is produced by
lets. Problems can still arise in part due
with the recipient’s HLA class I type can fractionation of pooled plasma.
to the inevitable deficiencies of stored
be selected. Platelet transfusion can Solutions for clinical use include
blood. Shortage of clotting factors and
cause non-haemolytic reactions and can human albumin 4.5/5%, human
platelets in transfused blood may exac-
transmit infection as for red cells. albumin 20% and plasma protein
erbate haemorrhage. It is important to
fraction (PPF). Albumin solutions are
monitor haemostasis by checking the
Granulocyte (neutrophil) used for the treatment of severe
basic coagulation screen and replacing
transfusion hypoproteinaemia, particularly when
components accordingly. Metabolic dis-
Granulocyte transfusion is infrequently associated with a low plasma volume.
turbances are less common but include
used in neutropenia and the indications Concentrated solutions can help
hyperkalaemia, hypocalcaemia, acidosis
are uncertain. produce a diuresis in hypo-
and citrate toxicity. Rapid transfusion
albuminaemia (e.g. in hepatic
can cause hypothermia; this can be min-
cirrhosis).
imised by carefully controlled blood Transfusion of plasma ■ Immunoglobulins. These can be
warming. and plasma products
‘specific’ and used in passive
Alternatives to allogeneic A wide range of plasma products is prophylaxis against a range of
blood transfusion available for therapeutic use: infections (e.g. varicella zoster,
Use of the patient’s own blood for trans- tetanus) or to prevent haemolytic
■ Fresh frozen plasma (FFP). Plasma is
fusion rather than allogeneic blood disease of the newborn (anti-Rhesus
collected from whole blood or
minimises the risk of infection. Selected D). ‘Non-specific’ immunoglobulins
derived from plasmapheresis prior to
patients awaiting elective surgery can are used for passive prophylaxis
rapid freezing. FFP contains the full
‘pre-deposit’ blood in the weeks prior to against hepatitis A, treatment of
range of coagulation factors and
the operation. An alternative approach, hypogammaglobulinaemia and in
indications for use are shown in
now more favoured, is to use specially selected autoimmune disorders (e.g.
Table 42.2. The normal dose in an
designed equipment to ‘salvage’ blood ITP).
lost during surgery and reinfuse it back
into the patient. Other strategies to
reduce blood transfusion include the Blood transfusion –
active treatment of anaemia, strict appli- clinical practice
cation of transfusion triggers, anaes- ■ Beforered cell transfusion is undertaken the indication should be confirmed and the
thetic measures or drugs (e.g. tranexamic optimal red cell preparation selected.
acid) to reduce blood loss and biological ■ Red cell transfusion can cause both immediate complications (e.g. haemolytic transfusion
agents such as erythropoietin. Ques- reaction) and delayed complications (e.g. infection, iron overload).
tions remain regarding optimal transfu- ■ Platelet transfusion may be helpful in the management of thrombocytopenia.
sion practice and there is a need for
■ A wide range of plasma products is available for transfusion. Selection of the appropriate
more randomized controlled trials. Pos-
product requires an understanding of the therapeutic benefit and possible side-effects.
sible future developments include man-
■ There are alternatives to allogeneic blood transfusion (e.g. erythropoietin) which should be
ufactured haemoglobin solutions and
considered in selected patients.
platelet substitutes.
86 9 SPECIAL SITUATIONS

43 The immunosuppressed patient


Many patients with blood disorders Table 43.1  Possible factors predisposing
are immunosuppressed. Patients with to infection in haematology patients
aggressive haematological malignancies Cellular defects
such as leukaemia and non-Hodgkin’s ■ Neutropenia and neutrophil dysfunction
lymphoma have their immune function ■ Lymphopenia and lymphocyte dysfunction

initially compromised by the disease and Humoral defects


■ Reduced antibody production
then further depressed by chemother-
Anatomic defects
apy. Others have more subtle deficien-
■ Reduced mucosal barriers (e.g. mucositis)
cies. Patients with ‘benign’ diseases such ■ Indwelling venous catheters
as immune thrombocytopenia (ITP) and Splenectomy (see p. 10)
hereditary spherocytosis who have had
splenectomy performed are also at
increased risk of infection.
An increased susceptibility to infec-
tion can arise from multiple factors
(Table 43.1). Neutropenia and neutrophil
dysfunction are probably the most
important causes of infectious complica-
tions in patients with leukaemia. Unlike
Fig 43.2  Aspergillosis complicating
prolonged neutropenia.
many other forms of immunosuppres-
sion, neutropenia is easy to quantify – deadly fungal pathogens. Infection is
the risk of infection rises appreciably at Fig 43.1  Skin infection caused by usually via the inhalation of airborne
Streptococcus faecalis in a neutropenic
counts below 0.5 × 109/L and is greatest spores and is mainly pulmonary. A chest
patient.
where the count is below 0.1. Lympho- X-ray may show pneumonia and cavita-
penia and lymphocyte dysfunction are Bacterial infection in neutropenic tion (Fig 43.2) but it is an insensitive
seen in lymphoid malignancy and after patients may be overt – for instance a diagnostic method. Other infected sites
chemo- and radiotherapy. Defects in chest infection with a productive cough can include the paranasal sinuses, skin,
humoral immunity are particularly seen or the presence of infected skin lesions central nervous system and eye. Even in
in patients with chronic lymphoid (Fig 43.1). However, bacterial sepsis can disseminated disease, blood and sputum
malignancies and in myeloma. The like- equally present with non-specific cultures are rarely positive. Thirty per
lihood of infection is related to the sever- malaise and a pyrexia. In the latter case cent of cases of invasive aspergillosis
ity of hypogammaglobulinaemia. extensive cultures including blood, nose, remain undiagnosed and untreated at
Other common immunosuppressive throat, stool and urine are indicated. death. Strategies for earlier diagnosis
factors are the loss of mucosal or skin include regular galactomannan antigen
integrity due to damage from disease or Fungi testing, aspergillus PCR, and CT scan-
treatment, and the presence of indwell- The incidence of invasive fungal infec- ning of the chest.
ing venous catheters. tions is increasing and they are a Pneumocystis jiroveci (previously
major cause of morbidity and mortal- Pneumocystis carinii) is a fungus which
ity in patients with haematological causes a potentially fatal bilateral pneu-
Types of infection
malignancy. monia in patients with depressed cell-
Bacteria The most widespread fungal patho- mediated immunity. In haematological
Bacterial infections in neutropenic gen is Candida. Oral and colonic car- practice it mostly affects patients receiv-
patients are often caused by the spread riage of the organism is common in ing intensive chemotherapy regimens or
of commensal flora to previously sterile healthy people. Invasive Candida infec- stem cell transplantation and patients
sites. Fatal septicaemia can result from tion is most likely in neutropenic infected with the HIV virus. Bronchoal-
Gram-negative bacilli such as Pseu- patients with indwelling catheters and veolar lavage is useful in diagnosis.
domonas aeruginosa, E. coli, Klebsiella severe mucositis. Disseminated candi-
spp. and Enterobacter spp. Gram-positive diasis usually presents with a persistent Viruses
cocci currently cause the majority of fever and no diagnostic clinical features. Most viral infection in immunosup-
documented bacteraemias. The skin Possible organ involvement includes the pressed patients is caused by reactiva-
pathogen Staphylococcus epidermidis kidney, lung, heart and liver. Cutaneous tion of latent organisms. Patients with
often colonises indwelling venous cath- emboli may lead to a nodular skin erup- deficient cell-mediated immunity (e.g.
eters. The use of broad-spectrum antibi- tion while exudative retinal lesions can acute lymphoblastic leukaemia (ALL),
otics can lead to the emergence of be seen through the ophthalmoscope. stem cell transplantation, chronic lym-
toxin-producing Clostridium difficile in Candida spp. are grown from blood cul- phocytic leukaemia) are particularly sus-
the stools. Methicillin-resistant Staph. tures in only 20% of patients with inva- ceptible. Important pathogens include
aureus (MRSA) and bacteria produc- sive candidiasis. The addition of candida herpes simplex, varicella zoster and
ing extended-spectrum beta-lactamases PCR significantly improves detection cytomegalovirus (CMV). Clinical mani-
(ESBLs) leading to antibiotic resistance rates. festations range from relatively trivial
are becoming increasingly problematic Aspergillus species, particularly mouth ulcers attributable to herpes
in hospitals. Aspergillus fumigatus, are potentially simplex through herpes zoster
The immunosuppressed patient 87

Table 43.2  Groups of antibiotics used in prior to starting antibiotics and a chest
the empirical treatment of infection in X-ray is helpful: investigations, however,
neutropenia should not substantially delay treat-
Group Examples ment. A microbiological diagnosis is
Antipseudomonal Azlocillin, piperacillin made in only half of these cases.
penicillins
The empirical antibiotic regimens are
Aminoglycosides Gentamicin, amikacin
designed to provide protection against
Cephalosporins Ceftazidime
commonly implicated organisms, par-
Quinolones Ciprofloxacin
ticularly those causing life-threatening
Carbapenems Meropenem, imipenem
infection (e.g. Pseudomonas). Regimens
Glycopeptides Teicoplanin, vancomycin
are constantly changing – the major
groups of drugs are summarised in
Depressed cell-mediated Table 43.2.
immunity and Monotherapy (e.g. meropenem)
Fig 43.3  Herpes zoster following allogeneic hypogammaglobulinaemia may be used but in patients at highest
stem cell transplantation.
Impaired cell-mediated immunity leads risk a combination (e.g. piperacillin/
(shingles) (Fig 43.3) with the risk of dis- to an increased risk of Pneumocystis tazobactam and gentamicin) is usually
semination to the potentially fatal CMV jiroveci (carinii) pneumonia and viral preferred.
pneumonitis which complicates alloge- infections. Standard prophylaxis against Persistent pyrexia or clinical deteriora-
neic stem cell transplantation. Measles Pneumocystis is oral co-trimoxazole or tion on first-line antibiotics is a difficult
can be a fatal illness in children with nebulised pentamidine where this is not management problem. Often the infec-
ALL. There may be no specific diagnos- tolerated. Aciclovir is effective in reduc- tious agent is unknown. The usual
tic features of viral infection and it must ing the incidence of viral infections. The approach is to continue investigations
be considered as a possible cause of a more toxic drug ganciclovir can be used while making a change in the antibiotic
febrile illness in the immunosuppressed after stem cell transplantation to give regimen. A lack of response prompts
patient. PCR-based diagnosis may allow additional protection against CMV. consideration of empirical antifungal
earlier therapy of CMV infection after Patients with low-grade lymphoprolif- treatment. To limit drug exposure,
allogeneic stem cell transplantation. erative disorders and myeloma can have entirely empirical therapy can be replaced
significant hypogammaglobulinaemia by a ‘pre-emptive’ strategy where only
and suffer recurrent infection. Regular patients with ‘probable’ fungal infection
Prevention of infection in infusions of immunoglobulin may be (e.g. suggested by CT of chest) are treated
the immunosuppressed helpful in these cases. with antifungal agents. Growth factors
patient (e.g. G-CSF) may be given to shorten the
Neutropenia Post-splenectomy period of neutropenia.
General measures include the isolation See page 10.
of the patient, laminar airflow rooms, Treatment of specific infections
strict hygiene and avoidance of possible Treatment of infection Liposomal amphotericin B has generally
contaminants (e.g. uncooked food). been the drug of choice for treatment of
Simple precautions such as hand The pyrexial neutropenic patient established Aspergillus infection and in
washing by staff are crucial in reducing A common clinical problem in haema- the empirical antifungal role outlined
infection rates. tology is the management of the patient above, but voriconazole and caspofun-
Antimicrobial prophylaxis may reduce with neutropenia who becomes unwell gin are reasonable alternatives. Azoles,
the incidence of infection but there are and/or develops a pyrexia. A subgroup particularly fluconazole, are commonly
well-defined adverse effects. For instance, of very carefully defined ‘low-risk’ used in the treatment of Candida infec-
quinolone antibiotic prophylaxis reduces patients may require only oral broad- tion. Herpes simplex and varicella zoster
the number of bacterial infections spectrum antibiotics but high-risk infections are best treated with aciclovir.
in patients with chemotherapy and patients can rapidly succumb to bacte- Ganciclovir or foscarnet is used for
transplant-induced neutropenia but this rial infection and need prompt inpatient CMV infection after allogeneic stem cell
must be balanced against the side-effects empirical treatment with broad- transplantation. Pneumocystis jiroveci
of the drug and the potential emergence spectrum intravenous antibiotics even (carinii) pneumonia is effectively treated
and dissemination of antimicrobial- before the infectious pathogen is identi- by either high-dose co-trimoxazole or
resistant organisms. Increased use of fied. Blood and other cultures are taken pentamidine.
antimicrobial agents increases the vul-
nerability of patients to nosocomial The immunosuppressed patient
infections (e.g. Clostridium difficile) and
■ Many patients with blood disorders are immunosuppressed. Possible factors predisposing
community-acquired infections. Prophy-
to infection include neutropenia, lymphopenia, reduced antibody levels and anatomical
laxis also complicates the treatment of a
defects.
subsequent episode of febrile neutrope-
■ Bacteria, fungi and viruses can all cause severe systemic infection in an immunosuppressed
nia. It appears that the best way to
patient.
exploit the benefits of prophylaxis is to
■ Measures to prevent infection in the immunosuppressed patient include isolation of the
restrict its use to patients at highest risk
patient, strict hygiene and selective prophylactic use of antimicrobial agents.
such as those with a previous history of
■ Infection in a neutropenic patient generally requires empirical treatment with broad-
neutropenic fever. Similar considera-
spectrum antibiotics. Persisting fever or clinical deterioration necessitates a change in
tions apply to the use of prophylaxis antibiotics and/or antifungal treatment.
against fungal infections.
88 9 SPECIAL SITUATIONS

44 Pregnancy

Haematological changes
40% increase
Several haematological changes occur in plasma volume
normal pregnancy (Fig 44.1). Beginning Dilutional
anaemia
in the sixth week there is an increase in 15–25% increase
plasma volume accompanied by an red cell mass
increase in red cell mass. The plasma
volume expansion peaks at around 24 Platelets
(late pregnancy)
weeks when it is approximately 40%
greater than in a non-pregnant woman.
ΠNumber
Normal pregnancy ΠFe
Ø Volume
As the increase in red cell mass is more
modest (15–25%) a dilutional anaemia is Ø Factors VII, VIII, X
inevitable. In practice the haematocrit Ø Fibrinogen
and haemoglobin level start to fall at 6–8 Fig 44.1  Common haematological changes in normal pregnancy.
weeks and reach a trough at around 20
weeks. It is unusual for the haemoglobin The other major type of anaemia in in pregnancy, occurring in 6–10% of all
level to fall below 100 g/L and if this pregnancy is megaloblastic anaemia. pregnant women. A few women have
happens another cause for anaemia This usually results from deficiency of an obvious systemic disorder such as
should be sought. Negative iron balance folate. As for iron, folate requirements pre-eclampsia; disseminated intravascu-
can be regarded as routine in pregnancy are increased during pregnancy and lar coagulation (DIC) in pregnancy is
and as discussed below frank iron defi- the diet is frequently inadequate to meet further discussed below. However, the
ciency commonly occurs. this demand. Megaloblastic anaemia majority of women are systemically well
The other major changes which may most often presents as a macrocytic with an apparently normal pregnancy.
be regarded as a physiological conse- anaemia in the third trimester or post- In these cases thrombocytopenia can be
quence of pregnancy affect the coagula- partum. It is normal practice to give divided into two categories, with differ-
tion system. There are increases in the folate supplements in pregnancy. The ing clinical implications for the mother
levels of the coagulation factors VII, VIII amount of folate routinely administered and fetus.
and X and a marked increase in plasma orally should be large enough to avoid
fibrinogen. The resulting hypercoagula- megaloblastic anaemia but not so large Incidental (gestational)
bility is helpful in limiting the likeli- as to risk masking pernicious anaemia thrombocytopenia
hood of life-threatening bleeding at with vitamin B12 deficiency which does Incidental thrombocytopenia is the
delivery but it does lead to an increased occasionally occur in pregnancy. Folate most common cause of thrombocytope-
risk of thromboembolism. The platelet deficiency in pregnancy has been linked nia in pregnancy accounting for around
count falls about 10% during an uncom- with an increased incidence of neural 75% of cases. Thrombocytopenia is mild
plicated pregnancy. Later in pregnancy tube defects in the fetus and recommen- to moderate (70–150 × 109/L) and the
there may also be an increase in mean dations for planned pregnancies are the woman is otherwise well. There is no
platelet volume (MPV). use of folate supplements (400 µg daily) past history suggesting a cause for the
prior to conception and then particu- low platelet count and particularly no
larly in the first 12 weeks. Larger doses history of immune thrombocytopenia
Anaemia in pregnancy
of folate are recommended where (ITP). The disorder is not associated
There are several causes of anaemia in women are at high risk of conceiving a with maternal haemorrhage or fetal or
pregnancy. The most common scenario child with a neural tube defect (e.g. pre- neonatal thrombocytopenia. As there is
is an exacerbation of the usual dilutional viously affected pregnancy). There is no diagnostic test it is often difficult to
anaemia by deficiency of iron and/or no justification for the prescription of distinguish gestational thrombocytope-
folate. Erythropoietin levels increase multi-ingredient vitamin preparations nia from mild ITP until a non-pregnancy
less than in anaemic non-pregnant in pregnancy but a combined iron and platelet count is available.
women, possibly suppressed by hormo- folate tablet of adequate dosage may be
nal changes. prescribed. ITP in pregnancy
The identification of iron deficiency It should be remembered that not all The management of pregnancy in a
relies upon normal laboratory tests (p. anaemia in pregnancy is caused by defi- woman with known chronic ITP can be
25). However, even in women with no ciency states. Other blood disorders problematic as severe thrombocytope-
overt clinical deficiency there is a pro- may present in pregnancy and chronic nia may be a threat to the mother and
gressive fall in serum iron through preg- blood diseases such as sickle cell there is also a risk of the child becoming
nancy. Routine dietary supplementation anaemia can be especially difficult to thrombocytopenic. The latter complica-
with modest amounts of iron (e.g. manage at this time. tion arises as the causative IgG antiplate-
ferrous sulphate 200 mg daily) leads to let autoantibody in the mother freely
a significant increase in haemoglobin crosses the placenta and can target fetal
level at term compared with women
Thrombocytopenia
platelets. Fortunately, the majority of
receiving no supplements. Parenteral
in pregnancy
babies escape – severe thrombocytope-
iron is contraindicated in the first After anaemia, thrombocytopenia is the nia (less than 50 × 109/L) occurs in
trimester. most common haematological disorder around 10% of neonates and mortality
Pregnancy 89

Table 44.1  General guidelines for the


management of acute obstetric
haemorrhage
■ Secure venous access and insert a central line to
measure central venous pressure (CVP)
■ Seek additional (preferably senior) medical help
Abruptio placentae ■ Collect samples for urgent blood count,
Amniotic fluid embolism crossmatching and coagulation screen; liaise with
Septic abortion and intrauterine infection haematology laboratory
Retained dead fetus ■ Restore blood volume – may have to use
Hydatiform mole unmatched blood of patient’s ABO and Rh group
Placenta accreta (preferred to group O Rh negative)
Pre-eclampsia and eclampsia ■ Address source of bleeding
■ Blood product replacement as necessary

Fig 44.2  Causes of DIC in pregnancy.


delivery. Current practice is to avoid
use of oral anticoagulants in
from intracranial bleeding in those Both the anticoagulants commonly pregnancy wherever possible. There
affected is less than 1%. All manage- used in clinical practice, heparin and is currently no place for the newer
ment decisions must thus acknowledge warfarin, require special consideration oral anticoagulants (i.e. direct
that fetal thrombocytopenia is uncom- in pregnancy. thrombin and anti-Xa inhibitors) due
mon and fetal mortality very rare. In to concerns regarding toxicity.
■ Heparin. Neither unfractionated
this context, aggressive treatment of all
standard heparin nor low molecular DIC in pregnancy
mothers with ITP with corticosteroids
weight heparin (LMWH) cross the DIC is associated with a wide variety of
and/or intravenous immunoglobulin
placenta. LMWH is widely used and situations in pregnancy (Fig 44.2). The
and routine delivery by caesarean
is both safe and effective in the chief characteristics and pathogenesis of
section are not justified. The fetal plate-
prevention and treatment of venous DIC are discussed on page 76. In preg-
let count is not routinely measured.
thromboembolism in pregnancy, nancy, DIC may manifest as a chronic
A conservative approach with normal
with significant bleeding, usually compensated state or as life-threatening
delivery and an immediate neonatal
from primary obstetric causes, in less haemorrhage. The latter is a frightening
cord blood platelet count is gaining
than 2% of cases. Anti-factor Xa levels medical emergency and there should be
support. If the baby’s count is low or
can be monitored but the optimal a planned regimen of management with
falling, intravenous immunoglobulin
therapeutic range is unclear. input from an obstetrician, haematolo-
can be given. In more severe thrombo-
■ Warfarin. Warfarin is not significantly gist, physician, anaesthetist and nurse
cytopenia, transcranial ultrasound can
secreted in breast milk and treatment (Table 44.1). It is imperative that the
be performed to exclude intracranial
is safe during lactation. However, it source of bleeding is identified and
haemorrhage. Because of its low inci-
readily crosses the placenta and is a addressed as soon as possible. It is often
dence of side-effects, intravenous
known teratogen, producing a shock which triggers DIC with a result-
immunoglobulin is probably the treat-
specific warfarin embryopathy at ant increase in bleeding.
ment of choice for severe maternal
around 6–12 weeks (approximately
thrombocytopenia.
5% incidence). Thus, heparin should HELLP syndrome
be substituted for warfarin in the first HELLP is an acronym for microangio-
trimester. There may be a risk of fetal pathic haemolysis (H), elevated liver
Coagulation haemorrhage secondary to warfarin enzymes (EL) and low platelets (LP).
abnormalities in throughout pregnancy, particularly if The syndrome complicates less than
pregnancy anticoagulant control is poor, and the 1% of all pregnancies but develops in
risk to mother and fetus becomes 10% of women with pre-eclampsia.
Thromboembolism and
unacceptable in the antepartum Both disorders generally remit within
anticoagulant therapy
period. It should therefore be several days after delivery and delivery
Pulmonary embolism (PE) remains a
discontinued at 36 weeks and of the fetus is therefore central to
major cause of maternal death. Approxi-
heparin substituted until after management.
mately half of fatal PEs occur antepar-
tum and half postpartum, the majority
of the latter in the first 2 weeks of the Pregnancy
puerperium. About 70% of women who ■ Normal pregnancy is accompanied by a modest dilutional anaemia.
develop venous thromboembolism in
■ Deficiency of iron and/or folate frequently exacerbates the normal dilutional anaemia.
pregnancy and the puerperium have
■ Thrombocytopenia is most often ‘incidental’ and of little significance. Immune
major risk factors. These include increas-
thrombocytopenia (ITP) may require treatment but a normal delivery is usual and severe
ing age, caesarean section, obesity, previ-
neonatal thrombocytopenia is rare.
ous thrombotic problems and familial
■ There is a hypercoagulable state in pregnancy and pulmonary embolism remains a major
thrombophilia. Hereditary throm-
cause of maternal death. Low molecular weight heparin is generally the preferred
bophilia (see p. 78) has also been linked anticoagulant for treatment or prevention of thrombosis.
with recurrent fetal loss, intrauterine
■ Disseminated intravascular coagulation (DIC) can complicate pregnancy and cause
growth restriction, pre-eclampsia and life-threatening haemorrhage.
placental abruption.
90 9 SPECIAL SITUATIONS

45 Paediatric haematology
Many of the blood disorders encountered Table 45.1  Normal white cell counts in children (× 109/L)
in children have been discussed in the Age White cell count Neutrophils Lymphocytes
preceding pages. For instance, acute lym- Birth (full term) 18 ± 8 5–13 3–10
phoblastic leukaemia is the most common Day 3 15 ± 8 3–5 2–8
leukaemia of childhood, haemophilia is 1 month 12 ± 7 3–9 3–16
usually diagnosed in infancy and the hae- 2–6 months 12 ± 6 1.5–9 4–10
moglobinopathies are a significant cause 2–6 years 10 ± 5 1.5–8 6–9
of ill health in children worldwide. 6–12 years 9±4 2–8 1–5
Chronic and severe diseases of the blood
Note: Normal haemoglobin values in childhood are shown on page 22, Table 1. The normal platelet count is the same in children and adults
pose particular problems in childhood (150–400 × 109/L).
and usually are best managed by a
against HDN due to RhD incompatibility
paediatrician with a special interest in
(see below), the most common cause of
haematology or in a combined paediatric/
the disorder is the formation of immune
haematology clinic. The child’s growth
antibodies against ABO; most cases are
and development, and educational needs
associated with only mild haemolysis.
often require special attention. In this
section we discuss some haematological Diagnosis
disorders encountered in paediatric prac- Severe HDN can result in intrauterine
tice which are not addressed elsewhere. death. In the newborn child the presenta-
tion is entirely dependent on the degree of
Normal values haemolysis but common features include Fig 45.1  Peripheral blood film in a newborn
child with severe HDN. Note the numerous
anaemia, jaundice, oedema and hepat-
It is important to appreciate that the nucleated red cells and polychromasia.
osplenomegaly. High levels of circulating
normal ranges for many haematological
unconjugated bilirubin may lead to high RhD-negative mothers who deliver a
tests vary with age. Table 45.1 illustrates
frequency deafness or deposition in the RhD-positive infant. A larger than average
reference values for the total white cell
basal ganglia with spasticity and other feto-maternal haemorrhage necessitates
count (WCC) and the differential count in
neurological symptoms and signs (‘ker- a greater dose of anti-D Ig. It is most
children. More detailed listings of normal
nicterus’). Further investigation of the likely that anti-D administration prevents
ranges of laboratory tests in childhood
anaemia reveals features typical of haemo- HDN by a negative modulation of the
can be found in specialised paediatric hae-
lysis (Fig 45.1) with a positive direct anti- primary immune response rather than
matology texts.
globulin test (DAT). In HDN due to RhD by simple removal of fetal RhD-positive
incompatibility the baby is RhD positive cells. General recommendations for Rh
Neonatal disorders
and the mother RhD negative with a high prophylaxis are shown in Table 45.2.
Haemolytic disease of level of anti-D. As some women undoubtedly become
the newborn sensitised earlier in a normal pregnancy,
Management
Haemolytic disease of the newborn routine antenatal prophylaxis is widely
Management of HDN is complex, requir-
(HDN) is a disease of the fetus and recommended.
ing close liaison between the haematol-
newborn child. The haemolysis is caused
ogy laboratory and obstetrician. In RhD Anaemia of prematurity
by maternal IgG antibodies traversing the
alloimmunisation, if maternal anti-D The haemoglobin concentration falls after
placenta and attaching to fetal red cells
levels are high and paternal testing birth in all babies but in premature infants
which are destroyed in the child’s reticu-
indicates RhD heterozygosity, the fetal it falls faster and to a lower level. At 1–3
loendothelial system. The antibodies are
Rh genotype can be determined non- months of age haemoglobin concentra-
directed against a fetal red cell antigen not
invasively by applying PCR technology tions of less than 70 g/L are common and
shared by the mother. Incompatibility for
to a maternal blood sample. Another in babies born at less than 32 weeks gesta-
one of a large number of different red cell
advance is velocimetry of the fetal middle tion this anaemia is often associated with
blood group systems can cause HDN but
cerebral artery during an affected preg- inadequate adaptive responses including
most cases of clinically significant disease
nancy. High peak systolic velocities predict tachycardia, tachypnoea and apnoeic
affect a Rhesus (Rh)D-positive child where
severe fetal anaemia and allow the selec- attacks. The anaemia is due in part to
the mother is RhD negative. Sensitisation
tive use of more invasive techniques such shortened red cell lifespan and the effects
of the mother (i.e. the formation of anti-D)
as fetal blood sampling and intrauterine of rapid growth but the fundamental
occurs following the haemorrhage of fetal
transfusion. Newborns may experience problem appears to be a poor erythropoi-
red cells into the maternal circulation.
ongoing anaemia and require exchange etin response. Erythropoietin levels are
This usually occurs at parturition follow-
transfusion. Later anaemia may respond highest in premature infants with the
ing a normal pregnancy but may also
to erythropoietin therapy. With optimal most severe anaemia and hypoxia but
arise earlier in pregnancy or following
management, a healthy child is the even in these cases levels are inadequate
abortion. ABO incompatibility between
outcome in more than 90% of cases. compared to those achieved in anaemic
mother and fetus gives some protection
adults. Recombinant erythropoietin is of
against sensitisation to RhD as fetal red RhD prophylaxis in RhD-negative
benefit in some infants.
cells are quickly destroyed by the mother’s mothers
naturally occurring anti-A or anti-B anti- The breakthrough in the prevention of Polycythaemia in the neonate
bodies. Unfortunately, in most cases baby HDN has been the introduction of proph- Polycythaemia in the neonate is most
and mother are ABO compatible. With ylaxis (Fig 45.2). A dose of Rh anti-D simply defined as a packed cell volume
the considerable success of prophylaxis immunoglobulin (Ig) is given to all (PCV) exceeding 0.65. Causes include
Paediatric haematology 91

Table 45.2  Recommendations for Table 45.3  Some causes of disorders, and malignancies (e.g.
Rh prophylaxis1 thrombocytopenia in the neonate thymoma). However, two types of
Rh prophylaxis after delivery ■ DIC in various severe systemic disorders PCRA are unique to childhood:
Anti-D (usually 500 IU) is given within 72 hours in ■ Intrauterine infection (e.g. rubella, cytomegalovirus) Diamond–Blackfan anaemia and tran-
RhD-negative mothers where the infant is RhD positive ■ Platelet antibodies: sient erythroblastopenia.
(or group undetermined). If there is a large – autoimmune (maternal ITP)
feto-maternal haemorrhage (assessed in a Kleihauer – alloimmune Diamond–Blackfan anaemia
test) additional anti-D is given – drugs This is a rare heterogeneous disorder
Rh prophylaxis and abortions ■ Hereditary/congenital disorders:
caused by defects in structured ribosomal
In RhD-negative mothers anti-D is given after all – Wiskott–Aldrich syndrome
– thrombocytopenia with absent radii (TAR) proteins. The majority of cases are spo-
therapeutic abortions and after spontaneous or
threatened abortions later than 12–13 weeks’ syndrome radic but various patterns of inheritance
gestation and in selected cases of threatened abortion ■ Post exchange transfusion have been documented. An anaemia with
■ Neonatal leukaemia
before 12 weeks (usual dose 250 IU before 20 weeks the features of red cell aplasia usually
and 500 IU after 20 weeks) ■ Giant haemangioma
presents within the first 12 months of
Rh prophylaxis during pregnancy
life. This runs a chronic course and can
Anti-D is given after possible sensitising events in Immune thrombocytopenia (ITP) may be
be combined with developmental abnor-
RhD-negative women. These include: amniocentesis, seen in infants born to mothers with ITP
chorionic villus sampling, abdominal trauma, external malities. There is an increased risk of
where there is passive transfer of IgG
cephalic version, antepartum haemorrhage, ectopic haematological malignancy and other
across the placenta. Alloimmune throm-
pregnancy (usual dose of anti-D is 250 IU before 20 cancers. Beyond blood transfusion, thera-
weeks and 500 IU after 20 weeks). Anti-D (500 IU) bocytopenia arises where the healthy
peutic options include corticosteroids,
should be given to non-sensitised RhD-negative mother becomes sensitised against a fetal
mothers at 28 and 34 weeks ciclosporin and allogeneic stem cell
platelet antigen in a manner analogous to
transplantation.
1
United Kingdom guidelines.
HDN; the platelet antigen HPA-1a is most
commonly implicated. Transient erythroblastopenia
of childhood
placental transfusion (e.g. delayed clamp-
Iron deficiency in infancy This is a transient form of red cell aplasia
ing of the cord), intrauterine hypoxia,
of probable immune origin which must
endocrine disorders (e.g. maternal diabe- Iron deficiency has already been discussed
be distinguished from Diamond–Blackfan
tes) and genetic disorders (e.g. trisomy 21). (p. 24) but some aetiological factors in
anaemia. It generally affects older children
Polycythaemia is often well tolerated but infancy are unique to this period of life.
(1–4 years) and may be diagnosed simul-
if severe it may cause hyperviscosity with Blood loss may still be the major cause
taneously in siblings or in seasonal clus-
congestive heart failure, respiratory dis- of deficiency but other factors worthy of
ters. In over half of cases there is a previous
tress, neurological disturbances and even consideration are decreased total body
viral illness. The normocytic anaemia may
gangrene. Partial exchange transfusion iron at birth (e.g. prematurity, feto-
be accompanied by mild neutropenia. Full
using a crystalloid solution to reduce the maternal haemorrhage, twins), the impact
recovery within 4–8 weeks is the rule.
haematocrit is indicated where a high of growth with increased demands for
PCV is associated with symptoms and iron, and dietary inadequacy (e.g. exces-
signs of hyperviscosity. sive dependence on unsupplemented Congenital
cow’s milk). dyserythropoietic
Thrombocytopenia in
anaemias (CDAs)
the neonate
Red cell aplasia
Some causes of thrombocytopenia in This is a group of rare inherited anaemias.
in childhood
neonates are listed in Table 45.3. In prac- There are various subtypes but common
and adolescence
tice the major divide is between seriously features include ineffective erythropoiesis
ill infants where the low platelet count is Pure red cell aplasia (PRCA) is character- and multi-nucleated erythroblasts. The
caused by disseminated intravascular ised by anaemia, reticulocytopenia and white cell and platelet counts are normal.
coagulation (DIC), and relatively well reduced or absent erythroid precursor Anaemia is usually first diagnosed in
infants where thrombocytopenia is most cells in the bone marrow. There are many infancy or childhood. It may be of normo-
often of immune aetiology or occurs sec- causes of PCRA including infection cytic or macrocytic type. Transfusion is
ondary to a specific inherited syndrome. (e.g. parvovirus B19), connective tissue required in more severe cases.

Deaths/
1000 births
2.0
Improved obstetric
1.8 and neonatal care Paediatric haematology
1.6
1.4 ■ Chronic and severe blood disorders in children are usually best
1.2 managed by a paediatrician with a special interest in haematology
Exchange transfusion

or in a combined paediatric/haematology clinic.


1.0
Premature delivery

Rh Ig prophylaxis

■ In haemolytic disease of the newborn (HDN), haemolysis is caused


0.8
Amniocentesis

by maternal IgG antibodies crossing the placenta and attaching to


0.6
Intrauterine
transfusion

fetal red cells. Most clinically significant cases affect a RhD-positive


0.4 fetus or newborn child where the mother is RhD negative.
0.2
■ RhD prophylaxis has much reduced the incidence of severe HDN.
0.0
1950 1960 1970 1980 ■ Prematurity is associated with a particular type of anaemia.
Year
■ In pure red cell aplasia in children it is important to distinguish
Fig 45.2  Perinatal deaths/1000 births caused by HDN. (From Derrick between Diamond–Blackfan anaemia and the more benign
Tovey, LA 1992 Haemolytic disease of the newborn and its prevention. In: transient erythroblastopenia of childhood.
Contreras, M (ed) ABC of Transfusion. BMJ Publishing.)
92 9 SPECIAL SITUATIONS

46 Haematology in the elderly


Average life expectancy is increasing. mainly due to a combination of age- particular considerations. Patients over
Elderly people (>65 years) are likely to related suppression of erythroid colony 60 years usually require lower doses of
make up more than 20% of the world’s formation, insensitivity to erythropoie- vitamin K antagonists to maintain a
population by 2050. This effect is par- tin and impaired iron utilisation. therapeutic range than younger patients.
ticularly marked in the developed world Whatever its aetiology, anaemia in the Some studies have suggested an
where in some countries the population elderly is a relevant finding. It is associ- increased risk of adverse events such as
over 65 years already outnumbers those ated with reduced physical and cogni- bleeding in older patients on anticoagu-
below 20 years. An ageing population tive functioning, an increased chance of lants. It is not clear whether this is
has implications for the practice of clini- falls, an aggravation of comorbidity such related to age alone or the presence of
cal haematology. There are significant as cardiac and neurological disease, and comorbidity. It is sensible to monitor
age-related changes in haematopoiesis reduced survival. A low haemoglobin elderly patients more frequently. It is
and haemostasis. Many blood diseases, level should not be readily dismissed as likely that the newer oral anticoagulants
especially malignant disorders, are more part of ‘normal ageing’. (see p. 102) will also have to be used
common in older people. The manage- Where the anaemia has an explained cautiously in this group of patients.
ment of blood diseases in the elderly is cause (e.g. iron deficiency), treatment is
often complicated by frailty, comorbid- specific for that disorder. Erythropoeitin
Haemophilia and
ity and a need for extra care in the hos- may be used in chronic renal failure.
other inherited
pital and community. Blood transfusion is needed in only a
bleeding disorders
minority of cases (e.g. for the sympto-
matic anaemia of myelodysplastic syn- Haemophilia is not generally thought of
Haematopoiesis
drome), and in the very elderly and frail as a disease of the elderly. Recent
and ageing
must be undertaken cautiously to avoid changes in management such as the
There is an age-associated decline in fluid overload and the exacerbation of prophylactic use of coagulation factors
haematopoietic stem cells and haemat- cardiac failure. The treatment of unex- have focused on children and young
opoietic cell function. The reasons for plained anaemia in the elderly remains adults. However, at least in the West,
this are unclear and much of our knowl- controversial with no clear guidelines. regular coagulation factor replacement
edge is derived from studies of mouse means that life expectancy is now
models rather than humans. It is prob-
Thrombosis and
able that this decline arises from a com-
anticoagulation
bination of accumulated genetic and
epigenetic changes, alterations in the Old age is a relatively hypercoagulable Nutritional
deficiency
marrow microenvironment and sys- state. Ageing leads to increased plasma
temic effects such as inflammation. Prac- levels of fibrinogen and various clotting
tical consequences of these biological factors and delayed fibrinolysis. The
Anaemia of Anaemia
changes include reduced immune func- incidence of both arterial and venous chronic disease unexplained
tion, an increased incidence of cancer, thrombosis increases with age (Fig 46.2).
and a lesser tolerance of haematopoietic Advanced age is not in itself a contrain-
stress (e.g. chemotherapy). Stem cells dication to the use of anticoagulant
from older donors are less efficient at therapy – older patients may derive the
reconstituting haematopoiesis in trans- greatest benefit – but there are Fig 46.1  Causes of anaemia in the elderly.
plant recipients.
500

Anaemia
400
Incidence rate per 100,000/year1

Anaemia is a common clinical problem in the elderly. The


prevalence rises rapidly after 50 years and approaches 20% in
people aged over 80 years. In general, one third of cases will 300
have an identifiable nutritional deficiency (iron, vitamin B12 or
folate) (Fig 46.1). Where iron deficiency is the cause of anaemia
200
it is often secondary to gastrointestinal blood loss, and under-
lying bowel pathology (e.g. colonic carcinoma) should be
excluded. Another third of cases have the anaemia of chronic 100
disease. These patients have an obvious chronic inflammatory
condition (see p. 36) and will often have a measurable acute
phase response (e.g. elevated C-reactive protein). In the final 0
third of elderly patents, there is no clear cause for the anaemia 0 20 40 60 80
(sometimes termed ‘anaemia unexplained’). This entity is a Age (years)
diagnosis of exclusion and has been the focus of much recent 1 New episodes of venous thromboembolism
interest. A few cases may be explained by myelodysplastic Fig 46.2  The incidence of venous thrombosis increases
syndrome or other rarer causes of anaemia but it is probably exponentially with age.
Haematology in the elderly 93

approaching that of the whole male


population. This means that older Acute myeloid leukaemia 67
patients increasingly present with both
Acute lymphoblastic leukaemia 13
the clinical problems of haemophilia
(e.g. joint arthropathy and pain, blood- Chronic myeloid leukaemia 65
borne infections) and age-related ill-
nesses such as cancer and cardiovascular Chronic lymphocytic leukaemia 72
disease. The treatment of such comor-
bidities routinely involves invasive prac- Hodgkin’s lymphoma 38
tical procedures and drugs which further
derange haemostasis, and patients will Non-Hodgkin’s lymphoma 66
often need intensive coagulation factor
replacement to counter the increased Myelodysplastic syndrome 76
risk of bleeding. The same considera-
Myeloma 69
tions apply to other inherited bleeding
disorders. 0 10 20 30 40 50 60 70 80 90 100
Age (years)
Haematological Fig 46.3  Median ages of presentation of haematological malignancies.
malignancy and
In the very elderly (>80 years), it may
chemotherapy Table 46.1  Guidelines for the management
be judged that intensive chemotherapy of chemotherapy in older cancer patients
Haematological malignancy can occur at is contraindicated. This might also apply Geriatric assessment for patients over 70 years
any age but most types are more to younger patients with very significant Adjust doses to individual GFR in patients over 65
common in the elderly. The median age comorbidity. Where chemotherapy is years
Prophylactic use of G-CSF in patients over 65 years
at diagnosis for all blood neoplasms undertaken, this should follow a careful
receiving regimens comparable to CHOP1
combined is around 70 years (Fig 46.3). assessment of the benefit and risk to the Maintenance of haemoglobin level around 120 g/L
There is a male excess except for over 80 individual patient. A formal geriatric Use of drugs with low toxicity where feasible
years when more women are diagnosed. assessment should be considered in
GFR: glomerular filtration rate; G-CSF: granulocyte colony-
This may be because of greater female patients 70 years and older. This can stimulating factor.
life expectancy. Some haematological identify potential reversible conditions 1
See page 61.
malignancies in the elderly are diag- which might exacerbate the complica-
nosed incidentally, are indolent, and tions of treatment including anaemia,
require little or no intervention. Exam- depression, poor nutrition and a lack of as they potentially combine high levels
ples are described in other sections and available care and social support. Other of efficacy with fewer side-effects than
include monoclonal gammopathy of practical steps to reduce the complica- conventional chemotherapy.
undetermined significance (MGUS), tions of chemotherapy are dose adjust- Population studies show lower sur-
monoclonal B-cell lymphocytosis and ment for renal dysfunction (based on vival rates for older people with all types
early stage chronic lymphocytic leukae- the glomerular filtration rate), the pro- of haematological malignancy. This
mia. Other neoplasms are aggressive phylactic use of growth factors (e.g. group of patients is generally under-
and will reduce quality of life and shorten granulocyte colony-stimulating factor represented in formal clinical trials
life expectancy in the absence of effective (G-CSF)) to shorten the period of mye- of chemotherapy. Many are excluded
treatment. losuppression and the selection of drug because of multiple comorbidities.
Older patients tolerate chemotherapy regimens with relatively low toxicity. An Where older patients are entered into
less well than younger patients and they example of the latter is the avoidance of such trials the results must be inter-
are less likely to be cured of their malig- anthracycline drugs in patients with preted cautiously as the selective nature
nancy (Table 46.1). Changes in pharma- cardiac failure. Targeted therapies (e.g. of trial entry means that the results may
codynamics account for part of this imatinib in chronic myeloid leukaemia) not be applicable to the whole elderly
reduced effectiveness and increased tox- are particularly attractive in the elderly population.
icity. Reducing the dose of chemother-
apy drugs to minimise side-effects will
reduce cure rates. Other factors leading
to poorer outcomes in the elderly are
Haematology in the elderly
increased comorbidity and adverse
disease characteristics. For example, ■ An ageing population has implications for the practice of clinical haematology. Many blood
elderly patients with acute myeloid leu- disorders (e.g. malignant diseases) are commoner in the elderly.
kaemia have a higher incidence of poor ■ There are significant age-related changes in haematopoiesis and haemostasis.
risk cytogenetic changes compared with ■ Anaemia is a common clinical problem in the elderly; in a third of cases there is no clear
younger patients. They also have an cause.
increased number of significant chemo- ■ Old age is a relatively hypercoagulable state.
therapy side-effects including more pro-
■ Older people tolerate chemotherapy less well than younger people and are less likely to be
found myelosuppression, neurotoxicity cured of their haematological malignancy.
and cardiotoxicity.

tahir99-VRG & vip.persianss.ir


94 9 SPECIAL SITUATIONS

47 Palliative care in haematological malignancy


The World Health Organization defines palliative care as ‘an
approach that improves the quality of life of patients and their
Disease-modifying/
families facing the problems associated with life-threatening curative treatment
illness, through the prevention and relief of suffering by
Focus
means of early identification and impeccable assessment and of care
treatment of pain and other problems, physical, psychosocial
and spiritual’. As the modern specialty of palliative medicine Palliative care
was born in the hospice movement, there has been a tendency
to regard it as an end-of-life intervention, an option only when
Presentation Death
disease-modifying cancer treatments have been exhausted. It Time
Bereavement
is more correctly viewed as being entirely complementary to care
ongoing anti-tumour treatment – for instance, a patient with
Fig 47.1  Current model of cancer management.
myeloma presenting with refractory bone pain is likely to
benefit from expert symptom control early in their illness (Fig
47.1). Good palliative care requires a multidisciplinary team Home/
primary care
approach with staff expert in communication and control of
symptoms and with the organisational skills to coordinate
care. Patients commonly derive palliative care input from
diverse sources (Fig 47.2). There is not space here to compre- Hospital Home/hospice
hensively review all aspects of palliative care but a number of outpatient
applications with particular relevance to patients with haema- Patient
tological malignancy will be discussed.

Palliative chemotherapy
and radiotherapy Hospital
inpatient
Hospice
inpatient
Chemotherapy and radiotherapy are primarily employed as
disease-modifying treatments but they may also be used in a
palliative context to relieve symptoms and improve quality of
Fig 47.2  Schematic view of a patient’s access to palliative care
services.
life. Any side-effects of such treatment must be carefully
weighed against the likely symptom control. Chemotherapy likely to require an opiate analgesic given at an appropriate
may thus be used to limit the degree of troublesome lym- dose and interval, possibly in combination with a specific
phadenopathy in advanced lymphoma or to reduce the sys- co-analgesic dependent on the nature of the pain. There is no
temic upset from a high malignant cell burden in end-stage one optimal opiate dose and the correct dose must be achieved
leukaemia. Similarly, attenuated radiotherapy can give local by proactive titration so that the patient is pain free without
relief from tumour infiltration in lymphoma and can reduce unacceptable toxicity. For most patients with chronic cancer
pain from myeloma bone lesions (Fig 47.3). Surgical interven- pain, the oral route is preferable but parenteral, transdermal
tions are used more sparingly but lymphomatous pleural and rectal preparations are also widely employed. Pain pre-
effusions can be drained and the severe pain of myelomatous vention must be complemented by management of drug side-
spinal disease can be ameliorated by the operation of verte- effects – patients on opiates will generally need laxatives and
broplasty where the vertebrae are reinforced with a cement- antiemetics. Particular care is necessary when analgesics are
like substance. changed. This requires an understanding of their relative
strengths and durations of action.
Opiates may be combined with co-analgesics to maximise
Management of cancer pain
their effect. Examples in haematological practice include the
Pain is a common symptom of cancer, particularly in patients use of corticosteroids (usually dexamethasone) to tackle
with advanced or refractory disease. The means are available symptoms of raised intracranial pressure or nerve infiltration
to give the great majority of patients good quality pain relief secondary to lymphoma and the prescription of NSAIDs and
but, in practice, this goal may be compromised by the time, bisphosphonates to optimise control of bone pain in myeloma.
skill and commitment required of the medical team. Before Tricyclic antidepressants can be a useful adjunct in neuro-
initiating therapy, it is important to fully assess the severity pathic pain.
(pain scales are available) and nature of the pain – visceral
pain is often described as a dull ache, somatic pain may be
sharp and postural, while neuropathic pain can be ‘burning’
Control of non-pain symptoms
or ‘numbing’. Fatigue. In the haematology clinic, many patients with leu-
Pharmacological therapy is the mainstay of pain manage- kaemia, lymphoma and myeloma complain of demoralising
ment in patients with haematological malignancy. Patients fatigue. This is usually of multifactorial aetiology with a
experiencing mild pain on no analgesia may be commenced mixture of physiological and psychosocial factors, the latter
on an oral non-opiate agent (e.g. paracetamol, nonsteroidal including anxiety and sleep disruption. Anaemia is the most
anti-inflammatory drug (NSAID)) but more severe pain is common reversible physical cause and regular administration

tahir99-VRG & vip.persianss.ir


Palliative care in haematological malignancy 95

Fig 47.4  A syringe driver used to give continuous drug infusions.

bleeds into the lung parenchyma. Once distress caused by setbacks and the
correctable causes of dyspnoea have toxicity of treatment. Psychotherapeutic
been excluded, other options include approaches may be on an individual or
the selective administration of oxygen, group basis and have been shown to
opiates and sedation to minimise the diminish any sense of isolation and to
subjective sensation of breathlessness. boost optimism. The provision of good
Care at of the end of life. In patients quality information, exploiting the
with advanced disease and refractory internet and other new technologies, is
symptoms in the final days of life, seda- complementary to this process. Where
Fig 47.3  The need for palliative care in a
patient with myeloma. An MRI scan of the tion may be used in the form of continu- there is significant anxiety and depres-
spine in a patient with myeloma showing ous drug infusions via portable syringe sion – and correctable causes have been
vertebral collapse. Back pain is a common drivers (Fig 47.4). Strict criteria must be excluded – anxiolytics and antidepres-
symptom in this disease. used for patient selection and fully sants can be beneficial. Support must
informed consent must be obtained also be given to the patient’s family, par-
from the patient (where possible) and ticularly at the time of bereavement.
of subcutaneous erythropoietin their family. Such measures should only
improves haemoglobin levels and be resorted to after consultation with
Complementary therapy
quality of life in selected patients with the palliative care team.
myeloma, lymphoma and other cancers. Patients may turn to complementary
Nausea and vomiting. These symp- and alternative medicine. Staff specialis-
Psychosocial oncology
toms are common, both due to tumour ing in traditional cancer medicine may
infiltration and as a side-effect of chemo- It is important (but not always easy) to be ignorant of and even threatened by
therapy and analgesics. It is crucial distinguish the inevitable emotional such modalities. However, they should
to identify the cause. A centrally acting upset following a diagnosis of a life- not hesitate to advise against any unor-
antiemetic drug (e.g. ondansetron) is threatening disease such as leukaemia thodox intervention which is likely to
indicated for drug-induced nausea from a more severe disturbance that lead to harm. There is some evidence for
whereas a pro-kinetic agent (e.g. meets the criteria for a mental disorder. the use of hypnosis in pain relief, for
metoclopramide) is more appropriate In practice, there is a need for a multidis- acupuncture in the management of
for gastric stasis or functional bowel ciplinary strategy with the involvement drug-induced nausea and vomiting, and
obstruction. In more difficult cases, it is of specialist haematology/oncology for massage and meditation techniques
important to combine antiemetics logi- nurses, psychologists, psychiatrists and in decreasing distress and improving
cally and to consider alternative routes social workers. High quality communi- sense of well-being. Centres specialising
of administration. cation between the patient and their in the treatment of haematological
Anorexia and cachexia. Patients physician and other members of malignancy are increasingly offering
with loss of appetite and weight loss the caring team is crucial to blunt the these therapies to their patients.
need expert dietary assessment and
advice. Oral corticosteroids can improve
Palliative care in
appetite and lead to weight gain. Poor
haematological malignancy
nutrition may be exacerbated by drug-
induced mucositis, painful neutropenic ■ Palliative
care is designed to improve the quality of life of patients and their families – it is
mouth ulcers and oral infections such as complementary to ongoing anti-cancer treatment.
candida and herpes simplex. This can be ■ Optimal provision of palliative care requires a multidisciplinary team with specialist skills.
minimised by good mouth care and ■ Good communication is a vital therapeutic tool.
early recognition of infections.
■ Chemotherapy and radiotherapy may be used to palliate symptoms.
Dyspnoea. Lymphoma can cause
■ Pain is a common symptom of haematological malignancy and is often poorly treated.
dyspnoea because of bulky mediastinal
Patients with advanced disease may require an actively titrated opiate analgesic and an
lymphadenopathy or lung infiltration.
appropriate co-analgesic.
Patients with advanced leukaemia more
■ Normal emotional distress following the diagnosis of a haematological cancer must be
commonly develop the symptom during
distinguished from mental disorders requiring specific treatment.
fulminant respiratory infections or after

tahir99-VRG & vip.persianss.ir


96 9 SPECIAL SITUATIONS

48 Systemic disease
Clinical haematologists spend a consid- Table 48.1  Haematological changes in renal disease
erable part of their time investigating Abnormality Clinical association
blood abnormalities in patients with dis- Red cells Anaemia Chronic renal failure
eases of other organ systems. Some of Polycythaemia Renal carcinoma, cystic disease, hydronephrosis, parenchymal
the more common diagnostic chal- disease, Bartter’s syndrome, renal transplantation
Burr cells Renal failure
lenges are discussed here.
Haemostasis Abnormal platelet function Renal failure
Thrombocytopenia
Renal disease Disordered coagulation1

Diseases of the kidney are associated


1
The complex coagulation abnormalities of renal failure usually lead to a bleeding tendency but nephrotic syndrome is associated with an
increased incidence of thrombosis.
with a remarkably wide range of possi-
ble haematological abnormalities (Table seen in cancer patients. The mechanism
48.1). is thought to be activation of the normal
Anaemia is almost inevitable in clotting system with low-grade intravas-
chronic renal failure. The pathogenesis cular coagulation and secondary fibri-
is complex but impaired erythropoietin nolysis. In the laboratory, common
production is the principal cause. Other findings are elevated levels of clotting
possible contributory factors include the factors and a shortened prothrombin
release of inhibitors of erythropoiesis, time and activated partial thromboplas-
mild haemolysis and iron deficiency. tin time. It is presumed that cancer cells
The anaemia of renal failure is typically secrete thromboplastin which initiates
normocytic and normochromic. A char- clot formation. Treatment of venous
acteristic finding in the blood film is the Fig 48.1  Burr cells in the blood in renal thrombosis in malignancy is difficult, as
failure.
presence of burr cells (Fig 48.1). The best anticoagulant control is often poor. Low
treatment of anaemia is resolution of worsen bleeding by interfering with the molecular weight heparin is generally
the underlying renal problem (e.g. by normal interaction between platelets preferred to warfarin.
transplantation), but where this is not and vascular endothelium. Disseminated intravascular coagula-
feasible, recombinant erythropoietin is tion (DIC) can complicate malignancy.
the treatment of choice. Intermittent It may be an acute haemorrhagic state
Liver disease and alcohol
bolus administration generally leads to but is more often a chronic low-grade
a marked improvement in anaemia and Significant liver disease is associated disorder with no bleeding. It is particu-
transfusion independence. A failure of with haemostatic problems (see p. 77) larly likely to accompany carcinomas of
the anaemia to respond to erythropoie- and red cell abnormalities including the prostate, stomach, colon, breast,
tin should prompt a search for other macrocytosis and target cells. Excessive ovary, lung, gallbladder and melanoma.
aetiologies such as iron deficiency. alcohol consumption commonly causes DIC is further discussed on page 76.
Paradoxically, some forms of renal macrocytosis and thrombocytopenia.
disease can lead to increased red cell
Connective tissue
production and clinical polycythaemia
Malignancy disorders
(see Table 48.1). This arises either from
inappropriate secretion of erythropoie- Anaemia is seen in around half of Systemic disorders such as rheumatoid
tin by a kidney tumour or from local patients with non-haematological malig- arthritis, systemic lupus erythematosus
renal hypoxia promoting erythropoietin nant tumours. The anaemia of chronic (SLE) and mixed connective tissue
release from normal cells. Polycythae- disease is the most common aetiology disease often lead to abnormal blood
mia can be the presenting feature of (p. 36) but other causes include chemo- counts. In practice the most common
renal carcinoma and rapid identification therapy, blood loss, haemolysis and finding, particularly in rheumatoid
of the malignancy may allow curative marrow infiltration. Invasion of the arthritis, is the anaemia of chronic
surgical treatment. Benign diseases such bone marrow by solid tumours can disease. Immune thrombocytopenia is
as polycystic disease and hydronephro- result in a pancytopenia and a character- more often seen in SLE and this hetero-
sis probably cause polycythaemia by istic leucoerythroblastic blood picture geneous disorder may also be compli-
inducing renal ischaemia. The poly- with circulating nucleated red cells and cated by the presence of the lupus
cythaemia of renal disease is not an myelocytes (Fig 48.2a). Clumps of malig- anticoagulant (p. 79). Neutropenia can
appropriate physiological response and nant cells may be seen in a bone marrow arise in several connective tissue disor-
patients with high haematocrits can aspirate but a bone marrow trephine is ders; the triad of long-standing rheuma-
derive benefit from regular venesection. a more reliable way of demonstrating toid arthritis, splenomegaly and
Chronic renal failure is also associated solid malignancy (Fig 48.2b). neutropenia is termed Felty syndrome.
with a large number of possible platelet Malignancy can be associated both There may be a small increased risk
and coagulation abnormalities. The with a hypercoagulable state and a of haematological malignancy in
increased risk of bleeding in these bleeding tendency. The presence of patients with rheumatoid arthritis. In
patients is generally caused by the hypercoagulability was first suggested Sjögren’s syndrome there is a substan-
complex interaction of abnormalities by the increased incidence of deep vein tially increased risk of non-Hodgkin’s
shown in Table 48.1. Anaemia tends to thrombosis and pulmonary embolism lymphoma.

tahir99-VRG & vip.persianss.ir


Systemic disease 97

(a) (b)
Fig 48.2  Patient with prostatic carcinoma and invasion of the bone marrow. (a) Leucoerythroblastic blood picture: note the nucleated red cell and
myelocyte. (b) Bone marrow trephine specimen showing replacement of normal haematopoiesis by carcinoma.

adults. Clinical features often include corticosteroids can be helpful in unusu-


Infections
malaise, fever, pharyngitis, lymphaden- ally difficult cases.
Infections are probably the most opathy, splenomegaly and hepatitis.
common cause of abnormal blood There is a small risk of splenic rupture. HIV infection
counts in a typical haematology labora- The haematological hallmark of the Progressive HIV infection has many
tory. Different infections are associated disease is the presence of numerous possible haematological consequences
with different abnormalities but it is atypical lymphocytes in the blood (Fig (Table 48.2). These result from a combi-
possible to make some generalisations. 48.3). These lymphocytes are mainly nation of a direct effect of the virus,
Bacterial infections commonly cause a activated T-cells produced as an immu- opportunistic infection and side-effects
neutrophil leucocytosis. The neutrophils nological response to EBV-infected from the drugs used in treatment. The
are classically ‘left-shifted’ (i.e. reduced B-lymphocytes. Other possible blood blood changes are often similar to those
nuclear segmentation) with increased changes are neutropenia, thrombocyto- seen in other viral infections but a
cytoplasmic granulation (toxic granula- penia and a cold-type autoimmune chronic decline in the lymphocyte count
tion). Very severe bacterial infections haemolytic anaemia. The differential is a particular feature. Examination of
such as disseminated tuberculosis can diagnosis is essentially other viral dis- the bone marrow often reveals non-
induce a leukaemoid reaction with eases, but where the blood abnormali- specific features such as changes in
immature myeloid cells appearing in ties are severe the disease may be cellularity, fibrosis, trilineage myelodys-
the blood. confused with acute lymphoblastic leu- plasia, increased plasma cells and prom-
Viral infections most commonly cause kaemia. The diagnosis is supported by inent haemophagocytosis. The presence
a transient lymphocytosis with reactive positive Paul–Bunnel or Monospot tests of granulomas can signify infection by
changes in the cells. Two types of viral which rely on the detection of heter- atypical mycobacteria or other oppor-
infection merit more detailed descrip- ophile antibodies that appear in the tunistic pathogens. In clinical practice
tion: infectious mononucleosis and HIV serum. If these tests are negative and the major haematological problems
infection. there remains clinical suspicion of the associated with HIV infection are
disorder then EBV-specific serodiagnos- immune thrombocytopenia (ITP) and
Infectious mononucleosis tic tests should be performed. Treat- lymphomas. The latter are typically
Infectious mononucleosis (or glandular ment of infectious mononucleosis aggressive B-cell malignancies with
fever) is a disorder caused by the is essentially symptomatic, although extranodal involvement.
Epstein–Barr virus (EBV). It predomi-
nantly affects adolescents and young Systemic
Table 48.2  Possible haematological disease
changes in HIV infection
■ Renal disease can cause anaemia,
Blood Lymphopenia
Anaemia
polycythaemia and abnormalities in
Neutropenia
platelets and coagulation.
Thrombocytopenia ■ Malignancy often causes anaemia.
Atypical lymphocyte morphology Invasion of the bone marrow by solid
Anisopoikilocytosis tumour is a cause of a
Macrocytosis1 leucoerythroblastic blood picture.
Bone marrow Variable changes in cellularity ■ Bacterial
and viral infections are
Dysplasia
common causes of abnormal blood
Increased plasma cells
counts.
Increased fibrosis
Haemophagocytosis ■ Infectiousmononucleosis is a disease
Opportunistic infection (e.g. caused by the Epstein–Barr virus.
granulomas) Numerous atypical lymphocytes are
Lymphoid aggregates seen in the blood.
Lymphoma ■ The many possible blood changes of
Other Positive direct antiglobulin test (DAT) HIV infection result from a combination
Lupus anticoagulant of a direct viral effect, opportunistic
Fig 48.3  Atypical lymphocyte in infectious infection and drugs used in treatment.
mononucleosis. 1
Particularly in patients receiving the drug zidovudine.

tahir99-VRG & vip.persianss.ir


98 9 SPECIAL SITUATIONS

49 The developing world


The term ‘developing world’ is used to describe the majority
of tropical countries which are ‘hot, humid and poor’. An
alternative term is the ‘less economically sound’ nations, as
these countries are often advanced in human and cultural
resources. Haematological practice is different to that in most
developed countries. Genetic diseases such as the haemoglob- Male and female
inopathies and red cell enzymopathies are frequent in many gametocytes Exflagellating
male gametocyte
tropical regions. Deficiency anaemia and haemolytic anaemia
are often secondary to infections such as ancylostomiasis Female gametocyte
being fertilised by
(hookworm) and malaria. Medical treatment regarded as male gamete
routine in the developed countries is commonly unavailable.
Zygote
For instance, only about 20% of the world’s haemophiliac
Maturation
population has access to factor VIII replacement therapy. Merozoites of female Stomach
With the ever-increasing availability of ‘exotic’ holidays and Immature gamete wall
gametocytes
regular foreign travel within immigrant populations, doctors
Ookinete
in the developed world are seeing more tropical diseases. In Schizont Young
the patient with unexplained symptoms such as malaise and oocyst
fever, or signs such as splenomegaly, a history of travel should
not be overlooked.
Segmenting
Ring form oocyst

Malaria Trophozoite Merozoites


Malaria is a protozoal disease, the infectious agent being Plas- Ruptured
oocyst
modium falciparum, P. vivax, P. ovale or P. malariae. Mortality
from the disease has fallen over the last decade but it remains Sporozoites
a serious health risk throughout the tropics and subtropics invading
salivary glands
where insecticide resistance of anopheline mosquitoes and
multiple drug resistance of malarial parasites make control
and treatment challenging. According to the World Health
Organization, there were 216 million cases of malaria and an
estimated 655 000 deaths in 2010. Most deaths occur in chil- Hypnozoite
dren living in Africa where the disease accounts for 20% of
Fig 49.1  Life cycle of the malarial parasite.
all childhood mortality.
some very ill patients with malaria initially have no detectable
Pathogenesis parasites in the blood as there is sequestration of parasite-
The life cycle of the malaria parasite is illustrated in Figure laden red cells in the tissues. It is beyond the scope of this
49.1. When taking a meal of blood an infected mosquito initi- book to make a detailed comparison of the four species but
ates human infection by the inoculation of malarial sporo- some typical appearances are shown in Figure 49.2. Supple-
zoites. These rapidly pass to the liver where they enter mentary methods of parasite detection include antigen and
hepatocytes and divide. After several days, enormously antibody detection and DNA-based techniques.
increased numbers of parasites (merozoites) depart the liver
and invade red cells. Here the merozoites develop via ring Clinical features
forms and trophozoites into schizonts. Rupture of the sch- Malaria has a different clinical presentation in non-immune
izont releases 12–20 merozoites back into the blood, thus and immune patients.
perpetuating the cycle. The duration of the blood cycle varies
between malarial species, explaining the different periodicity Non-immune patient
of fever in each type. A further mosquito becomes infected The interval between the mosquito bite and the onset of
when it feeds on blood containing gametocytes, the sexual symptoms is typically 1–2 weeks. Common symptoms are
form of the parasite. rigors, sweats, headache, vomiting, diarrhoea and muscle
pains. P. vivax and P. ovale are classically associated with
Diagnosis bouts of fever on alternate days and P. malariae on every third
Although malarial parasites may be detected in normal blood day. Possible clinical signs include a rising temperature, tachy-
films, their identification is generally easier in Leishmann or cardia, herpes labialis, jaundice, dehydration and splenomeg-
Giemsa stain at a higher pH. A thick film is best for detection aly. P. falciparum infection is the most dangerous form of
and a thin film for determination of the species. Prolonged malaria. The onset can be insidious and the fever has no
inspection of the film is sometimes necessary to spot malarial particular pattern. Life-threatening complications such as cer-
parasites as there can be a low level of parasitaemia. Where ebral malaria (with development of coma), acute renal failure
malaria is suspected on clinical grounds repeated samples and blackwater fever (rapid intravascular haemolysis), can
may be needed to make or exclude the diagnosis. P. falciparum suddenly develop in a patient previously not particularly ill.
is often associated with higher parasite counts. Paradoxically, Children are particularly at risk of a sudden demise.

tahir99-VRG & vip.persianss.ir


The developing world 99

(a) (b) (c)


Fig 49.2  Malarial parasites in the blood. (a) Ring-forms in P. falciparum malaria. (b) Schizonts in P. ovale malaria. (c) Gametocytes in P. vivax malaria.

Endemic malaria doxycycline. Where there is doubt, ■ Trypanosomiasis. The parasites are
In indigenous populations, malaria expert advice should be sought. Simple extracellular and motile.
presents variably depending on the preventative measures such as protec-
degree of endemicity, the age of the tive clothes, mosquito nets and insect
Iron deficiency in
patient and the development of immu- repellent creams also help reduce the
hookworm infection
nity. Thus in hyperendemic areas where risk of infection. The recently discovered
there are seasonal variations, adults red cell surface receptor which allows P. Hookworms infect approximately a
develop considerable immunity, malaria falciparum to invade may provide a billion people worldwide. They are a
causing only short episodes of fever and target for a future vaccine. major cause of gastrointestinal blood
a palpable spleen. In holoendemic areas loss and iron deficiency anaemia in trop-
there is infection through the year and ical regions. Worms attach to the upper
Visceral leishmaniasis small intestine and remove blood from
usually the disease manifests as a tran-
(kala-azar) the host; the daily loss can be as great
sient low parasitaemia with no symp-
toms. In hypoendemic areas, epidemics This protozoal disease is transmitted by as 250 mL. Management of anaemic
occur and the disease resembles that in sandflies and caused by the organism patients should include both treatment
the non-immune. Tropical splenomegaly Leishmania donovani. It is a cause of of worms with an effective anti-
syndrome is the development of massive massive splenomegaly. The organism helminthic agent and oral iron supple-
splenomegaly in adults in hyperen- may be detected in a blood film within ments to replenish stores.
demic areas. The patient has a low para- monocytes or neutrophils but bone
sitaemia with an exaggerated immune marrow aspiration is more sensitive.
Endemic Burkitt’s
response and very high levels of IgM.
lymphoma
Other parasitic diseases
Treatment and prophylaxis detectable in the blood
Endemic Burkitt’s lymphoma is an
Ill patients should be rested and rehy- aggressive B-lymphoblastic lymphoma
drated. A rational choice of drug treat- These include the following: which is found particularly in African
ment requires knowledge of both the children. In areas where malaria is
■ Filariasis. Microfilariae are released
clinical syndrome and the likelihood of holoendemic it is the most common
into the blood during an acute attack
drug resistance. The mainstay of treat- childhood cancer. The disease is associ-
of disease. As the organisms are
ment of severe malaria is quinine. This is ated with Epstein–Barr virus (EBV)
motile, examination of a wet
given intravenously. The dosage must be infection and the chromosomal rear-
preparation is useful.
carefully calculated to avoid under- rangement t(8;14). The classic clinical
■ Babesiosis. This tick-borne disease
treatment or toxicity. Recent trials have presentation is with a massive tumour
only occasionally affects humans.
suggested that artesunate, a derivative of of the jaw or other extranodal disease.
Trophozoites which resemble small
the plant compound artemisin, is both Cure rates exceeding 90% are possible
ring-forms of P. falciparum can be
easier to give and more effective than with combination chemotherapy.
found in red cells.
quinine in severe malaria. Its use in
Africa may be limited by its cost. Special-
ist advice should be sought in difficult
The developing world
cases. ■ The incidence of many haematological disorders and the availability of treatment is
Chemoprophylaxis is advised for non- different in the developing world and developed countries.
immune travellers entering malarial ■ Malariais a protozoal disease transmitted to humans by anopheline mosquitoes. It is a
areas. Specific recommendations depend major health problem in tropical and subtropical regions.
on the risk of exposure to malaria, the ■ Laboratory diagnosis of malaria depends on the identification of parasites in thick and thin
extent of drug resistance, the efficacy of blood films.
drugs, drug side-effects and patient- ■ Optimum drug treatment of established malaria and the best choice of prophylaxis require
related criteria (e.g. pregnancy, renal expert knowledge of clinical syndromes and possible drug resistance.
impairment). Drugs used include chlo- ■ Hookworm infection is a major cause of iron deficiency in tropical areas.
roquine, proguanil, mefloquine and

tahir99-VRG & vip.persianss.ir


100 10 RECENT ADVANCES IN HAEMATOLOGY

50 Molecular biology
Molecular techniques now play a central role in the diagnosis
and management of blood disorders, particularly haemato-
logical malignancies. This is a rapidly changing field and the
following is a summary of some of the most commonly used
and newest technologies and applications.

Selected techniques used in the analysis


of DNA
Polymerase chain reaction (PCR)
The object of PCR is to amplify a preselected sequence of
DNA many times over. This amplification greatly facilitates
subsequent analysis of the DNA sequence for point mutations
and polymorphisms, and often allows direct analysis of the
product by gel electrophoresis without the use of probes. Fig 50.1  Use of FISH in a case of chronic myeloid leukaemia (CML).
The method can only be briefly described here. Essentially Probes for BCR and ABL are used – juxtaposition of the two probes (giving a
yellow signal as in cell lower right) indicates the BCR–ABL rearrangement
two specific oligonucleotide primers are added to the DNA.
seen in Ph+ CML.
These have sequences matching the regions flanking the
region of interest. A DNA polymerase is added and the
mixture heated, causing the DNA to dissociate into two single Germinal-centre Activated
Type 3
strands. Following cooling the single strands bind to the oli- B-cell – like B-cell – like
gonucleotides which are in excess. The oligonucleotide then
acts as a primer for DNA polymerase and is extended to form
a new double-stranded molecule. With each repeat of the
cycle the amount of DNA is doubled. Generally about 30
cycles are used and amplification of approximately 106 can be
achieved.
Genes

Fluorescence in situ hybridisation (FISH)


FISH describes the hybridisation of specific DNA or RNA
sequences in situ to cellular targets attached to microscope
slides. The most popular probes are chromosome-specific
DNA sequences which generate a brilliant signal in both
metaphase and interphase nuclei. The technique is particu-
larly useful in the demonstration of chromosomal mono-
somies or trisomies but chromosome translocations (Fig
50.1), deletions and amplification of specific genes can also be
1.0
detected. The results of FISH may be further improved by
image processing.
Probability

Comparative genomic hybridisation


This technique is designed to detect regions in the genome 0.5 Germinal-centre B-cell like
which are undergoing quantitative changes. Different tumours
Type 3
show distinct genomic hybridisation patterns of gains and Activated B-cell like
losses. The method is especially useful in the analysis of
leukaemias. 0.0
0 2 4 6 8 10
Microarrays/gene profiling Overall survival (yr)
Microarray technology allows the simultaneous profiling of Fig 50.2  Subgroups of diffuse large B-cell non-Hodgkin’s lymphoma
tens of thousands of genes thus painting a molecular portrait (DLBCL) according to gene expression profiles. In the top panel each
of a tumour cell. Several methods are available for analysis of column represents a single DLBCL and each row a single gene. Red areas
a large number of RNA transcripts. These include comple- indicate increased expression and green areas reduced expression. Survival
after chemotherapy is different in the three groups. (Reprinted with
mentary DNA microarrays, oligonucleotide microarrays and
permission from Margalit O et al 2005 Micro-array based gene expression
serial analysis of gene expression (SAGE). The most com- profiling of hematologic malignancies: basic concepts and clinical
monly used ‘platforms’ are the two microarray technologies applications. Blood Reviews 19: 226.)
in which each experiment reveals the expression levels of over
20 000 genes. cDNA fragments and oligonucleotides can be complex gene expression data generated requires powerful
spotted onto glass slides. The DNA arrayed on the slide is statistical analysis. Microarrays are enhancing our under-
generally referred to as the ‘probe’ and the cDNA or cRNA standing of haematological malignancy (see below and Fig
derived from the sample is referred to as the ‘target’. The 50.2).

tahir99-VRG & vip.persianss.ir


Molecular biology 101

Haematological malignancy
Diagnosis and classification
Leukaemias and lymphomas were origi-
nally diagnosed and classified on the
basis of their morphological appear-
ance. As is discussed in the disease sec-
tions, optimum management of these
disorders now requires the supplemen-
tation of traditional clinical and mor-
phological information with detail of
immunophenotypic, karyotypic and
molecular characteristics. Molecular
analysis allows the confirmation of spe-
Fig 50.3  Whole genome sequencing for detection of mutations in chronic lymphocytic cific disease markers (e.g. BCR-ABL in
leukaemia. For each tumour genome, copy number (solid lines), density of mutations per 5-Mb
window (bars) and protein-coding mutations (dots) are shown. The shaded rectangle indicates the chronic myeloid leukaemia) and also
location of the 13q14 deletion present in three of the four cases. Chromosome numbers are listed reveals key prognostic information
below the four profiles. (Reprinted by permission from Macmillan Publishers Ltd: Puente XS et al 2011, (e.g. Ig gene mutation in chronic lym-
Whole genome sequencing identifies recurrent mutations in chronic lymphocytic leukaemia. Nature phocytic leukaemia). Microarray-based
475: 101–105.) gene expression and next-generation
sequencing studies, described above, provide novel insights
Relative frequency
of leukaemic cells into the biology of leukaemia, lymphoma and myeloma. Sim-
1 plification of this expensive research technology is likely to
10 -1 permit its eventual use in the hospital laboratory.
Detection limit
10-2 morphological Minimal residual disease
techniques Traditional definitions of remission in leukaemia have relied
10-3
on crude morphological criteria. Many patients in remission
10-4 subsequently relapse, implying the existence of occult neo-
10-5 plastic cells undetectable by normal morphological or cytoge-
Detection limit of netic methods – so-called minimal residual disease (MRD) (Fig
10-6 immunological
50.4). Reliable detection of MRD potentially allows improved
marker analysis and
10-7 PCR techniques management with escalation of therapy for patients with per-
'Cure' sistent disease and the avoidance of excessive treatment in
0
0 1 2 3 4 patients showing a good response to previous intervention.
I-Rx M-Rx Follow-up in years Detection of MRD relies upon the presence of disease markers
I-Rx = induction chemotherapy; M-Rx = maintenance chemotherapy that can be targeted (e.g. PML-RAR α in acute promyelocytic
Fig 50.4  The detection of minimal residual disease. The greater leukaemia). In childhood and adolescent ALL the tandem
sensitivity of PCR and immunological marker analysis compared with application of flow cytometry and PCR can be used to study
traditional morphological techniques in the detection of residual leukaemic MRD in almost all patients and this information is being
cells can be seen. (Reproduced with permission of JJM van Dongen, employed in clinical trials. In CML quantitative PCR assay of
Department of Immunology, Erasmus University, Rotterdam, and Medicultura BCR-ABL transcripts is routinely used to direct management.
International B.V.)
Very low levels of BCR-ABL mRNA predict a good clinical
Next-generation sequencing outcome.
Massively parallel sequencing (also termed next- or second-
generation sequencing) results in the simultaneous genera-
Stem cell transplantation
Molecular techniques can be used both to monitor MRD
tion of millions of short DNA sequences. In studies of
post-transplant and to improve the level of HLA matching
haematological malignancy, it is important to sequence both
between unrelated donors and recipients.
the tumour cells and normal tissue (e.g. skin) from the patient
to identify relevant acquired (somatic) mutations. In whole
genomic sequencing (Fig 50.3), the object is to sequence the
entire genome. More specific next-generation techniques
Molecular biology
include exome and transcriptome sequencing.
■ Molecular biology techniques routinely used in haematology
include the polymerase chain reaction (PCR), fluorescence in situ
Application of molecular biology hybridisation (FISH) and comparative genomic hybridisation.
in haematology ■ Molecular techniques play a key role in carrier detection and
Carrier detection and antenatal detection in antenatal detection in genetic disorders such as thalassaemia and
haemophilia.
genetic disorders
Molecular techniques are crucial in antenatal diagnosis and ■ In haematological malignancy, molecular techniques refine
diagnosis and classification, and improve detection of minimal
genetic counselling in genetic disorders of the blood. For
residual disease (MRD) after therapy.
instance, PCR technology to detect DNA point mutations or
■ Recent developments include microarray-based gene expression
deletions in chorionic villus samples, enabling first trimester
profiling and whole genome sequencing of haematological
testing for thalassaemia. Extraction of fetal cells or DNA from malignancies.
maternal blood may allow less invasive prenatal diagnosis.

tahir99-VRG & vip.persianss.ir


102 10 RECENT ADVANCES IN HAEMATOLOGY

51 Potential advances in treatment


This section addresses some potential include immunomodulating drugs such molecular weight heparin is a more
advances in the treatment of blood dis- as lenalidomide. Such considerations recent addition. All of these drugs have
orders only briefly alluded to in the may also apply in other haematological disadvantages, none fulfilling all the cri-
coverage of specific diseases. Inevitably malignancies. For instance, encouraging teria for an ‘ideal’ anticoagulant, namely
any such listing of advances is subjec- clinical results have been obtained in a high efficacy to safety index, a predict-
tive. Most clinicians would expect an chronic lymphocytic leukaemia follow- able dose response with no need for
expanding role for the targeting of the ing the targeting of the tumour cells laboratory monitoring, administration
microenvironment in haematological with genetically modified autologous by parenteral and oral routes, a rapid
malignancy, the wider use of new oral T-cells. onset of action, a safe antidote, freedom
anticoagulant agents, and the introduc- from non-anticoagulant side-effects and
tion of gene therapy for single-gene minimal interaction with other drugs.
New oral
disorders such as haemophilia and Any new agent must aspire to meet as
anticoagulant drugs
thalassaemia. many of these requirements as possible
Anticoagulant drugs are used both in the without being prohibitively expensive.
treatment and prevention of throm- Attempts to develop better anticoagu-
Targeting the
boembolic disease (see pp. 80–81). Con- lants have focused on specific steps or
microenvironment
ventional heparin and warfarin were enzymes in the coagulation pathway.
in haematological
discovered over 60 years ago while low Agents under evaluation include inhibi-
malignancy
tors of the factor VIIIc/tissue factor
The tumour microenvironment is cur- Tumour cell pathway, factor Xa inhibitors, activated
rently thought to play a crucial role in protein C and soluble thrombomodulin
cancer biology. A better understanding and direct thrombin inhibitors (Fig 51.2).
of the complex interactions between Direct thrombin inhibitors and direct
tumour and non-malignant cells is likely factor Xa inhibitors are now entering
to translate into better characterization clinical practice and their use is likely to
of tumour pathophysiology and novel change the current practice of antico-
approaches to therapy. In haematologi- agulation. Dabigatran etexilate is a
cal malignancy, follicular lymphoma prodrug. It is rapidly absorbed and
(see also p. 61) provides an excellent converted in the liver to the active
model for the study of the microenvi- form dabigatran which acts as a direct
ronment. This disease accounts for thrombin inhibitor binding both free
approximately a quarter of all cases Non-malignant cells and clot-bound thrombin. The drug is
of non-Hodgkin’s lymphoma. Modern (e.g. T-cells, macrophages) administered orally and no therapeutic
treatment with a combination of chemo- Fig 51.1  A schematic view of the monitoring (i.e. laboratory testing of
therapy agents and the anti-CD20 microenvironment in follicular lymphoma. drug levels or activity) is required.
monoclonal antibody rituximab has Cytotoxic CD8+ lymphocytes exert anti-tumour Rivaroxaban is a direct inhibitor acti-
effects (red arrows) while tumour cells seek
improved overall survival but cure is vated X (factor Xa) and is also given
self-preservation (green arrows) by stimulating
elusive. Better therapeutic strategies are CD4+ T-cells to secrete favourable cytokines, orally without the need for routine
needed. Technologies such as immuno- inhibiting cytotoxic T-cells, and erecting a monitoring. Apixaban and edoxaban
cytochemistry, functional cytotoxicity ‘tumour barrier’. are other oral factor Xa inhibitors under
assays and 3-D confocal imaging have
improved our understanding of the role
of the microenvironment in this disease and offered the pros- Coagulation pathway Possible anticoagulant
pect of new treatments which tip the balance against the Tissue factor pathway
Initiation Tissue factor / VIIa
tumour cells. The various interactions are complex (Fig 51.1) inhibitors
but it is clear that CD8+ cytotoxic lymphocytes play an active
role and have the potential to be employed in the treatment IX
of follicular lymphoma. Most simplistically, a higher content X
of T-cells appears to confer a better prognosis while pro- Thrombin Protein C pathway
generation IXa, VIIIa (e.g. activated protein C)
tumour macrophages may worsen the outlook. Follicular lym-
phoma cells can manipulate the microenvironment to their
own advantage. Stimulation of CD4+ T-cell cytokine release Xa, Va Xa inhibitors
promotes tumour cell growth and survival. The tumour cells II (e.g. rivaroxaban)
also employ mechanisms to block entry of cytotoxic T-cells
into the tumour site and to inhibit recruitment and trafficking Thrombin Thrombin inhibitors
of previously healthy T-cells. The challenge is to find ways of Thrombin (e.g. dabigatran)
activity
supplementing current immunotherapy regimens (e.g. rituxi-
mab and chemotherapy) by tipping the balance in the tumour Fibrin
microenvironment towards enhanced T-cell anti-tumour
activity and away from immunosuppression. Possible agents Fig 51.2  Possible new anticoagulants. See text for discussion.

tahir99-VRG & vip.persianss.ir


Potential advances in treatment 103

development. The indications for all candidates for cure by gene therapy. 1
these agents are gradually increasing as Haemophilia is a particularly attractive Establish the
genetic aetiology of the disease
they are tested in clinical trials. There target disease as only a very small sus-
are still questions regarding long-term tained increase in factor VIII or IX levels
safety. Also, there are no specific anti- (1–2%) significantly reduces the bleed-
dotes to reverse the anticoagulant effects. ing tendency. A variety of methods for Haemophilia A is a sex-linked inherited
Fortunately, the drugs have a short half- transferring genes for factor VIII and IX disorder. The gene is on the
life and the bleeding risk is relatively are under investigation. No single tech- X chromosome
low. nique has emerged as being definitely
superior. Animal studies with viral- 2
Factor VIII gene
derived vectors suggest that it is possible
Gene therapy
to provide sustained therapeutic levels
Gene therapy is potentially a very pow- of the clotting factors. Allowing the pos- Deletion
erful therapeutic tool applicable to a sibility that gene therapy for haemo-
Haemophilia A is caused by a mutation of
wide range of diseases including several philia may involve unforeseen risks, a the factor VIII gene. The normal factor
blood disorders. After years of failure, number of carefully designed clinical VIII gene has been identified
and cloned
there have been recent successes in the studies in consenting patients are under
treatment of infants with immunodefi- way. In an early trial, a small number
3
ciency and in selected patients with of patients with severe haemophilia B Normal
malignancy. The basis of the technique were treated using a non-pathogenic, factor VIII gene-vector
is the insertion of a new functional gene non-integrating adeno-associated virus Patient cells
into a cell (Fig 51.3). The gene is intro- (AAV) as a DNA carrier. An intravenous (e.g. hepatocytes)
duced into the target cell by use of a schedule with gradually increased doses
Design a suitable vector for the
‘vector’. Disabled viruses are commonly was used. Patients achieved increased insertion of the normal factor VIII
used as vectors because they can under- factor IX levels with sustained clinical gene into patient cells
take tasks necessary for successful gene benefit, some being able to stop prophy-
transfer such as binding to the target cell lactic factor IX treatment. In thalassae- 4
and delivering the viral genome to the mia initial efforts at gene therapy have Patient cells with
nucleus for transcription. Non-viral been directed against diseases of the normal factor VIII gene inserted
vectors based on plasmid DNA pro- β-globin gene. The therapeutic strategy Factor
duced in bacteria and combined with involves insertion of a normally func- VIII
lipids are also used. The optimum vector tioning γ-globin or β-globin gene into
Obtain adequate levels of factor VIII
and delivery method is likely to vary the patient’s haematopoietic stem cells. synthesis over a prolonged period
depending on the disease under treat- At the time of writing, one patient with to 'cure' the disease
ment. Early protocols mainly involved β-thalassaemia has been rendered trans-
an ex vivo approach where the gene was fusion independent following gene Fig 51.3  The essential steps necessary for
successful gene therapy in haemophilia A.
inserted into cells taken from the patient. therapy but it is unclear if this was for-
However, more recently the vector is tuitous or if this success can be repro-
normally given directly to the patient (in duced in larger trials.
vivo approach). The problems have been Gene therapy is also likely to have a malignant cell and cure the patient with
numerous and have included difficulties role in malignant disorders. Leukaemia minimal side-effects. Alternatively, gene
in characterising and accessing the is essentially a genetic disease and, in therapy might be used to augment the
target cells, poor efficacy of gene trans- theory, gene therapy could eventually be patient’s own immune response against
fer, short-lived expression of the newly used to correct the abnormality in the malignant cells.
introduced gene, and safety issues. The
latter especially relate to viral vectors
which have caused clinical symptoms of Potential advances in treatment
infection and induced massive immuno-
■ The tumour microenvironment plays a crucial role in some haematological malignancies
logical responses. Two children cured of (e.g. follicular lymphoma). It is likely to be increasingly targeted.
their immunodeficiency by gene therapy
■ New anticoagulant agents are under active development. Oral thrombin and factor Xa
subsequently developed leukaemia as a inhibitors potentially give safe anticoagulation without laboratory monitoring.
result of insertional mutagenesis.
■ Theenormous promise of gene therapy is likely to be soon translated into clinical benefits.
As single-gene disorders, both haemo- There is a real prospect of successful treatment of haemophilia.
philia and thalassaemia are good

tahir99-VRG & vip.persianss.ir


104 11 PRACTICAL PROCEDURES

52 Venepuncture and venous access

Obtaining a sample of venous blood from a patient is the vacuum is released. It is important to understand how the
most commonly performed practical procedure in haematol- system works before undertaking venepuncture.
ogy. The technique is apparently straightforward but poorly
performed venepuncture can both upset the patient and com- Precautions
promise the quality of the sample. Gaining venous access for Blood should not be taken from a vein proximal to an intra-
the delivery of fluids, blood or drugs is also fundamental to venous infusion as the sample can be diluted. Neither should
good haematological practice. This section is an overview of eczematous or infected areas be used for venepuncture. If
venepuncture and venous cannulation. These skills are best patients are known to have a blood-transmissible infection
learnt by practice with expert supervision. (e.g. hepatitis B or C, HIV) or are at increased risk of such an
infection this must be indicated on the specimen bottle and
request form. Due care must be taken as this is sensitive
Taking a venous blood
information – labels stating an infective risk are generally
specimen (venepuncture)
available. In view of the possibility of needle-stick injuries,
The patient should be the correct patient – check their iden- those performing venepuncture should be vaccinated against
tity! Most serious haemolytic transfusion reactions arise from hepatitis B.
careless identification of patients and incorrect form labelling.
Patients should sit or lie comfortably in such a way that no Common problems
serious injury could result from a faint. The operator washes Venepuncture is not always easy. If blood is not aspirable fol-
his hands and wears plastic gloves – insist on gloves that fit lowing perceived entry of the vein it is worth withdrawing the
properly. The procedure is explained to the patient and the needle slowly with suction applied as the vein may have been
necessary consent obtained. The presence of a little transient transfixed. If a vein cannot be located in the antecubital fossa
pain when the needle is inserted should be acknowledged but it is permissible to use veins at the wrist or on the dorsum of
not exaggerated. the hand. If two attempts fail a more experienced colleague
Under normal circumstances blood is most easily taken
from a vein in the antecubital fossa; the median cubital vein
Left arm
is preferred (Figs 52.1 and 52.2). It is considerate to ask anterior view
whether the patient is left- or right-handed and then to choose
the non-dominant arm. A tourniquet is applied well proximal
to the site. This should cause distension of the veins but not
discomfort. Gentle palpation is the best method of identifying Cephalic
a vein and checking its patency. If a suitable vein proves vein
elusive it may help to gently tap the area or to warm the arm Median
cubital vein
in water. The skin over the chosen vein is thoroughly cleaned
with antiseptic solution. Usually a 21- or 22-gauge needle is
used but a smaller size (e.g. 23) can be used where the veins
are fragile, or in children. The syringe should be adequate for
the sample – where larger blood samples necessitate more
than one syringe a ‘butterfly needle’ may be preferred to a
conventional venepuncture needle. The needle is inserted
bevel uppermost along the line of the vein at an angle of
around 20°. There is a distinctive ‘give’ as the vein is entered.
Blood is aspirated into the syringe slowly to avoid haemolysis.
The tourniquet is released and the needle withdrawn after a Fig 52.1  Veins at the antecubital fossa. The median cubital vein is
dry swab has been held to the site. Pressure should be applied preferred for routine venepuncture.
by the patient or an assistant with the arm held straight or
slightly elevated. The needle is removed from the syringe –
not resheathed – and placed directly into a sharps container.
The specimen is expelled gently from the syringe into the
relevant bottles. Mixing with anticoagulant is best achieved by
gently inverting the bottle several times – violent shaking will
damage the sample. An adhesive plaster can be applied to the
venepuncture site (check for allergy) when bleeding has
stopped.
The above describes the procedure for a conventional
needle and syringe. Increasingly, venepuncture is performed
using closed evacuated container systems where a double-
ended venepuncture needle is screwed into a holder and the
evacuated tube inserted into the holder following entry of the
vein. Blood is automatically aspirated into the tube as the Fig 52.2  Taking blood from a vein in the antecubital fossa.
Venepuncture and venous access 105

should be sought. As a last resort a sample can be taken from


the femoral vein. The operator must be familiar with the
anatomy of the femoral region as the vein lies close to the
femoral artery and nerve.

Children
In babies and infants a blood sample is often more easily
obtained from a stab wound made with a lancet (capillary
blood). The usual site is the heel, although fingers and ear-
lobes can be used. Venepuncture may also be from scalp
veins.
Fig 52.3  A large gauge plastic cannula.
protected with a sterile dressing and the
Venous access
cannula secured with a bandage or
Peripheral venous cannulation adhesive tape.
Almost all haematology patients admit- The most common problem is failure
ted to hospital require a drip to infuse to locate a vein in the favoured sites. A
fluids, blood products or drugs. Before more experienced operator may be suc-
inserting a cannula into a vein, an appro- cessful. Where problems persist in expe-
priate giving set should be prepared in rienced hands, other veins such as those
accordance with instructions and the in the region of the ankle or the subcla-
bag or bottle containing the infusion vian, jugular or saphenous veins may be
fluid inverted and hung on the drip cannulated. Regular inspection of the
stand. The set should be properly drip site and careful hygiene will mini-
primed and all bubbles excluded. The mise the chance of infection. Where
operator must wash hands and wear there is local inflammation or an other-
gloves. It is vital to ensure that the wise unexplained bacteraemia, the
patient is comfortable and fully under- cannula should be removed and another
stands the procedure and that necessary site used. Fig 52.4  Chest X-ray showing a central
consent is obtained. The choice of venous catheter in situ.
cannula depends both on the quality Central venous cannulation
of the veins and the duration and type Insertion of wide-lumen silicon rubber site on the anterior chest wall. A catheter
of infusion. For short-term infusions or catheters (generally referred to as cuff within the tunnel promotes the for-
small veins a winged metal cannula Hickman catheters) is routinely under- mation of fibrous tissue which helps
(butterfly needle) is often suitable. In taken in clinical haematology where secure the device. The procedure is
other circumstances a larger gauge recurrent intravenous access is required. usually performed in the operating
plastic cannula is used (Fig 52.3). In Examples include: theatre by a surgeon or anaesthetist.
adults, 18–20-gauge catheters provide Once in place the catheter may be used
■ patients with haematological
good flow rates without too much dis- for several months. Strict aseptic tech-
malignancy receiving intensive
comfort for the patient. nique is necessary as infection with
chemotherapy
The best site is the non-dominant coagulase-negative staphylococci is the
■ patients with thalassaemia having
forearm or the dorsum of the hand. The most common complication.
regular blood transfusions
antecubital fossa is best avoided as it is
■ children with haemophilia A on
uncomfortable to have the elbow immo-
prophylactic factor VIII treatment.
bilised. A tourniquet is applied and the
Intravenous administration of
skin cleaned as for venepuncture. The The catheter is normally inserted into cytotoxic drugs (chemotherapy)
skin at the site may be stretched slightly the subclavian vein and the location of should only be undertaken
to immobilise the vein. The cannula the distal tip checked on X-ray (Fig 52.4). following appropriate training
assembly (metal needle and surround- The proximal end of the catheter can be in the use of these agents.
ing plastic cannula) is introduced tunnelled under the skin with an exit
through the skin and into the vein. Once
blood enters the cannula chamber or is
easily drawn into a syringe, the tourni- Venepuncture and
quet is released and the metal needle venous access
withdrawn from the plastic cannula ■ Obtaininga venous blood sample (venepuncture) is a commonly performed practical
which may be advanced further into the procedure in haematology; poor technique can upset the patient and ruin the sample.
vein. The pre-prepared giving set is ■ In babies and infants, capillary blood sampling is often easier than venepuncture.
attached to the cannula and fluid
■ Peripheral venous cannulation is commonly performed to infuse fluids, blood products and
allowed to enter the vein while the inser- drugs.
tion site is carefully inspected for
■ Where there is serious difficulty in locating a vein for venepuncture or cannulation, more
possible extravasation. The needle is experienced help should be sought.
promptly disposed of in a sharps recep-
■ For recurrent venous access, insertion of an indwelling central venous catheter can be
tacle. To minimise the chance of the drip
helpful.
being infected or dislodged the site is
106 11 PRACTICAL PROCEDURES

53 Bone marrow aspiration and trephine biopsy

The indications for performing bone marrow aspiration and


trephine biopsy procedures have previously been discussed
(p. 19). In this section the practical aspects of obtaining these
samples are outlined. More detailed accounts can be found in
books of practical procedures, but ultimately the only way to
perfect techniques is to practise under expert supervision.
Although the anterior iliac crest is occasionally preferred,
most operators get the best specimens from the posterior iliac
crest. The sternum is now rarely used. This is, in part, due to
the small risk of causing catastrophic damage to the mediasti-
num, but mainly because it is not possible to obtain a trephine
biopsy. Only the posterior iliac crest approach is described
here.

Bone marrow aspiration


As for all procedures the sequence of events should be
explained to the patient, reassurance given and consent
obtained. A degree of discomfort should be acknowledged but
it should be emphasised that this is transitory. In most adults,
local analgesia is adequate but sedation is considered where Fig 53.1  Anatomy of the posterior iliac crest. Possible sites for marrow
sampling are indicated in red.
patients are unusually anxious. A general anaesthetic is the
norm in children. A clean, no touch
technique is mandatory and operators
should wear gloves. Stringent asepsis is
needed in immunosuppressed cases.
The patient lies in the left or right
lateral position and the skin over the
posterior iliac crest is cleaned with anti-
septic prior to screening with sterile
drapes. The crucial next stage is to prop-
erly identify the bony landmarks (Fig
53.1). This is straightforward in most Fig 53.2  Bone marrow aspirate needle.
patients but can be problematic in obese
subjects. If there are real difficulties in Marrow aspirate smears must be
locating the posterior iliac crest then the made promptly at the bedside before
anterior crest or the sternum may be the marrow clots. If a larger volume is
considered or the procedure may be needed for tests such as cytogenetics
performed under CT guidance. A local and immunophenotyping, it is best to
anaesthetic is infiltrated into the skin use a second syringe as large samples
and then down to the periosteum. dilute the marrow with peripheral blood
Before use it should be checked that the and reduce the quality of the morpho-
marrow aspirate needle stylet is easily logical preparations. If it proves difficult
withdrawn and the guard is removed or impossible to aspirate marrow it is
(this is only required for sternal aspi- worth replacing the stylet and carefully Fig 53.3  Aspiration of bone marrow from
rates). The needle (Fig 53.2) is inserted advancing or retracting the needle a the posterior iliac crest.
through the skin and subcutaneous short distance before repeating aspira-
tissues at the site of local anaesthetic tion. It is important to remember that a applied to the puncture site. Outpatients
infiltration until the periosteum is ‘dry tap’ can result from marrow pathol- should probably be observed for at least
encountered. It is pushed through the ogy (particularly fibrosis or solid malig- an hour before being allowed home
periosteum with a deliberate screwing nancy) and is not always caused by poor (more if sedated). Troublesome haemor-
motion (alternating clockwise and anti- technique. rhage from the site is rare but it is sen-
clockwise) – a ‘give’ is felt as the marrow Once the aspirate needle is with- sible to correct a severe coagulation
cavity is entered. The stylet is withdrawn drawn, firm pressure is applied to the defect before undertaking the proce-
and a syringe attached to the needle (Fig site for a few minutes and then a sterile dure. Thrombocytopenia alone is gener-
53.3). Approximately 0.5 mL of marrow dressing or plaster used as protection. ally not a problem.
is aspirated into the syringe. The patient The patient lies on his back for 15 Patients often ask how quickly the
should be warned that this stage often minutes to ensure a period of recupera- ‘results’ will be available. Aspirate slides
causes pain but that it is momentary. tion and that further light pressure is can be processed for microscopy (see
Bone marrow aspiration and trephine biopsy 107

Table 53.1  Ancillary tests which may be


performed on bone marrow
aspirate samples
■ Cytochemistry
■ Cytogenetics
■ Immunophenotyping
■ Molecular studies
■ Microbiological culture
■ Cell culture studies
■ Drug resistance studies

Fig 53.4  Bone marrow trephine needle.

p. 19) within a few hours but most ancil- biopsy onto a glass slide before putting
lary tests (Table 53.1) take longer. it into histological fixative. This ‘touch
preparation’ is not useful for subtle mor-
phological diagnosis but can permit
Bone marrow
rapid identification of malignant infiltra-
trephine biopsy
tion. It usually takes several days to
In practice the trephine procedure is process the trephine biopsy. Aftercare is
usually performed immediately follow- the same as for the aspirate, although as
ing the aspirate at the same site. It is it is a slightly more invasive procedure
(a)
helpful to enlarge the aspiration punc- the patient also having a trephine may
ture site slightly with a scalpel blade. require a longer period of recuperation.
There is sometimes more prolonged dis- Nevertheless, trephine biopsies are rou-
comfort than in the aspirate procedure tinely performed in the outpatient clinic.
and sedation is indicated in anxious
adults, and a general anaesthetic is nec-
Bone marrow harvesting
essary in children. A number of different
disposable needles are available – the Bone marrow can be harvested from a
Jamshidi type is illustrated in Figure patient (for autologous stem cell trans-
53.4. Smaller needles are available for plantation) or from a donor (for alloge-
paediatric use. neic stem cell transplantation). The
It is important to ensure that the procedure is performed under a general
device is complete and that the stylet anaesthetic, the marrow being collected (b)
can be easily withdrawn. The trephine from the iliac crests using multiple Fig 53.5  Obtaining a trephine biopsy from
needle is inserted in a similar fashion to punctures with specialised harvest the posterior iliac crest. (a) Insertion of the
the aspirate needle through the perios- needles. Normally, approximately 1 litre needle. (b) CT guidance is useful in obese
teum and approximately 0.5 cm into is harvested from an adult in under patients.
the cortex (Fig 53.5a) – when properly an hour. Donors are hospitalised for
inserted the needle should easily support around 48 hours. Serious side-effects are as peripheral blood stem cells are more
its own weight. The stylet is removed rare but some short-lived discomfort commonly used than bone marrow in
prior to advancing the needle 2–3 cm over the aspiration sites is common. The both autologous and allogeneic trans-
using the same oscillatory movement. procedure is now undertaken less often plants (see page 56).
The needle is aimed towards the anterior
iliac crest. The method for breaking off
the biopsy varies with the needle used. Bone marrow aspiration and
Some have devices designed to grip the trephine biopsy
biopsy and ensure its retention. The
■ The optimal site for both bone marrow aspiration and trephine biopsy procedures is the
needle is then withdrawn taking care not posterior iliac crest.
to catch the skin and lose the biopsy in
■ Local
analgesia is often adequate but nervous adults require sedation and children
subcutaneous tissue. A special blunt normally require a general anaesthetic.
probe is provided to push the biopsy out
■ Marrow aspiration smears may be stained for microscopy immediately after the procedure
of the needle. The probe is inserted (with whereas trephine biopsies are processed over several days.
great care to avoid injury to the operator)
■ Seriousside-effects from posterior iliac crest aspiration and trephine biopsy are very rare.
at the sharp end of the needle so as not
Occasionally there can be excessive haemorrhage or local infection at the site.
to traumatise the sample.
■ Bone marrow can be harvested from the iliac crests in patients (for autologous stem cell
If the aspirate is a ‘dry tap’ it is
transplantation) or healthy donors (for allogeneic stem cell transplantation).
worthwhile gently dabbing the trephine
108 APPENDICES

Appendices
Appendix I: Reference ranges in normal adults
These figures are for guidance only. ‘Normal’ reference ranges vary in different
populations and in different laboratories. Patient results should always be
compared with local reference ranges.

Blood count
Haemoglobin Male 130–180 g/L Female 115–165 g/L
Packed cell volume Male 0.40–0.52 Female 0.37–0.47
Red cell count Male 4.5–5.9 × 1012/L Female 3.8–5.2 × 1012/L
MCV 80–96 fl
MCH 27–32 pg
MCHC 315–345 g/L
Reticulocytes 50–100 × 109/L (0.5–2.5%)
White cell count 4.0–12.0 × 109/L
Neutrophils 2.00–7.50 × 109/L
Lymphocytes 1.50–4.00 × 109/L
Monocytes 0.20–1.00 × 109/L
Eosinophils 0.02–0.40 × 109/L
Basophils 0.02–0.20 × 109/L
Platelet count 150–400 × 109/L

MCV, mean corpuscular volume; MCH, mean corpuscular haemoglobin; MCHC, mean corpuscular haemoglobin concentration.

Acute phase response


ESR1 Male 0–5 mm/h Female 0–7 mm/h
Plasma viscosity 1.50–1.72 mPa·s (25°C)
C-reactive protein 0–10 mg/L

1
Higher levels (up to 15 mm/h) may be seen in the elderly.
ESR, erythrocyte sedimentation rate.

Other
Ferritin Male 21–300 µg/L Female 15–150 µg/L
Serum iron 13–32 µmol/L
TIBC 45–70 µmol/L
Transferrin 1.2–2.0 g/L
Serum vitamin B12 160–760 ng/L
Serum folate 3–20 µg/L
Red cell folate 160–640 µg/L

TIBC, total iron binding capacity.


Appendices 109

Appendix II: Selected immunophenotypic


(cell surface) markers

Cluster differentiation (CD) Normal reactivity/Comments


designation
CD2 T-lymphocytes
CD3 T-lymphocytes
CD4 Helper/inducer T-lymphocytes
CD5 T-lymphocytes, B-lymphocyte subset (expressed in B-CLL)
CD7 T-lymphocytes
CD8 Cytotoxic/suppressor T-lymphocytes
CD10 Precursor B-lymphocytes (expressed in common ALL)
CD11c Monocytes, granulocytes, NK cells, activated T-lymphocytes, hairy cells
CD13 Monocytes, granulocytes (expressed in AML)
CD14 Monocytes
CD19 B-lymphocytes
CD20 B-lymphocytes (except pre-B)
CD22 B-lymphocytes
CD33 Monocytes, myeloid cells (expressed in AML)
CD34 Haematopoietic stem cells
CD36 Platelets, monocytes (platelet GP IIIa)
CD38 Plasma cells, some lymphocytes
CD41 Platelets (GP IIb)
CD42a/b Platelets (GP Ib)
CD45 Leucocytes
CD55 Broad (decay accelerating factor1)
CD56 NK cells
CD57 NK cells
CD59 Broad (membrane inhibitor of reactive lysis1)
CD61 Platelets (GP IIIa)
CD68 Macrophages, neutrophils
CD75 B-lymphocytes
CD79 B-lymphocytes
CD103 Hairy cells
CD114 Granulocytes, monocytes
CD117 (expressed in AML) Mast cells, plasma cells, expressed in AML

1
Deficient in paroxysmal nocturnal haemoglobinuria.
CLL, chronic lymphocytic leukaemia; ALL, acute lymphoblastic leukaemia; NK, natural killer; AML, acute myeloid leukaemia; GP, glycoprotein.

Appendix III: International prognostic index (IPI) for


non-Hodgkin’s lymphoma (NHL)
The IPI was initially designed for predicting the outcome of diffuse large B-cell
lymphoma (see non-Hodgkin’s lymphoma section for further discussion of IPI).
However, it has been shown to also have validity in other types of NHL. Patients
in the higher risk groups have poor outcomes with conventional chemotherapy
regimens.

Pretreatment criteria Score 0 Score 1


Age (years)1 60 or under Over 60
Stage (Ann Arbor) I or II III or IV
Number of extranodal sites of disease 1 or less Greater than 1
Performance status (ECOG/WHO scale2) 0 or 1 2 or greater
Serum lactate dehydrogenase (LDH) Low or normal High
These five scores are added to define the risk group as follows:
Low 0 or 1
Low intermediate 2
High intermediate 3
High 4 or 5

1
An age-adjusted model for patients less than 60 years is also available.
2
The ECOG/WHO performance status scale is defined as follows: (0) able to carry out all normal activities without restriction; (1) restricted in
physically strenuous activity but ambulatory and able to carry out light work; (2) ambulatory and capable of all self-care but unable to carry
out any work; up and about more than 50% of waking hours; (3) capable of only limited self-care; confined to bed or chair for more than 50%
of waking hours; (4) completely disabled; cannot undertake any self-care; totally confined to bed or chair; (5) dead.
110 CASE HISTORIES

Case histories
These case histories and the attached vital to exclude gastrointestinal 3. Simple blood tests to clarify the
questions are designed to illustrate bleeding despite the lack of cause of the anaemia should include
themes discussed in the main text. gastrointestinal symptoms. Clinical a reticulocyte count and inspection
They are in no particular order. For examination should include rectal of a blood film. Her reticulocyte
Case 1, there is an obvious clue in one examination. A colonoscopy count was raised at 9% and the
of the anaemia sections to help you get revealed a large bowel carcinoma blood film revealed large numbers
started. For the subsequent cases, you which was successfully resected. of spherocytes (see p. 28, Fig. 14.2).
will have to dig a little deeper into the A diagnosis of hereditary
book for the answers. Relevant page spherocytosis was made and was
numbers are shown is square brackets
Case 2 [pp. 28–29]
later also confirmed in the mother.
next to each case number. Where not A 15-year-old girl presents to her In the Haemolytic anaemia II
appended, the normal ranges for the primary care practitioner. According to section, you should be able to find
laboratory test results are listed in her parents, she is constantly pale and an acquired form of haemolytic
Appendix I. tired. They have also noticed occasions anaemia which is also a cause of
Good luck! when her eyes are a little yellow. She spherocytes in the blood film.
eats a normal diet and there is nothing
else of note in the remainder of the
Case 1 [pp. 24–25]
history. The mother has had life-long Case 3 [pp. 40–41]
A 53-year-old Caucasian man attends similar symptoms but these have been
A previously well 26-year-old man
his primary care practitioner only mild and never investigated. She
presents to the accident and
complaining of gradually increasing thinks other members of her family
emergency department of his local
fatigue and shortness of breath on might also be affected. The doctor
hospital with a 24 hour history of
exertion. He has also noted a few decides to check a full blood count as
spontaneous bruising and bleeding
kilograms weight loss but there are no he suspects anaemia:
from his gums. He has also felt
other symptoms. His doctor decides to
Haemoglobin 94 g/L unusually tired for the last few weeks.
check a full blood count, the results of
MCV 92 fl On examination he is pale with
which are summarised:
MCH 31 pg numerous small bruises and a
Haemoglobin 75 g/L White cell count 8.3 × 109/L scattered petechial rash. The accident
MCV 69 fl Platelet count 301 × 109/L and emergency doctor suspects
MCH 24 pg that this might be a blood disorder
1. What type of anaemia is this?
White cell count 9.2 × 109/L and he takes blood samples for a
2. The mother and other family
Platelet count 460 × 109/L full blood count and some tests of
members have similar symptoms
coagulation:
1. What type of anaemia is this? so this might be an inherited form
2. What is the most likely cause in of anaemia. Do you know of any Haemoglobin 84 g/L
this man? What simple blood causes of inherited anaemia? MCV 95 fl
investigation would confirm this? 3. What further simple blood tests can MCH 31 pg
3. If you confirm the anaemia be performed in the haematology White cell count 25.4 × 109/L
aetiology you suspect, are further laboratory to help elucidate the Platelet count 62 × 109/L
investigations necessary? cause of the anaemia in this girl? Prothrombin time 22 seconds (9–12)
Activated partial 58 seconds
Notes to Case 1 Notes to Case 2 thromboplastin (26–36)
1. This is a microcytic hypochromic 1. This is a normocytic normochromic time
anaemia. anaemia. Fibrinogen level 1.1 g/L (1.7–4.5)
2. The most likely cause in a 2. Inherited causes of anaemia include
The automated haematology analyser
Caucasian man of this age is iron the haemoglobinopathies (e.g.
does not generate a white cell
deficiency. Measurement of serum thalassaemias, sickle cell
differential count but the biomedical
ferritin is a simple test to confirm syndromes), disorders of the red
scientist in the haematology laboratory
the diagnosis. It is usually reliable. cell membrane (e.g. hereditary
has inspected a blood film and reports
A low level always indicates iron spherocytosis) and abnormalities of
that the white cell count is increased
deficiency but a normal level does red cell metabolism (e.g. glucose-6-
because of numerous leukaemic
not guarantee normal stores as phosphate dehydrogenase
blast cells with marked cytoplasmic
ferritin is increased in chronic deficiency). A thorough family
granularity. An urgent request for
inflammation and liver disease. The history is very important in the
specialist clinical haematology advice
result here was 7 µg/L. This low diagnosis of these disorders. In
is made.
level confirms the presence of iron them all, the anaemia is at least
deficiency. This may also be the in part due to haemolysis. The 1. What is the most likely
cause of the slight thrombocytosis. patient’s history of intermittent diagnosis?
3. A cause for iron deficiency must jaundice also supports a diagnosis 2. How urgent is the treatment of this
always be sought. In this man, it is of haemolytic anaemia. blood disorder?
Case histories 111

Notes to Case 3 Notes to Case 4 Symptoms may only become


1. The combination of the symptoms, 1. The lack of symptoms, absence of apparent after 6 months when the
the blood count, the coagulation test palpable lymphadenopathy and the baby begins to crawl.
results and the appearance of the normal haemoglobin level and 2. The most common form of
leukaemic blast cells strongly platelet count are consistent with haemophilia is haemophilia
suggest that this is acute myeloid stage A disease. A which is characterised by a
leukaemia with t (15;17) (q22;q12) 2. This is an entirely incidental deficiency of factor VIII. A factor
(M3:M3v). This subtype of the diagnosis of early stage chronic VIII assay was performed and this
disease is also known as acute lymphocytic leukaemia in an elderly revealed a very low level (1 unit/dL)
promyelocytic leukaemia. It is person. The prognosis is very good of the coagulation factor consistent
associated with a high incidence and there is no immediate need with severe disease.
of disseminated intravascular for treatment. The patient can be
coagulation and a high risk of reassured and periodically followed Case 6 [pp. 68–69]
spontaneous bleeding into vital up in the haematology outpatient
A 62-year-old woman is admitted to
organs. The blood film is shown clinic.
hospital with a chest infection. She has
on page 40 as Figure 20.2b.
no history of any blood disorder and a
2. Very urgent. Immediate treatment Case 5 [pp. 72–73] normal blood count. Her infection is
with the differentiating agent
treated with intravenous antibiotics.
all-trans retinoic acid (ATRA) A 10-month-old male infant is brought
She is also prescribed a daily
reduces the risk of early death from to the accident and emergency
subcutaneous dose of low molecular
bleeding. Tests to make a more department by his very anxious
weight heparin as prophylaxis against
definitive diagnosis (e.g. molecular parents. They report that he has had
thromboembolism. After a week in
testing to detect the genetic more bruising than his older siblings
hospital she is feeling much better
abnormality) should not delay the and that over the previous 2 days he
with improvement in her pneumonia
initiation of this treatment where has developed swelling of the left knee
but the medical team notice that her
there is clinical suspicion of the which he is now reluctant to bend.
platelet count is starting to fall. By day
disorder. There is no history of trauma and he
10 of her admission it is 65 × 109/L. She
has been otherwise well with normal
has no symptoms of bleeding. On the
Case 4 [pp. 46–47] development. There is no family
following day, she develops a swollen
history of a similar problem. On
A 72-year-old woman has a full blood left lower leg.
examination, the baby is irritable. He
count checked as part of the routine
is confirmed to have multiple small 1. What is the most likely cause of the
monitoring of her diabetes. She is
ecchymoses and a swollen left knee. thrombocytopenia?
feeling very well and wonders whether
The doctor decides to check a full 2. What is the most likely cause of the
her doctor really needs to take these
blood count and some first-line leg swelling?
blood samples. The full blood count
coagulation tests. The blood count is
is surprisingly abnormal, as follows:
normal but the coagulation screen is Notes to Case 6
Haemoglobin 121 g/L abnormal as follows: 1. This patient’s ‘isolated
MCV 88 fl thrombocytopenia’ (her
Prothrombin time 11 seconds (9–12)
MCH 30 pg haemoglobin level and white cell
Activated partial 82 seconds
White cell count 30.4 × 109/L count were normal) occurred
thromboplastin (26–36)
  Neutrophils 2.4 following her admission to
time
  Lymphocytes 21.5 hospital. A diagnosis of immune
Fibrinogen level 3.1 g/L (1.7–4.5)
  Monocytes 0.8 thrombocytopenia (ITP) is possible
  Eosinophils 0.2 1. Suggest a possible diagnosis. but the timing is suspicious of some
  Basophils 0.1 2. What further blood test would you other cause. She is not obviously ill
Platelet count 262 × 109/L request to confirm this diagnosis? enough for the low platelet count
to be a sign of disseminated
Inspection of the blood film reveals
Notes to Case 5 intravascular coagulation (DIC)
the increased lymphocytes to resemble
1. The presence of multiple bruises but it would be sensible to check
normal lymphocytes but they are
and joint swelling in a baby raises first-line coagulation tests. The most
perhaps a little larger. Numerous
the possibility of non-accidental likely cause is a newly commenced
‘smear cells’ are noted. Flow cytometry
injury. It is important to remember drug and heparin is the obvious
of the cells confirms a diagnosis of
that a bleeding disorder can lead candidate. The drug chart should
chronic lymphocytic leukaemia. The
to a very similar presentation. In be carefully scrutinised to exclude
blood film is illustrated in the chronic
this case, the combination of the other agents which might be
lymphocytic leukaemia section (Fig.
symptoms and the very prolonged responsible (see p. 69, Table 34.3).
23.2). Subsequent examination of the
activated partial thromboplastin 2. Heparin induced thrombocytopenia
patient is normal with no palpable
time (APTT) is very suggestive of (HIT) is a distinct syndrome
lymphadenopathy or
haemophilia. The lack of a family characterised by the development of
hepatosplenomegaly.
history does not rule out this antibodies to platelet factor 4 and
1. What is the stage of the disease? diagnosis as up to 30% of all new heparin. The thrombocytopenia is
2. What is your treatment plan for this cases of haemophilia are due typically non-severe and occurs
lady? to recent sporadic mutations. 5–10 days after starting heparin.
112 CASE HISTORIES

Unlike other drug-induced are designed to provide protection potential long-term side-effects of
thrombocytopenias, HIT leads to an against commonly implicated chemotherapy (e.g. increased risk of
increased risk of thromboembolism. organisms (see p. 87, Table 43.2). second malignancy, sterility, cardiac
This patient had developed a deep disease, lung damage, endocrine
vein thrombosis; an ultrasound Case 8 [pp. 58–59] dysfunction) should be fully
of the affected leg ruled out a acknowledged.
A previously well 32-year-old
haematoma and confirmed this
man presents to his primary care
diagnosis. Heparin was discontinued
practitioner complaining of a 12 week
Case 9 [pp. 78–79]
and an alternative anticoagulant
history of malaise, anorexia, weight A 27-year-old woman presents to the
substituted.
loss and drenching night sweats. It is hospital accident and emergency
calculated that he has lost 15% of his department complaining of a day’s
Case 7 [pp. 86–87] normal body weight. He has also history of a swollen right lower leg.
found a lump in his left neck which This developed shortly after a long
A 42-year-old man is diagnosed as
the doctor identifies as a 3 cm flight. An ultrasound scan of the leg
having stage IV diffuse large B-cell
enlarged non-tender cervical lymph confirms a deep vein thrombosis. The
non-Hodgkin’s lymphoma. He is treated
node. On more general examination, patient tells the doctor that she has
with the RCHOP chemotherapy
he has similar enlarged nodes in the a strong family history of venous
regimen. Two weeks after the first
axillary and inguinal regions but thrombosis and that she has previously
cycle, he presents to the haematology
nothing else. He is urgently referred tested positive as being heterozygous
outpatient clinic complaining of
to the local hospital where a cervical for the factor V Leiden mutation. This
general malaise. There are no more
lymph node biopsy confirms the is confirmed in her hospital notes
specific symptoms. He is initially
suspected diagnosis of classical – other thrombophilia tests were
reviewed by the clinic nursing staff
Hodgkin’s lymphoma (nodular normal. She is otherwise entirely well
who report him to appear pale and
sclerosing histological subtype). A and has had no previous episode of
unwell. He has a pyrexia, a slight
blood count reveals moderate anaemia thrombosis.
tachycardia and a normal blood
and an eosinophilia. A CT scan of the
pressure. A blood sample is taken for 1. What is the increased risk of venous
neck, chest and abdomen confirms the
investigations including a full blood thrombosis in people with the
enlarged cervical, axillary and inguinal
count: factor V Leiden mutation?
nodes and also shows significant
2. Was it the correct decision to screen
Haemoglobin 115 g/L mediastinal and intra-abdominal
her for this mutation?
MCV 83 fl lymphadenopathy. The liver and spleen
3. How will the presence of
MCH 31 pg are normal. Bone marrow examination
heterozygosity for the factor V
White cell count 0.8 × 109/L is also normal.
Leiden mutation alter the immediate
  Neutrophils 0.1
1. What is the stage of this man’s clinical management?
  Lymphocytes 0.6
Hodgkin’s lymphoma?
  Monocytes 0.1
2. Suggest a treatment plan. Notes to Case 9
Platelet count 132 × 109/L
3. What is the prognosis? 1. Heterozygosity for the factor V
1. You are asked for your opinion? Leiden mutation gives a 4–8-fold
What diagnosis do you suspect? Notes to Case 8 increase in risk for venous
2. What is your management plan? 1. He has lymph node involvement thrombosis. This is a relatively small
above and below the diaphragm but risk and thrombosis often only
Notes to Case 7 no evidence of disease outside nodal occurs where there are coexistent
1. This man has severe neutropenia areas (e.g. liver, bone marrow). He risk factors (e.g. prolonged
following chemotherapy. His has night sweats and significant immobility). Homozygosity carries
malaise and pyrexia are very likely weight loss. This is stage IIIB a much greater venous thrombotic
due to neutropenic sepsis. It is disease (see p. 59, Fig. 29.3). risk, around 50–100-fold.
common for the symptoms of 2. Stage IIIB disease is ‘advanced stage 2. We must presume that she was
infection in these patients to be disease’. He should receive a full screened because one or more of
vague with no localisation. course of chemotherapy with the her relatives tested positive for the
2. Suspected neutropenic sepsis is a possible addition of radiotherapy for factor V Leiden mutation and/or
medical emergency. A significant bulky disease. The ABVD regimen is other thrombophilia abnormalities
delay in the empirical most commonly used. ‘Alternating’ after an episode of venous
administration of intravenous or ‘hybrid’ regimens containing a thrombosis. Testing strategies for
broad-spectrum antibiotics leads to larger number of drugs are under thrombophilia are controversial
increased mortality. He should have investigation. The role of PET but there is generally no indication
blood and other relevant cultures scanning in staging and assessment for case finding of asymptomatic
performed prior to starting of response to treatment is also relatives with low risk
antibiotics but investigations should under review. thrombophilia (such as factor V
never significantly delay antibiotic 3. In younger patients, the prognosis Leiden heterozygosity). Her testing
treatment. The antibiotics can be of Hodgkin’s lymphoma is generally was very likely unnecessary.
commenced in the clinic before good with cure rates of 80% 3. Patients with heritable
transfer to the haematology ward. achievable even in advanced disease. thrombophilia should have a deep
The empirical antibiotic regimens With such high cure rates, the vein thrombosis treated with
Case histories 113

heparin and warfarin as in patients drugs and viral infections must be level and packed cell volume (also
without heritable thrombophilia. excluded (p. 52, Tables 26.1 and known as the haematocrit).
Thus, the presence of the factor V 26.2). This man had idiopathic 2. This is secondary polycythaemia
mutation does not affect her aplastic anaemia. caused by the hypoxia of chronic
immediate management. The 3. The blood results are consistent lung disease. The clinical history,
precise role of thrombophilia testing with very severe aplastic anaemia low oxygen saturation and raised
in clinical decision making remains (p. 53, Table 26.4). erythropoietin level are all
nuclear. 4. In a young fit person with very supportive of this diagnosis. In this
severe aplastic anaemia, the best patient, further investigations for
definitive therapy is likely to be the cause of polycythaemia, such as
Case 10 [pp. 52–53] an allogeneic stem cell transplant screening for the JAK2 mutation
from an HLA-matched sibling or and bone marrow examination, are
A 25-year-old man presents to his
unrelated donor. In the meantime, unnecessary (p. 64, Fig. 32.2).
primary care practitioner with a 6
he will need judicious use of red 3. The management of secondary
week history of increasing fatigue and
cell and platelet transfusions to polycythaemia is essentially that of
shortness of breath on exertion. He
manage symptoms arising from the underlying cause. This man’s
has also noticed an intermittent sore
anaemia and thrombocytopenia. If polycythaemia is likely to improve
throat and that he has been bruising
he develops a systemic infection, he if he stops smoking and if his
more easily than usual. He has
will need prompt broad-spectrum respiratory function can be
been previously well and is on no
antibiotic treatment in view of his improved. The role of venesection
medication. On examination, he
severe neutropenia. in this situation is not well defined.
appears pale and there are a few small
It may be helpful where the
ecchymoses. His doctor suspects
haematocrit is very high (e.g. greater
anaemia and decides to perform a full Case 11 [pp. 64–65]
than 0.55).
blood count:
A 74-year-old man presents to his
Haemoglobin 64 g/L primary care practitioner complaining Case 12 [pp. 50–51, 84–85]
MCV 94 fl of a week’s history of cough and
A 62-year-old woman with
MCH 30 pg increased dyspnoea. His medical
myelodysplastic syndrome receives
Reticulocyte count 0.1% records reveal that he is a smoker and
regular blood (red cell) transfusions to
White cell count 0.9 × 109/L that he has a history of chronic lung
improve her haemoglobin level and
  Neutrophils 0.1 disease. The doctor’s initial diagnosis
relieve symptoms of anaemia. She has
  Lymphocytes 0.7 of a chest infection is confirmed by a
received these without problems every
  Monocytes 0.1 chest X-ray and he prescribes a course
3–4 weeks for the previous 6 months.
Platelet count 9 × 109/L of oral antibiotics. As the patient has
On this occasion, the nurses on the
also complained of more longstanding
On receipt of the blood count result, hospital haematology day unit inform
fatigue he decides to check some blood
the doctor makes an urgent referral the duty doctor that the patient has
tests including a full blood count:
to the local hospital haematology become suddenly unwell only a
department. Here, the blood count is Haemoglobin 196 g/L few minutes after the start of the
repeated and the result confirmed. Packed cell volume 0.54 transfusion of the first unit of red cells.
There are no abnormal cells in the (haematocrit) She is complaining of chest and loin
blood film. A bone marrow aspirate MCV 95 fl pain and has become acutely short
and trephine procedure is performed. MCH 31 pg of breath. Her clinical observations,
This shows a grossly hypocellular White cell count 9.8 × 109/L normal before the start of the
bone marrow appearance with no Platelet count 182 × 109/L transfusion, now show pyrexia,
abnormal cells. tachycardia and hypotension. The
In view of this abnormal result, the
doctor is alarmed.
1. How could you describe the patient is referred to the haematology
abnormal blood count in one word? department at the local hospital. More 1. What diagnosis should he suspect?
2. What is the likely diagnosis? investigations are performed including 2. What immediate action should he
3. What is the severity of this the following: take?
disorder?
Oxygen saturation 89% (95–100)
4. Suggest a treatment plan. Notes to Case 12
Serum erythropoietin level 28 mU/mL
1. The doctor should strongly suspect
(2–20)
Notes to Case 10 a potentially fatal haemolytic
1. Pancytopenia. There is reduction in 1. What is the abnormality in the transfusion reaction. The acute onset
haemoglobin level, white cell count blood count? of the symptoms within minutes of
and platelet count. 2. What is the most likely cause of this starting the red cell transfusion and
2. The marked hypocellularity of the abnormality? the changes in temperature, pulse
bone marrow is consistent with 3. What is the best treatment of the and blood pressure are all highly
aplastic anaemia. There are no blood abnormality? suggestive of this.
abnormal cells to suggest infiltration 2. He should immediately stop the
by leukaemia or another malignant Notes to Case 11 transfusion and maintain the
disorder. Possible causes of 1. Polycythaemia. This is indicated by venous access with crystalloid. The
secondary aplastic anaemia such as the significantly raised haemoglobin transfused unit should be checked
114 CASE HISTORIES

(is another patient about to get a treatment with high dose melphalan the level of fetal haemoglobin in red
‘wrong’ unit due to a mix-up?) and and autologous stem cell cells.
the blood bank informed. Urgent transplantation. He should also
measures to resuscitate the patient receive bisphosphonate therapy.
Case 15 [pp. 76–77]
should be undertaken as necessary.
If an ABO incompatible transfusion A 55-year-old woman is admitted to
Case 14 [pp. 34–35]
is confirmed, or even suspected, hospital with a chest infection. Despite
senior medical advice should be A 25-year-old man with sickle cell prompt antibiotic treatment she
quickly sought to ensure proper anaemia (Hb SS) presents to the deteriorates and develops the
investigation, to guide the casualty department of his local symptoms and signs of severe sepsis.
management of complications hospital with a 24 hour history of She is transferred to the intensive care
(e.g. renal failure, disseminated increasing back and shoulder pain unit. Here it is noted that she has
intravascular coagulation) and to unresponsive to oral analgesics. He has oozing of blood from her cannulation
establish the need for intensive care had numerous episodes of similar pain sites. The medical team request a full
input. in the past, some of which have led to blood count and coagulation tests:
admission to hospital. He is clearly in
Haemoglobin 102 g/L
Case 13 [pp. 62–63] distress with severe pain but the doctor
MCV 81 fl
can elicit no other symptoms.
A previously well 59-year-old man MCH 30 pg
His medication includes a folate
presents with a 3 month history of White cell count 9.2 × 109/L
supplement, penicillin and
persistent increasingly severe back and Platelet count 31 × 109/L
hydroxycarbamide. His full blood count
rib pain. He also complains of loss of Prothrombin time 26 seconds (9–12)
is as follows:
energy. There is nothing diagnostic on Activated partial 58 seconds
examination although he appears pale Haemoglobin 72 g/L thromboplastin (26–36)
and unwell. The doctor decides to MCV 90 fl time
arrange some investigations including MCH 32 pg Fibrinogen level 0.8 g/L (1.7–4.5)
blood tests. These show the patient White cell count 9.8 × 109/L Fibrin degradation Increased
to have a moderate normocytic Platelet count 365 × 109/L products (FDPs)
normochromic anaemia with a blood
1. Why does the patient not have 1. What is the likely cause of the
film reported as ‘rouleaux formation’.
more obvious symptoms from his patient’s bleeding?
The biochemical tests reveal mild renal
significant anaemia? 2. How should this acquired bleeding
failure and hypercalcaemia.
2. Explain the aetiology of his pain. disorder be managed?
1. This man has a haematological 3. How would you manage his
malignancy. What is the likely symptoms? Notes to Case 15
diagnosis? 4. What is the mechanism of action of 1. The combination of
2. Suggest further investigations to hydroxycarbamide in this disease? thrombocytopenia, prolonged
confirm the suspected diagnosis. prothrombin time (PT) and
3. Suggest a treatment plan. Notes to Case 14 activated partial thromboplastin
1. When anaemia is chronic as in time (APTT), reduced fibrinogen
Notes to Case 13 sickle cell disease the symptoms are level and increased fibrin
1. The combination of chronic severe often less pronounced than when degradation products (or D-dimers)
back and rib pain, normocytic anaemia of similar severity (i.e. in a very ill patient with widespread
normochromic anaemia, rouleaux similar haemoglobin level) develops bleeding is characteristic of
formation in the blood film, renal acutely. Also, because HbS releases disseminated intravascular
failure and hypercalcaemia is highly oxygen more readily than HbA, the coagulation (DIC).
suggestive of myeloma. symptoms of anaemia are often 2. The cornerstone of management
2. Vital tests to establish the diagnosis surprisingly mild. of DIC is the treatment of the
of myeloma include serum and 2. He has a vaso-occlusive crisis. This underlying disorder – in this case
urine electrophoresis to detect a is caused by polymerisation of sepsis. In addition, bleeding may
monoclonal protein and bone deoxygenated HbS leading to be reduced by the transfusion of
marrow examination (aspirate and inflexible sickle cells becoming platelets, fresh frozen plasma (FFP
trephine biopsy) to assess the lodged in small vessels. This in turn – a source of coagulation factors)
number of clonal plasma cells. leads to oxygen deprivation of and cryoprecipitate (a source of
The degree of bone disease can be tissues and avascular necrosis of the fibrinogen). Careful monitoring of
determined with a combination of bone marrow. Over 90% of hospital the bleeding symptoms and
traditional X-rays (‘skeletal survey’) admissions for patients with sickle repeated blood counts and
and MRI scanning. cell disease are for a painful crisis. coagulation tests are vital to guide
3. The above tests confirmed myeloma 3. Management should include rest, the appropriate use of these blood
with numerous bony lesions in the warmth, intravenous fluids, opiate products. Recombinant activated
ribs and spine. In a younger fitter analgesia and reassurance. Patients protein C may be of benefit in DIC
patient such as this man, induction who are ill should be referred associated with sepsis and could be
chemotherapy (see p. 63) is urgently for more specialist care. considered here.
generally followed by stem cell 4. Hydroxycarbamide can reduce the
harvesting and intensification of severity of the disease by increasing
115

Index
Page numbers ending in ‘b’, ‘f ’ and ‘t’ refer to Boxes, Figures and Tables respectively

lymphocytosis, 9 Adult T-cell leukaemia lymphoma haemolytic see Haemolytic


A management and outcome, 43 (ATLL), 39f, 49 anaemia
poor prognosis, factors Afibrinogenaemia, 75 hypersplenism, 11
AA see Aplastic anaemia (AA) predicting, 43t Afro-Caribbean population, sickle iron deficiency see Iron
Abdomen, examination, 17 subtypes, 42 cell anaemia, 34 deficiency anaemia
Abdominal lymph nodes, Acute monocytic leukaemia, 40f Agglutination, cold (cold macrocytic, 22, 25, 27
examination, 16 Acute myeloid leukaemia (AML) autoimmune haemolytic management, 23
Abnormalities and acute lymphoblastic anaemia), 31f megaloblastic see Megaloblastic
blood count, 15 leukaemia, 40 AIHA see Autoimmune haemolytic anaemia
clotting tests, first-line, 20t age of presentation and AML anaemia (AIHA) microcytic, 22
coagulation see Coagulation characteristics, 41f Albumin, 20, 85 morphological classification, 22
disorders allogeneic stem cell Alcohol misuse, 15, 96 myelodysplastic syndromes, 50
common haematological, 14t transplantation, 56 Aldosterone, 4 myelofibrosis, 67
fibrinogen, 75 and aplastic anaemia, 53 Alemtuzumab, 48, 53, 55 myeloma, 62
genetic see Genetic blood count and film, 40 ALK1 (gene mutation), 71 normochromic, 22
abnormalities bone marrow aspirate and Alkylating agents, 54 normocytic, 22
haemoglobin, 29 trephine, 40 ALL see Acute lymphoblastic packed cell volume, 22
lymphocytes, 9 chemotherapy, 41 leukaemia (ALL) pernicious, 16, 26–27, 88
neutrophils, 7 chromosome deletions and Allergic disorders, 7, 15 platelet function disorders, 71
red cells additions, 38 Alloantibodies, 30–31 in pregnancy, 22, 88
indices, 22 classification, 40 Allogeneic stem cell of prematurity, 90
metabolism, 29 clinical features, 40 transplantation prevalence, 22
splenic, 10–11 common genetic abnormalities, acute lymphoblastic leukaemia, prolymphocytic leukaemia, 48
stomach and ileum (vitamin B12 41t 43 symptoms attributable to, 14
deficiency), 27 cytochemistry, 40–41 adult T-cell leukaemia T-cell large granular lymphocyte
ABO blood grouping, 82–83 cytogenetics, 41 lymphoma, 49 leukaemia, 48–49
neonatal disorders, 84, 90 diagnosis, 40–41 aplastic anaemia, 53 unexplained, in elderly people,
platelet transfusion, 85 epigenetic therapies, 55 bone marrow harvesting, 107 92
ABVD (doxorubicin, bleomycin, essential thrombocythaemia, chronic lymphocytic leukaemia, see also Haemoglobin; Red cells
vinblastine, dacarbazine) progression from, 66 47 Anaemia of chronic disease (ACD),
chemotherapy protocol, 59 FLT3 inhibitors, 55 chronic myeloid leukaemia, 45 36–37
Aciclovir, 87 fluorescence in situ defined, 56 Anagrelide, 66
Acidosis, 85 hybridisation, 38f herpes zoster following, 87f Anaphylaxis, 25
Acrocyanosis, 30 immunophenotyping, 41 myelodysplastic syndromes, 51 Angiogenesis, bone marrow
Actin, 4 management, 41 non-myeloablative (‘reduced abnormality, 67
Actin–myosin assembly, cell molecular biology, 41 intensity’), 57 Angular stomatitis, 24f, 26
membrane, 6 monoclonal antibodies, 55 and syngeneic SCT, 56 Ankyrin, 4
Activated partial thromboplastin multi-drug resistance, 54 thalassaemias, 33 Anopheline mosquitoes, 98
time (APTT) and myelodysplastic syndromes, Alloimmune thrombocytopenia, Anorexia, malignant disease, 95
acquired haemophilia, 77 39, 50 91 Antecubital fossa, veins at, 104
disseminated intravascular and myeloproliferative disorders, All-trans-retinoic acid (ATRA), 41, Anterior iliac crest, 106–107
coagulation, 76 39 55 Anthracycline, 41, 54
haemostasis, 12–13 peripheral blood film, 39f Alpha-1-antitrypsin, acute phase Anti-angiogenic agents, 55
and heparin, 80 platelet function disorders, 71 response, 21 Antibiotic treatment, 87, 87t
laboratory haematology, 20 polycythaemia, 65 Alpha-2-macroglobulin, 20 Antibodies
prolonged, in haemophilia, 72 prognosis, 41 Alteplase, 81 antileucocyte, 84
vitamin K deficiency, 77 smoking, 15 AML see Acute myeloid leukaemia HTLV-1, 49
Von Willebrand disease, 75 stem cell transplantation, 41 (AML) immune, 82–83
see also Prothrombin time (PT) supportive care, 41 Anaemia isoantibodies, 30–31
Activated protein C (APC), 78, 102 tyrosine kinase inhibitors, 55 acute lymphoblastic leukaemia, monoclonal, 55
Activating receptors, natural killer Acute obstetric haemorrhage, 89t 42 naturally occurring, 82
(NK) cells, 9 Acute phase response acute phase response, 20 Antibody screening, 82–83
Acupuncture, 95 in elderly people, 92 aetiological classification, Anti-CD33 (Mylotarg), 55
Acute graft-versus-host disease, 56 lack of, in essential 22–23 Anti-chymotrypsin, acute phase
Acute immune thrombocytopenia thrombocythaemia, 66 aplastic see Aplastic anaemia response, 21
(ITP), 68, 68t, 69f measurement, 20–21 (AA) Anticoagulation
Acute lymphoblastic leukaemia Acute promyelocytic leukaemia, blood count, 18 in elderly people, 92
(ALL) 40–41 of chronic disease see Anaemia heparin, 80
and acute myeloid leukaemia, Acute venous thrombosis, 79 of chronic disease (ACD) new agents, 81
40 Adaptive immune response, 9 classification, 22–23, 23f new drugs, 102–103
in adults, 43 Adeno-associated virus (AAV), congenital dyserythropoietic in pregnancy, 89
allogeneic stem cell 103 anaemias, 91 thrombolytic therapy, 81
transplantation, 56 Adenopathy, 16 definition, 22 warfarin, 80–81, 80f
in children, 43 Adenosine triphosphate (ATP), 5 Diamond–Blackfan, 91 Anti-D immunoglobulin (Ig), 90
classification, 42 Adhesion molecules dilutional, in pregnancy, 88 Antidiuretic hormone (ADH), 4
clinical features, 42 bone marrow structure, 2 in elderly people, 22–23, 92 Antiemetics, 95
diagnosis, 42–43 neutrophils, 6 Fanconi’s anaemia, 52 Antifibrinolytic agents, 70, 73
incidence, at different ages, 42f splenic structure, 10 general features, 22 Antigen-presenting cells, 8f, 10
116 INDEX

Antiglobulin test, 83f Autologous stem cell see also Blood count; Blood aplastic anaemia, 53
Anti-helminthic agents, 99 transplantation, 56–57 groups; Blood tests; Blood complications, 84–85
Antileucocyte antibodies, 84 acute lymphoblastic leukaemia, transfusion; Plasma; Platelet delayed complications, 84–85
Anti-leukaemic drugs, 54 41, 43 function disorders; Platelets in elderly people, 92
Antimetabolites, 54 bone marrow harvesting, 107 Blood count granulocytes, 85
Antimicrobial prophylaxis, 87 Hodgkin’s lymphoma, 59 abnormalities, past medical haemolytic reactions, 84
Antiphospholipid antibody myeloma, 63 history, 15 immediate complications, 84
syndrome, 79 Automated haematology counters, acute lymphoblastic leukaemia, incompatibility, 82
Antithrombin (AT), 13, 79–80 18 42 infection, 84
Antithymocyte globulin (ATG), 51, Axillary lymphadenopathy, acute myeloid leukaemia, 40 massive, 85
53 lymphoma, 60f aplastic anaemia, 52 myelodysplastic syndromes, 51
Anti-tissue transglutaminase, 25 Axillary nodes, examination, 16 Hodgkin’s lymphoma, 58–59 non-haemolytic reactions, 84
Aorto-gonad-mesonephros (AGM) Azacitidine, 55 iron deficiency anaemia, 25 platelets, 71, 85
region, embryo, 2 Azathioprine, 30 laboratory haematology, 18 practicalities, 84
APCR (activated protein resistance Azoles, 87 lymphocytes, 9, 46 red cells, 84–85, 84t
ratio), 78–79 neutrophils, 6 safety factors, 82
Apixaban, 102–103 pernicious anaemia, 24 sickle cell anaemia (HbSS), 35
Aplastic anaemia (AA) B report, 18–19 testing before, 82–83
aetiology, 52 Von Willebrand disease, 75 transfusion associated
classification, 52 Babesiosis, 99 Blood films circulation overload, 84
clinical features, 52 Backache, myeloma, 62 acute lymphoblastic leukaemia, transfusion-related acute lung
diagnosis, 52–53 Bacterial infection, 86, 97 42 injury, 84
immunosuppression, 52–53 see also Antibiotic treatment acute myeloid leukaemia, 39f, see also under Platelets; Stem cell
management, 53 Basophils, 7 40 transplantation (SCT)
removal of cause, 53 B-cell lymphomas, 55, 97 anaemia of chronic disease, B-lymphocytes, 8–9
restoring normal BCL2 gene, non-Hodgkin’s 36–37 antigen-stimulated mature, in
haematopoiesis, 53 lymphoma, 60 aplastic anaemia, 52 CLL, 46
severity measurement, 53 BCR-ABL oncogene, 38 chronic lymphocytic leukaemia, cold autoimmune haemolytic
Apoptosis, 38, 60 chronic myeloid leukaemia, 46, 46f anaemia, 30
Arteriovenous malformations 44–45 chronic myeloid leukaemia, 44, germinal-centre, in HL, 58
(AVMs), 71 molecular biology, 101 45f splenic structure, 10
Ashkenazi Jews, factor VI treatments, 55 cold autoimmune haemolytic Bone disease, myeloma, 62–63
deficiency, 75 Bence Jones protein, myeloma, anaemia, cold agglutination Bone marrow
Aspergillus fumigatus, 86–87 62 in, 31f acute myeloid leukaemia
Aspirate, bone marrow Bendamustine, 47, 54 essential thrombocythaemia, 66f subtypes, appearance in,
acute lymphoblastic leukaemia, Bernard–Soulier syndrome, 70 haemolytic disease of the 40f
42 Biliary obstruction, vitamin K newborn, 90f angiogenesis, 67
acute myeloid leukaemia, 40 deficiency, 77 hereditary spherocytosis, 29 aspirate see Aspirate, bone
advantages and disadvantages, Bilirubin, 5 hyposplenism, 11f marrow
19 Biopsy iron deficiency, 25f examination, 19
aplastic anaemia, 52–53 lymph node making, 18–19 failure of production, in
aspiration procedure, 106–107 Hodgkin’s lymphoma, 58, 58f megaloblastic anaemia, 27f thrombocytopenia, 68
chronic lymphocytic leukaemia, non-Hodgkin’s lymphoma, microangiopathic haemolytic haematopoiesis, 2–3, 3f
46 60–61 anaemia, 31f harvesting, 107
examination procedure, 19 trephine see Trephine biopsy, morphological terms, 19t normal, 2f
hairy cell leukaemia, 48 bone marrow myelofibrosis, 66f prostatic carcinoma, 97f
increased reticuloendothelial Bisphosphonate, myeloma, 63, 94 myeloma, 62f regulators, haematopoiesis, 2–3,
iron stores in anaemia of Black people, neutrophil counts, 7 pernicious anaemia, 24 3f
chronic disease, 37f Blast cells sickle cell anaemia (HbSS), 34f specific conditions, effects
iron deficiency anaemia, 25 acute lymphoblastic leukaemia, thalassaemias, 33f Hodgkin’s lymphoma, 58–59
in megaloblastic anaemia, 26f 42 thrombocytopenia, 68f–69f myelodysplastic syndromes,
needle, 106f acute myeloid leukaemia, 40 warm autoimmune haemolytic 50
pernicious anaemia, 26 morphology, 42f anaemia, 30f myelofibrosis, 66
polycythaemia, 64 Blast crisis, chronic myeloid Blood groups myeloma, 62, 62f
systemic disease, 96 leukaemia, 44–45 ABO group, 82–85, 90 stem cell hierarchy, 2, 3f
thrombocytopenia, 69 Bleeding gel system, using, 82f, 83 structure, 2
Aspirin disseminated intravascular incompatibility, 82, 90 trephine appearance, in human
essential thrombocythaemia, 66 coagulation, 76 tests, 82–83 T-cell leukaemic lymphoma,
platelet function disorder caused haemophilia A, 72–73, 72f Von Willebrand disease, 75 39f
by, 70–71 intracranial see Intracranial Blood tests Bortezomib, 55, 63
polycythaemia, 64–65 bleeding antibody screening, 83 Bovine spongiform
Asplenia syndrome, 10–11 liver disease, 77 antiglobulin, 83f encephalopathy (BSE), 84
Asplenism, 10–11, 11t myeloma, 62 blood grouping, 82–83 Brentuximab, 55, 59
ATG see Antithymocyte globulin thrombocytopenia, 14, 68 chronic myeloid leukaemia, 44f Bronchoalveolar lavage, 86
(ATG) tranexamic acid, 73 crossmatching, 83 Bruising, acquired haemophilia,
ATRA see All-trans-retinoic acid Von Willebrand disease, 74–75 donor blood, 82 76f
(ATRA) Bleeding time, 70–71 prior to transfusion, 82–83 Bullae, haemorrhagic, 76f
Autoantibodies, 27, 30 Bleomycin, 59 see also Venepuncture; Venous Burkitt’s lymphoma, 42, 60–61
Autoimmune haemolysis, 46 Blood access endemic, 99
Autoimmune haemolytic anaemia clotting of see Clotting of blood Blood transfusion Burr cells, renal failure, 96
(AIHA), 30 lymphocytes, mature, 8f acute myeloid leukaemia, 41 Busulfan, 54
Autoimmune thrombocytopenia, ordering practicalities, 83 allogeneic, alternatives to, 85 ‘Butterfly needle,’ venepuncture,
46 peripheral see Peripheral blood anaemia, 23 104
Index 117

Chronic granulomatous disease, 7 Coagulation cascade Cyanmethemoglobin method,


C Chronic immune defects, 14 automated haematology
thrombocytopenia (ITP), haemostasis, 12, 13t counters, 18
Cachexia, malignant disease, 95 68t, 69 Coagulation disorders Cyanotic congenital heart disease,
Cancer see Malignant disease Chronic lymphocytic leukaemia acquired, 76–77 65f
Candida infection, 86 (CLL) chronic renal failure, 96 Cyclical neutropenia, 7
Cannulation, 105 clinical features, 46 disseminated intravascular Cyclooxygenase-1 (COX-1), 70–71
Carbon dioxide, transport, 4 diagnosis, 46 coagulation see Cyclophosphamide, 54
Cardiac overactivity, anaemia, 22 in elderly people, 46, 93 Disseminated intravascular acute lymphoblastic leukaemia,
Cardiopulmonary bypass, platelet humoral immunity defects, 86 coagulation (DIC) 43
function disorders, 71 incidence, 38 factor deficiencies, 75 aplastic anaemia, 53
Caspofungin, 87 lymphocytosis, 9 fibrinogen abnormalities, 75 chronic lymphocytic leukaemia,
CD4 surface marker management, 46–47, 47f haemophilia, acquired, 76f, 77 47
HIV/AIDS, 9 monoclonal antibodies, 55 history taking, 14 diffuse large B-cell lymphoma, 61
and T-lymphocytes, 8 prognosis, 47t hypercoagulability in malignant follicular lymphoma, 61
treatment advances, 102 staging, 46 disease, 96 myeloma, 63
CD8 surface marker, 8 whole genome sequencing, liver disease, 77 Cytochemistry, 40–42
CD20 antigen, 55 101f in pregnancy, 89 Cytogenetics, 41, 43
Cell cycle, 54 Chronic myeloid leukaemia vitamin K deficiency, 76–77 Cytokines, 6, 10, 66
Cellulose acetate electrophoresis, (CML) Coagulation factors, 12 Cytomegalovirus (CMV)
35f accelerated phase, 44–45 Coeliac disease, splenic atrophy, pneumonitis, 87
Central nervous system (CNS) advanced disease, 45 10–11 red cell transfusion
disease allogeneic stem cell Cold autoimmune haemolytic complications, 84
acute lymphoblastic leukaemia, transplantation, 56 anaemia, 30 thrombocytopenia, 68
42 chronic phase, 44–45 Collagen receptors, 12 Cytopenia, supportive care, 55
monocytic/monoblastic clinical features, 44 Colon carcinoma, iron deficiency Cytoplasm, platelets, 12
leukaemia, 40 diagnosis and monitoring, anaemia, 24f Cytosine arabinoside, 54
non-Hodgkin’s lymphoma, 60 44–45 Colonoscopy, iron deficiency, acute lymphoblastic leukaemia,
Central venous cannulation, 105 drug therapy, 45, 45t, 55, 93 25 43
Cerebral malaria, 98 and essential thrombocythaemia, Colony-forming units (CFUs), 2 acute myeloid leukaemia, 41
Cervical lymph nodes, 66 Colony-stimulating factors (CSFs), Cytoskeleton, 4
enlargement, 16 fluorescence in situ 3 Cytotoxic drugs
Chemotaxis, 6 hybridisation, 100f Combination chemotherapy, 54, major classes, 54
Chemotherapy pathogenesis, 44 99, 102 side-effects, 54
acute myeloid leukaemia, 41 Philadelphia (Ph) chromosome, Comparative genomic see also Chemotherapy
combination, 54, 99, 102 38–39, 44, 44f hybridisation, 100
consolidation, 41 predisposing factors, 39 Complementary therapy, 95
elderly people see under Elderly treatment, 45, 55 Complete remission (CR), acute D
people Chronic myelomonocytic myeloid leukaemia, 41
general principles, 54 leukaemia (CMML), 50 Computed tomography (CT) scan, Dabigatran etexilate, 102–103
high-dose, in stem cell Chronic renal failure 59, 61 Dacarbazine, 59
transplantation, 56–57 platelet function disorders, 71 Congenital dyserythropoietic Dactylitis, sickle cell anaemia, 34f
Hodgkin’s lymphoma, 59 systemic disease, 96 anaemias (CDAs), 91 Dasatinib, 45, 55
immunosuppression in, 86 Chronic thrombocytopenia, 68 Connective tissue disease, 71, 96 Daunorubicin, 54
induction, 41 Ciclosporin, 30, 53 Coombs’ test, autoimmune acute lymphoblastic leukaemia,
major classes of conventional Circulating pool, neutrophils, 6 haemolytic anaemia, 30 43
drugs, 54 Cirrhosis, hepatitis risk, 73 Cords of Billroth, 10 acute myeloid leukaemia, 41
multi-drug resistance, 54 Citrate agar electrophoresis, 21 Coronary atherosclerosis, 79 DDAVP (desmopressin) see
myelodysplastic syndromes, 51 Citrate toxicity, massive blood Corticosteroid treatment Desmopressin (DDAVP)
palliative, 94 transfusion, 85 anorexia and cachexia, in Decitabine, 55
phase-specific and phase Cladribine (2-CDA), 48, 49f malignancy, 95 Deep vein thrombosis (DVT), 78,
non-specific, 54 CLL see Chronic lymphocytic monocytopenia, 7 89
and Philadelphia chromosome, leukaemia (CLL) pain relief, 94 Desferrioxamine, subcutaneous, 32
43 Clostridium difficile, 86 in pregnancy, 89 Dendritic cells, follicular, 10
related treatments, 54–55 Clotting of blood, 12 see also Prednisolone; Steroid Desmopressin (DDAVP)
Children see Paediatric first-line tests, 20t treatment haemophilia, 73
haematology prothrombin time test, 20 Co-trimoxazole, 87 platelet function disorders,
Chlorambucil, 54 Clubbing of nails, polycythaemia, Counselling see Genetic 70–71
chronic lymphocytic leukaemia, 65f counselling Von Willebrand disease, 75
47 Cluster differentiation (CD), 21 C-reactive protein (CRP), 21, Developing world, diseases
follicular lymphoma, 61 CML see Chronic myeloid 36–37 affecting, 7, 22, 98–99
myeloma, 63 leukaemia (CML) Creutzfeldt-Jakob disease (CJD), as Dexamethasone, 63, 94
Chloroquine, 99 CMV see Cytomegalovirus contradiction to blood Dialysis, 71
Cholesterol, 4 (CMV) donation, 82 Diamond–Blackfan anaemia, 91
CHOP regimen, diffuse large Coagulation Crossmatching, blood tests, 83 DIC see Disseminated
B-cell lymphoma, 61 abnormal, symptoms Cryoprecipitate intravascular coagulation
Chromosomal abnormalities see attributable to, 14 acquired coagulation disorders, (DIC)
Genetic abnormalities disorders see Coagulation 76–77 Diet
Chromosome deletions and disorders transfusion, 85 malabsorption see
additions, leukaemia, 38 physiological pathways, 12–13, Crystalloid solution, 84, 91 Malabsorption
Chronic benign neutropenia, 7 13f CT scans see Computed vitamin B12 deficiency see
Chronic graft-versus-host disease, regulation of, 13 tomography (CT) scan Vitamin B12 deficiency
56 tests, 20 Cultures, cell, 2 and vitamin K deficiency, 77
118 INDEX

Differential white cell count, 18 Elderly people neutrophil counts, black people, Factor Xa, 12
Differentiating agents, 55 anaemia, 22–23, 92 7 Factor XI
Diffuse large B-cell lymphoma anticoagulation, 92 sickle cell anaemia, 34 activated partial thromboplastin
(DLBCL), 61, 100f chronic lymphocytic leukaemia Ethylene diamine tetra-acetic acid time, 20
Direct antiglobulin test (DAT), 30, in, 46, 93 (EDTA), 18, 21 coagulation disorders,
90 essential thrombocythaemia, 66 Etoposide, 54 deficiencies in, 75
Disseminated intravascular haematopoiesis, 92 Examination platelets, 12–13
coagulation (DIC) haemophilia and other inherited blood films, 18–19 Factor XII
acute myeloid leukaemia, 40 blood disorders, 77, 92–93 bone marrow, 19 activated partial thromboplastin
bleeding, 76 iron deficiency, 24, 92 of patient, 16–17 time, 20
diagnosis and management, 76 malignant disease, 92–93, 93t Exsanguination, 12 coagulation disorders,
fibrinogen, quantitation, 20 chemotherapy Extended spectrum beta- deficiencies in, 75
haemolytic transfusion reactions, contraindications, 41, 43, 93 lactamases (ESBLs), 86 platelets, 12–13
84 chronic lymphocytic Extracorporeal Factor XIII, 12–13, 75
haemorrhagic bullae and leukaemia, 46 photochemotherapy, adult Familial thrombophilia, 78–79
gangrene, 76f prolymphocytic leukaemia, 48 T-cell leukaemia lymphoma, Family history, 15
malignant disease, 96 thrombosis, 92, 92f 49 haemophilia A, 72
pathophysiology, 76f vitamin K supplementation, 92 Extrinsic pathway, platelets, 12–13, Fanconi’s anaemia, 52
in pregnancy, 88–89 Electrical impedance, cell counting 13f Fatigue, in malignant disease,
protein C and S deficiencies, and sizing, 18 Eyes, sickle cell anaemia, 34 94–95
78–79 Electronic crossmatching, 83 Felty syndrome, 96
thrombocytopenia in neonate, 91 Electron microscopy, 48f Fetus, sites of blood production in,
and thrombophilia, 78 Electrophoresis, 21 F 2f
DNA analysis techniques, 83, cellulose acetate, 35f Fibrinogen, plasma
100–101 electrophoretic strip, myeloma, FAB (French-American-British) abnormalities, 75
DNA synthesis, megaloblastic 62f classification, 39 acute phase response, 21
anaemia, 24 sickle cell anaemia, 34, 35f acute lymphoblastic leukaemia, pregnancy, increase in, 88
Donor blood, 82 thalassaemias, 33f 42 quantitation, 20
Donor lymphocyte infusions Embden–Meyerhof pathway, 5, 29 acute myeloid leukaemia, 40 Fibrinolysis/fibrinolytic system, 13
(DLIs), 57 Endemic Burkitt’s lymphoma, 99 myelodysplastic syndromes, Fibroblasts, Hodgkin’s lymphoma,
Doxorubicin, 54 Endemic malaria, 99 50 58
diffuse large B-cell lymphoma, End of life care, 95 Factor II, 12 Filariasis, 99
61 Endoglin (gene mutation), 71 Factor V Flow cytometry
Hodgkin’s lymphoma, 59 Endoscopy, iron deficiency, 25 activated partial thromboplastin acute myeloid leukaemia, 41
Doxycycline, malaria, 99 Endothelial cell barrier, 12 time, 20 haematopoiesis, 2
Drug history, 15, 69 Enterobacter spp., 86 coagulation disorders, laboratory haematology, 21
Drug-induced conditions Eosinophils, 7 deficiencies in, 75 platelet function disorders, 70
aplastic anaemia, 52t Epigenetics, 38–39, 55 platelets, 12–13 FLT3 inhibitors, acute myeloid
glucose-6-phosphate Epipodophyllotoxins, 54 prothrombin time test, 20 leukaemia, 55
dehydrogenase (G6PD) Epstein–Barr virus (EBV) Factor Va, 12 Fluconazole, 87
deficiency, 29 endemic Burkitt’s lymphoma, Factor V Leiden (FVR506Q), Fludarabine, 54
nausea and vomiting, 95 99 familial thrombophilia, 78 chronic lymphocytic leukaemia,
platelet function disorders, Hodgkin’s lymphoma, 58 Factor VII 47
70–71 mononucleosis, 97 platelets, 12–13 follicular lymphoma, 61
thrombocytopenia, 69 non-Hodgkin’s lymphoma, 60 pregnancy, increase in, 88 myeloma, 63
Drug treatment Erythrocyte sedimentation rate prothrombin time test, 20 prolymphocytic leukaemia, 48
anticoagulant therapy see (ESR) Factor VIIa, 12–13 Fluorescence in situ hybridisation
Anticoagulation acute phase response, factor VI deficiency, 75 (FISH), 38f, 100
side-effects see Side-effects measurement, 20 platelet function disorders, 70 Folate deficiency
specific conditions anaemia of chronic disease, Factor VIII megaloblastic anaemia, 26–27
chronic myeloid leukaemia, 36–37 activated partial thromboplastin in pregnancy, 88
45, 45t, 93 myeloma, 62 time, 20 Follicular lymphoma, 61, 102, 102f
leukaemia, 54 Erythropoiesis, 22–23, 64 C assay, 75 Fondaparinux, 80
malaria, 99 Erythropoietin, 4, 55 haemophilia, 72–73, 73f French-American-British
myeloma, 63, 94 anaemia of chronic disease, 37 clinical severity of classification see FAB
nausea and vomiting, 95 myelodysplastic syndromes, 51 haemophilia A and factor (French-American-British)
polycythaemia, 64–65 myeloma, 63 VIII level, 14, 72t classification
syringe drivers, continuous in pregnancy, 88 see also Haemophilia A Fresh frozen plasma (FFP)
infusions, 95 radioimmunoassay, estimation platelets, 12–13 acquired coagulation disorders,
see also Chemotherapy; by, 64 pregnancy, increase in, 88 76–77
Treatment of conditions; renal failure, in elderly, 92 Factor VIIIa, 12–13 indications for use, 85t
specific drugs Escherichia coli (E. coli), 31, 86 Factor VIIIc/tissue factor pathway Full blood count (FBC), 18
‘Dry tap,’ bone marrow aspiration, ESR see Erythrocyte sedimentation inhibitors, 102 Functional assay, vWF antigen, 75
106 rate (ESR) Factor IX Functional cytotoxicity assays,
Dyes, blood film, 18–19 Essential thrombocythaemia (ET) activated partial thromboplastin 102
Dysfibrinogenaemia, 75, 79 clinical features, 66 time, 20 Fungal infection, 86–87
Dyskeratosis congenita, 52 diagnosis, 66 haemophilia B, 73
Dyspnoea, 22, 95 management, 66 platelets, 12–13
polycythaemia, 64 Factor X G
splenic atrophy, 10–11 activated partial thromboplastin
E systemic enquiry, 15 time, 20 Gaisbock’s syndrome, 64–65
Ethnic origin platelets, 12–13 Gametocytes, 98
Edoxaban, 102–103 disorder type, 16 pregnancy, increase in, 88 Ganciclovir, 87
Ehlers–Danlos syndrome, 71 factor VI deficiency, 75 prothrombin time test, 20 Gangrene, 76f

tahir99-VRG & vip.persianss.ir


Index 119

G-CSF see Granulocyte colony- Graft-versus-tumour (GVT) effects, isoimmune, 30–31 haemophilia, 72–73
stimulating factor (G-CSF) 57 microangiopathic, 31, 31f marginal zone lymphoma, 60
Gel technology, blood grouping, Gram-negative bacilli, 86 and sickle cell anaemia, 34 thrombocytopenia, 68
82f, 83 Granulocyte colony-stimulating Haemolytic disease of the venepuncture precautions, 104
Gene profiling, 100 factor (G-CSF), 93 newborn (HDN), 90, 90f Hepatobiliary complications, sickle
General anaesthesia, bone marrow chemotherapy and related blood group incompatibility, 82, cell anaemia, 34
aspiration in children, 106 treatments, 55 90 Hepatocellular cancer, 73
General examination, 16 stem cell transplantation, 56–57 Haemolytic transfusion reactions, Hepatomegaly, 67
Gene therapy Granulocytes 84 Hereditary elliptocytosis, 29
haemophilia A, 73 blood transfusion, 85 Haemolytic uraemic syndrome Hereditary haemorrhagic
sickle cell anaemia (HbSS), 35 defined, 6 (HUS), 31 telangiectasia (HHT), 71
treatment advances, 103 Hodgkin’s lymphoma, 58 Haemophilia Hereditary pyropoikilocytosis, 29
Genetic abnormalities see also Basophils; Eosinophils; acquired, 76f, 77 Hereditary spherocytosis, 28f, 29,
17p deletions, in CLL, 47 Neutrophils in children, 15 86
adult T-cell leukaemia Growth factors on demand treatment, 72–73 Hereditary thrombophilia, 89
lymphoma, 48 aplastic anaemia, 53 in elderly people, 77, 92–93 Herpes simplex, 87
antenatal detection, 101 and bone marrow, 2–3 gene therapy, 103 Herpes zoster, 87f
aplastic anaemia, 52 haematopoietic growth factor joint examination, 16 Hexose monophosphate shunt
β-chain gene, in sickle cell therapy, 55 see also Haemophilia A; (pentose phosphate
syndromes, 34–35 neutropenia, 87 Haemophilia B pathway), 5
carrier detection, 101 Haemophilia A Hickman catheters, 105
haemophilia A, 72 access to therapy, 98 History taking
leukaemia, 38–39, 41t H bleeding, 72–73, 72f common haematological
acute lymphoblastic carrier state and genetic abnormalities, 14t
leukaemia, 42, 43t Haem, protoporphyrin of, 5 counselling, 73 presenting complaint, 14
acute myeloid leukaemia, 41 Haemarthroses (bleeding into clinical features, 72 systemic enquiry, 15
adult T-cell leukaemia joints), 14 clinical severity and factor VIII thrombophilia, 78
lymphoma, 49 Haematoma, haemophilia, 72 level, 14, 72t Von Willebrand disease, 74–75
chronic myeloid leukaemia, Haematopoiesis diagnosis, 72 HIV/AIDS
44–45 and ageing, 92 treatment complications, 72 CD4 count, 9
myelodysplastic syndromes, 50 aplastic anaemia, restoring in, 53 Haemophilia B, 73, 103 as contradiction to blood
myeloma, 62–63 bone marrow, 2–3, 3f Haemophilia centres, 72 donation, 82
non-Hodgkin’s lymphoma, 60 myelodysplastic syndromes, Haemophilus influenzae, 11 haemophilia, 72–73
Philadelphia chromosome see abnormalities in, 50 Haemorrhage and Hodgkin’s lymphoma, 58
Philadelphia (Ph) splenic structure, 10 acute obstetric, 89t immunosuppression, 86
chromosome Haematopoietic growth factor coagulation disorders, 14 systemic disease, 97
platelet function disorders, 71 therapy, 55 fibrinogen abnormalities, 75 thrombocytopenia, 68
polycythaemia, 64 Haematopoietic stem cells (HSCs), thrombocytopenia, 14, 68 Hodgkin’s lymphoma (HL)
in tropical regions, 98 2 see also Bleeding advanced stage disease, 59
see also Cytogenetics; Haematoxylin and eosin (H&E) Haemorrhagic bullae, 76f aetiology, 58
Epigenetics stain, 19 Haemosiderin, 28 age distribution, 58
Genetic counselling Haemoglobin Haemosiderinuria, 31f classical, 58
haemophilia A, 73 abnormalities, 29 Haemostasis, 12–13 classification, 58
sickle cell syndromes, 35 blood count, 18 complexity, 20 clinical presentation, 58
thrombophilia, 79 cellulose acetate electrophoresis, primary tests, 75 diagnosis, 58
Von Willebrand disease, 75 to separate, 35f Hairy cell leukaemia (HCL), 48, early stage disease, 59
Genitourinary complications, high concentration in 55 late effects of treatment, 59
sickle cell anaemia, 34 polycythaemia, 64 Ham test, paroxysmal nocturnal lymphocyte predominant
Gentamicin, 87 low concentration see Anaemia haemoglobinuria, 31 nodular, 58
Germ cells, prechemotherapy molecule, essential elements, 5f Haptoglobins, 21, 28 management, 59
storage, 54 normal concentrations, 22, 22t Hb-Bart hydrops syndrome, 32 monoclonal antibodies, 55
Gestational thrombocytopenia, 88 transport, 5 HbH disease, 32 prognosis, 59
Giemsa stain, 18–19, 98 Haemoglobinaemia, 30 HDN see Haemolytic disease of spleen, absent, 11
Glanzmann’s thrombasthenia, 70 Haemoglobin molecule (HbA), 5 the newborn (HDN) staging, 58–59, 59f
Globin chains, 5 Haemoglobinopathies Helicobacter pylori, 60, 68 Hormone replacement therapy
Glossitis in children, 15–16 HELLP syndrome (haemolysis, (HRT), 79
iron deficiency, 24f electrophoresis, 21 elevated liver enzymes and Howell–Jolly bodies, 10–11
pernicious anaemia, 26, 27f see also specific conditions, such low platelets), 89 HPA-1a (platelet antigen), 69
Glucose-6-phosphate as thalassaemia ‘Helper’ cells, 8 HTLV-1 antibodies, adult T-cell
dehydrogenase (G6PD) Haemoglobinuria, 30 Henoch–Schönlein purpura, 71 leukaemia lymphoma, 49
deficiency, 29 Haemolysis Heparin Human leucocyte antigens (HLA),
Glycoprotein (GP) la/Ha complex, autoimmune, 46 acute venous thrombosis, 79 8
12 autoimmune haemolytic cardiopulmonary bypass, 71 platelet transfusion, 85
Glycoprotein (GP) lb/IX/V anaemia, 30 low molecular weight (LMW), stem cell transplantation, 56
complex, 12, 70 chronic lymphocytic leukaemia, 79–80, 89, 102 Humoral immunity, 8
Glycoprotein (GP) llb/IIIa 46–47 in pregnancy, 89 defects, 86
complex, 12, 70 general features, 28–29 unfractionated, 80, 89 Hydronephrosis, 60
Gonadal failure, chemotherapy Haemolytic anaemia Heparin-induced Hydroxycarbamide
treatment, 54 acquired disorders, 30–31 thrombocytopenia (HIT), essential thrombocythaemia, 66
Gout, 64, 65f autoimmune, 30 69 myelofibrosis, 67
Graft-versus-host disease (GVHD), classification, 28t Hepatitis polycythaemia, 64–65
56, 57f diagnosis, 28–29 aplastic anaemia, 52 sickle cell anaemia (HbSS), 35
Graft-versus-leukaemia (GVL) general features and inherited as contradiction to blood Hypercoagulability, in malignant
effects, 57 disorders, 28–29 donation, 82 disease, 96

tahir99-VRG & vip.persianss.ir


120 INDEX

Hyperdiploid mutations, myeloma, acute myeloid leukaemia, 41 International prognostic index see also Renal disease; Renal
62–63 flow cytometry, 21 (IPI), 61 failure
Hypereosinophilic syndrome, 7 non-Hodgkin’s lymphoma, 61 International Prognostic Scoring Klebsiella spp., 86
Hyperhomocysteinaemia, 27 Immunosuppression System for Primary MDS Knee damage, haemophilia A, 72f
Hyperkalaemia, 85 allogeneic stem cell (IPSS), 50–51, 51t
Hypersensitivity reactions, 7 transplantation, 56 International sensitivity index
Hypersplenism, 11 see also Allogeneic stem cell (ISI), 81 L
Hyperviscosity syndrome, 21, transplantation Intracranial bleeding
63–64 antibiotic treatment, 87, 87t haemophilia A, 72 Laboratory haematology
Hypervolaemia, 11 aplastic anaemia, 52–53 thrombocytopenia, 68 blood and bone marrow, 18–19
Hypnosis, 95 bone marrow aspiration, 106 Intrinsic factor (IF), megaloblastic blood films see Blood films
Hypocalcaemia, 85 and chemotherapy, 86 anaemia, 24–25 coagulation and acute phase
Hypocellular myelodysplastic chronic lymphocytic leukaemia, Intrinsic pathway, platelets, 12–13, response, 20–21
syndrome, 52–53 46f 13f Lactate dehydrogenase (LDH), 61
Hypochromia, 22 depressed cell-mediated Iron Leishmania donovani, 99
Hypofibrinogenaemia, 75 immunity, 87 definition/role, 24 Leishmann stain, 98
Hypogammaglobulinaemia, 46, 87 infection types, 86–87 malabsorption, 24–25 Lenalidomide, 55
chronic lymphocytic leukaemia, neutropenia, 87, 87t normal cycle, 24f myelofibrosis, 67
47 prevention of infection, 87 oral, failure to respond to, 25t myeloma, 63
immunosuppression, 87 treatment of infection, 87 overload, 85 Leucocytosis, 6–7, 7t, 97
Hyposplenism, 10–11, 11f, 11t see also Infection reuse of, 5 Leucopenia
Hypothermia, 71, 85 Infection Iron chelation therapy hypersplenism, 11
Hypoxia-inducible factor (HIF), 4 and anaemia of chronic disease, aplastic anaemia, 53 myelodysplastic syndromes, 50
36f myelodysplastic syndromes, 51 symptoms attributable to, 14
aplastic anaemia, 52 thalassaemias, 33 Leukaemia
I bacterial, 86, 97 Iron deficiency acute lymphoblastic see Acute
blood transfusion, 84 anaemia see Iron deficiency lymphoblastic leukaemia
Ibrutinib, 47 chronic lymphocytic leukaemia, anaemia (ALL)
Idarubicin, 54 47 characteristics, 24 acute monocytic, 40f
Idiopathic aplastic anaemia, 52 fungal, 86–87 correction, 25 acute myeloid see Acute myeloid
Idiopathic erythrocytosis, 65 history taking, 14 in elderly people, 24, 92 leukaemia (AML)
Idiopathic venous thrombosis, 81 HIV/AIDS, 97 erythropoiesis, 64 adult T-cell leukaemia
Ileum abnormalities, vitamin B12 hookworm, 99 hookworm infection, 99 lymphoma, 39f, 49
deficiency, 27 immunosuppression, 86–87 identification in pregnancy, 88 aetiology, 38–39
Imaging lymphadenopathy secondary to, in infancy, 91 annual causes of death, 38f
computed tomography (CT) 16 unexplained, 16 anti-leukaemic drugs, 54
scan, 59, 61 lymphocytosis, 9 Iron deficiency anaemia blood count, 18
magnetic resonance imaging, 59, mononucleosis, 97, 97f and anaemia of chronic disease, chronic lymphocytic see Chronic
61 neutropenia, 7 37t lymphocytic leukaemia
positron emission tomography parasitic, 7, 98–99 causes, 24–25 (CLL)
(PET) scans, 59, 61 prevention, 87 clinical features, 24–25 chronic myeloid see Chronic
Imatinib red cell transfusion confirmatory tests, 25 myeloid leukaemia (CML)
chronic myeloid leukaemia, 45, complications, 84 diagnosis, 25 classification, 39
55, 93 systemic disease, 97 investigation of underlying definition, 38
hypereosinophilic syndrome, 7 systemic enquiry, 15 cause, 25 dyspnoea in, 95
Immune antibodies, 82–83 treatment, 87 management, 25 gene therapy, 103
Immune neutropenia, 7 viral see Viral infection Isoantibodies, 30–31 genetic abnormalities, 38–39
Immune thrombocytopenia (ITP), Infertility, chemotherapy Isoimmune haemolytic anaemia, hairy cell, 48
68–69, 68t, 69f treatment, 54 30–31 history taking, 14
chronic lymphocytic leukaemia, Inflammation, systemic enquiry, 15 immunophenotype, cells, 21
47
haematopoietic growth factor
Inguinal nodes, examination, 16
Inherited blood disorders
J immunosuppression, 86
incidence, 38
therapy, 55 connective tissue disease, 71 minimal residual disease, 21
Janus kinase 2 (JAK2) gene
and HIV/AIDS, 97 in elderly people, 92–93 predisposing factors, 39
mutations
paediatric haematology, 91 examples, 15t prolymphocytic, 48
essential thrombocythaemia, 66
in pregnancy, 88–89 familial thrombophilia, 78 social history, 15
myelofibrosis, 67
systemic disease, 96 haemolytic anaemia, 29 T-cell large granular lymphocyte,
polycythaemia, 64
see also Thrombocytopenia platelet function, 70–71 48–49
Janus kinase 2 (JAK2) inhibitors
Immunochemistry, 102 sickle cell syndromes, 34f Leukapheresis, peripheral blood
myelofibrosis, 67
Immunodeficiency, in chronic vascular purpuras, 71 stem cells harvested by, 56,
myeloproliferative disorders,
lymphocytic leukaemia, 46 see also Genetic abnormalities; 57f
55
Immunoglobulins Haemophilia Leukocytes, listed, 6
polycythaemia, 65
acute phase response, 20 Inhibitory receptors, 9 see also White cell count (WBC)
Joints
basic structure, 9f Interference phase microscopy, Light microscopy, spleen, 11f
bleeding into, 14
and B-lymphocytes, 9 splenic function Light scattering, cell counting and
examination, 16
intravenous, 30, 89 quantitation, 11 sizing, 18
haemophilia, effects on, 72
thrombocytopenia, 69 Interferon alfa, 55 Lipid bilayer, red cells, 4
transfusion, 85, 87 essential thrombocythaemia, 66 Liver disease
Immunological tolerance, natural, hairy cell leukaemia, 48 K and alcohol, 96
9 polycythaemia, 64–65 coagulation disorders, 20, 77
Immunophenotyping Interleukin 5, eosinophils, 7 Kaolin cephalin clotting time hepatitis see Hepatitis
acute lymphoblastic leukaemia, International normalised ratio (KCCT), 20 protein C and S deficiencies,
42 (INR), 20, 80–81 Kidney enlargement, 17 78–79

tahir99-VRG & vip.persianss.ir


Index 121

sickle cell anaemia, 34 poorly differentiated large cell, Marginal zone, spleen, 10 Monocyte colony-stimulating
and thrombophilia, 78 46 Marginal zone lymphoma, 61 factor (M-CSF), 7
Liver function tests, 53 T-cell leukaemic see Adult T-cell Massive cervical lymphadenopathy, Monocytes, 7
Local anaesthesia, bone marrow leukaemia lymphoma 16f Monocytopenia, 7, 48
aspiration, 106 Lymphopenia, 86 Mast cells, 7 Mononuclear phagocyte system, 7
Low molecular weight (LMW) May–Grünwald–Giemsa (MGG) Mononucleosis, infectious, 97
heparin, 79–80, 89, 102 stain, 18–19 Monospot test, mononucleosis, 97
Lumps, examination, 16 M MDR1 (multi-drug resistance M-proteins, myeloma, 62
Lymph, location of lymphocytes gene), 54 MRI scans see Magnetic resonance
in, 8 Macrocytic anaemias, 22, 25, 27 Mean cell haemoglobin (MCH), imaging (MRI)
Lymphadenopathy Macrocytosis, alcohol misuse, 15, 22, 25, 36–37 Mucosa-associated lymphoid type
chronic lymphocytic leukaemia, 96 Mean cell haemoglobin (MALT) lymphoma, 60–61
46 Macrophages, 7, 10 concentration (MCHC), 22 Multi-drug resistance, 54, 98
defined, 16 Magnetic resonance imaging Mean cell volume (MCV), 22, 25, Multimer analysis, Von Willebrand
history taking, 14 (MRI), 59, 61 36–37 disease, 74f, 75
Hodgkin’s lymphoma, 48f Malabsorption Mean corpuscular haemoglobin Multiple myeloma see Myeloma
infection, 16 folate, 27 (MCH), 32 Mutations see Genetic
massive cervical, 16f iron, 24–25 Mean corpuscular volume (MCV), abnormalities
mediastinal, in HL, 48f vitamin K, 77 32 MYC oncogene, non-Hodgkin’s
non-Hodgkin’s lymphoma, 60 Malaria, 98–99 Medical history, 15 lymphoma, 60
Lymph nodes history taking, 15 Mefloquine, 99 Myelodysplastic syndromes (MDS)
biopsy, 58, 58f, 60–61 removal of malarial parasites, 10 Megakaryocytes, increased in and acute myeloid leukaemia,
examination, 16–17 Malignant disease thrombocytopenia, 69 39, 50
location of lymphocytes in, 8 chemotherapy see Megaloblastic anaemia and aplastic anaemia, 52–53
neck, 17f Chemotherapy B12 deficiency, 26–27 classification, 50
Lymphocyte predominant nodular colon cancer, 24f bone marrow aspirate in, 26f clinical features, 50
Hodgkin’s lymphoma, 58 current management model, 94f clinical syndromes, 26–27 diagnosis, 50
Lymphocytes, 8–9 in elderly people see under diagnosis, 27 in elderly people, 92
atypical, infectious Elderly people folate deficiency, 26–27 epigenetic therapies, 55
mononucleosis, 97f end of life care, 95 in pregnancy, 88 genetic factors, 50
B-lymphocytes see hypercoagulability in, 96 treatment, 27 high-risk, 51
B-lymphocytes lymphocytosis, 9 Melphalan, 54, 63 hypocellular, 52–53
changes in disease, 9 median ages of presentation, 93f ‘Memory’ B-cells, 9 low-risk, 51
count, 9, 46 microarrays/gene profiling, 100 Menstruation, iron deficiency, morphology, 50
depletion, 56 microenvironment, targeting, 24–25 and myelofibrosis, 67
donor lymphocyte infusions, 102 Mercaptopurine, 43, 54 platelet function disorders, 71
57 minimal residual disease, 21, 101 Merozoites, 98 prognosis, 50–51
dysfunction, 86 molecular biology applications, Metabolism, red cell see Red cell treatment, 51
hairy cell leukaemia, 48f 101 metabolism Myelofibrosis
Hodgkin’s lymphoma, 58 non-pain symptoms, control, Methicillin-resistant Staphylococcus characteristics, 66–67
mature, in blood, 8f 94–95 aureus (MRSA), 86 clinical features, 67
mononucleosis, infectious, 97 pain management, 14, 94 Methotrexate, 43, 54 diagnosis, 67
natural killer (NK) cells, 9 palliative treatment see Palliative Methylene blue stain, blood film, essential thrombocythaemia,
T-lymphocytes see treatment 18–19 progression from, 66
T-lymphocytes prostatic carcinoma, bone Microangiopathic haemolytic management, 67
see also Chronic lymphocytic marrow invasion, 97f anaemia (MAHA), 31, 31f polycythaemia, 64–65
leukaemia (CLL) psychosocial oncology, 95 Microarrays/gene profiling, 100 prognosis, 67
Lymphocytosis rectal, 16 Microcytic anaemias, 22 ‘Myeloid’ antigens, 41
chronic lymphocytic leukaemia, renal carcinoma, 96 Microcytosis, 22 Myeloma
46, 93 secondary, mediastinal Microthrombus formation, in DIC, asymptomatic (‘smouldering’),
common causes, 9t irradiation, 59 76 62
infection, 9 staging Minimal residual disease (MRD), biology, 62
Lymphoid follicles, splenic chronic lymphocytic haematological malignancy, bisphosphonate, 63, 94
structure, 10 leukaemia, 46 21, 101 clinical features, 62
Lymphoid organs, 8–9 Hodgkin’s lymphoma, 58–59, Mitoxantrone, 54 complications, 63
Lymphoma 59f Molecular biology diagnosis, 62–63
axillary lymphadenopathy, 60f myeloma, 62–63 acute lymphoblastic leukaemia, humoral immunity defects, 86
B-cell, 55, 97 non-Hodgkin’s lymphoma, 43 hypogammaglobulinaemia, 87
Burkitt’s see Burkitt’s lymphoma 60–61 acute myeloid leukaemia, 41 management and outcome, 63
diffuse large B-cell, 61, 100f stem cell transplantation, 101 application in haematology, 101 palliative treatment, 63, 95f
dyspnoea in, 95 symptoms attributable to, 14 blood tests, 82 platelet function disorders, 71
follicular, 61, 102, 102f and systemic disease, 96 DNA analysis techniques, proteasome inhibitors, 55
and HIV/AIDS, 97 systemic enquiry, 15 100–101 staging, 62–63
Hodgkin’s lymphoma see see also Hepatocellular cancer; Monoclonal antibodies (MoAbs), Waldenström’s
Hodgkin’s lymphoma (HL) Leukaemia; Lymphoma; 55 macroglobulinaemia, 63
mantle cell, 61 Myeloma Monoclonal B-cell lymphocytosis, Myeloperoxidase
marginal zone, 61 Malpighian bodies, 10 46 acute lymphoblastic leukaemia,
mucosa-associated lymphoid MALT (mucosa-associated in elderly people, 93 42
type, 61 lymphoid type) lymphoma, Monoclonal gammopathy of acute myeloid leukaemia, 40–41
non-Hodgkin’s see Non- 60–61 uncertain significance Myeloproliferative disorders
Hodgkin’s lymphoma Mantle cell lymphoma, 61 (MGUS) and acute myeloid leukaemia,
pain management, 94 Marfan syndrome, 71 elderly people, 93 39
peripheral T-cell, 61 Marginal pool, neutrophils, 6 myeloma, 62 basophilia, 7

tahir99-VRG & vip.persianss.ir


122 INDEX

chronic myeloid leukaemia, 44 management, 61 non-pain symptoms, control, see also Fresh frozen plasma
JAK2 inhibitors, 55 monoclonal antibodies, 55 94–95 (FFP)
and myelofibrosis, 67 nodal involvement, 60 pain management, 94 Plasmacytomas, 62
platelet function disorders, 71 proteasome inhibitors, 55 psychosocial oncology, 95 Plasma-derived factor VIII, 72–73
and thrombophilia, 79 Sjögren’s syndrome, 96 see also Malignant disease Plasma protein fraction (PPF), 85
Myocardial infarction, staging, 60–61 Pallor, anaemia, 22 Plasmin, 13
thrombolytic therapy, 81 see also Hodgkin’s lymphoma Pancytopenia Plasmodium falciparum, 98–99
(HL); Lymphoma aplastic anaemia, 52 Plasmodium malariae, 98
Nonsteroidal anti-inflammatory chronic lymphocytic leukaemia, Plasmodium ovale, 98
N drugs (NSAIDs), 94 47 Plasmodium vivax, 98
Normochromic anaemias, 22 hairy cell leukaemia, 48 Plasticity concept, 2
Natural killer (NK) cells, 9 Normocytic anaemias, 22 hypersplenism, 11 Platelet-derived growth factor
Naturally occurring antibodies, 82 Normocytosis, 22 and leukocytes, 7 (PDGF), 66
Nausea and vomiting, malignant Nose bleeds, thrombocytopenia, myelodysplastic syndromes, 52f Platelet function disorders
disease, 95 14 non-Hodgkin’s lymphoma, 60 acquired, 70–71
Neck, lymph nodes, 17f Nucleoside analogues, hairy cell Pappenheimer (siderotic) granules, causes of abnormal function, 71t
Neisseria meningitidis, 11 leukaemia, 48 11 essential thrombocythaemia, 66
Neonates/neonatal disorders see Paracetamol, 94 history taking, 14
Newborns
Neural tube defects, 88
O Parallel sequencing, 101
Paraproteins, myeloma, 62
inherited, 70–71
vascular purpuras, 71
Neutropenia Parasitic infection, 7 Platelet function instruments, 70
Observation of patient, 16
antibiotic treatment, 87t malaria, 98–99 Platelets
Ofatumumab, 55
aspergillosis, 86f Paroxysmal nocturnal in acquired coagulation
Oligonucleotide microarrays, 100
bacterial infection, 86 haemoglobinuria (PNH) disorders, 76–77
Ondansetron, 95
growth factors, 87 aplastic anaemia, 53 activation, 10, 12
Opiate analgesia, 94
haematopoietic growth factor haemolytic anaemia, 31 adhesion, 12
Organelles, platelets, 12
therapy, 55 Partial exchange transfusion, 91 role of vWF in, 74f
Osmotic fragility, hereditary
hairy cell leukaemia, 48 Partial thromboplastin time with aggregation, 12, 70f
spherocytosis, 28f
history taking, 14 kaolin (PTTK), 20 Von Willebrand disease, 75
Oxygen, transport, 4–5
immunosuppression, 86 Parvovirus, aplastic anaemia, 52 blood transfusion, 71, 85
Oxygen dissociation curve, 5f
infection, 7 Paul–Bunnel test, mononucleosis, chronic renal failure,
isolated, causes of, 7t 97 abnormalities in, 96
prevention of infection, 87 P Pentamidine, 87 clumping of, in
pyrexial neutropenic patient, 87 Pentostatin, hairy cell leukaemia, thrombocytopenia, 68f
systemic disease, 96 P50 level, haemoglobin and oxygen 48 count, 68–69
T-cell large granular lymphocyte transport, 5 Periarteriolar lymphatic sheath, 10 cytoplasm, 12
leukaemia, 48–49 Packed cell volume (PCV), 22, Periodic acid Schiff (PAS), acute destruction, in
Neutrophilia/neutrophil 90–91 lymphoblastic leukaemia, 42 thrombocytopenia, 68
leucocytosis, 6–7 Paediatric haematology Peripheral blood dilution, in thrombocytopenia,
Neutrophils, 6–7 acute lymphoblastic leukaemia, blood film, in AML, 39f 68
dysfunction, 86 43 myelodysplastic syndromes, 50 essential thrombocythaemia, 66
transfusion, 85 anaemia, 22 Peripheral blood stem cells hyposplenism, 11
Newborns bone marrow aspiration, general (PBSC), 56–57 intrinsic and extrinsic pathways,
anaemia of prematurity, 90 anaesthesia, 106 Peripheral T-cell lymphomas, 61 12–13, 13f
haemolytic disease of the congenital dyserythropoietic Peripheral venous cannulation, laboratory testing of function, 70
newborn, 90 anaemia, 91 105 loose plug, 12
iron deficiency in, 91 Diamond–Blackfan anaemia, 91 Perls’ stain, 19, 31f low count see
polycythaemia in, 90–91 haemoglobinopathies, 15–16 Pernicious anaemia, 26–27, 27f Thrombocytopenia
RhD prophylaxis in RhD- haemophilia, 15 ethnic origin of patient, 16 organelles, 12
negative mothers, 90 iron deficiency in infancy, 91 in pregnancy, 88 procoagulant action, 12
sites of blood production in, 2f neonatal disorders, 90–91 Petechial haemorrhage, pro-thrombinase complex, 12
thrombocytopenia, 91 normal values, 90 thrombocytopenia, 68 role, 12
venepuncture, 105 red cell aplasia in children and PET scans see Positron emission transfusion of, 27, 85
vitamin K deficiency, 77 adolescents, 91 tomography (PET) scans and vasoconstriction, 12
see also Paediatric haematology; sickle cell syndromes, 35 PFA-100 (platelet function Plummer–Vinson syndrome,
Pregnancy spleen, absent, 11 instrument), 70 24–25
Next-generation sequencing, 101 transient erythroblastopenia of P-glycoprotein, 54 Pneumocystis jiroveci (carinii)
Nicotinamide adenosine childhood, 91 Phagocytes, 6, 10 pneumonia, 86–87
dinucleotide phosphate umbilical cord blood (UCB) Phagocytosis, 6 PNH see Paroxysmal nocturnal
(NADPH), 5 transplantation, 57 Philadelphia (Ph) chromosome haemoglobinuria (PNH)
Nilotinib, 45, 55 venepuncture, 105 acute lymphoblastic leukaemia, Polycythaemia
Nitroblue tetrazolium test, chronic Pain 43 acute phase response, 20
granulomatous disease, 7 bone, in myeloma, 62 chronic myeloid leukaemia, apparent, 64–65
Non-haemolytic transfusion malignant disease, 14, 62 38–39, 44, 44f approach to patient with, 64
reactions, 84 management of in palliative Phospholipids, red cells, 4 clinical syndromes, 64–65
Non-Hodgkin’s lymphoma care, 94 Piperacillin, 87 essential thrombocythaemia,
aetiology, 60 splenic, 14, 67 Plasma progression from, 66
classification, 60 vascular-occlusive crises, 35 fibrinogen see Fibrinogen, in newborn, 90–91
clinical presentation, 60 Palliative treatment plasma renal disease, 96
diagnosis, 60–61 chemotherapy and radiotherapy, transfusion, 85 secondary, 65, 65f
high- and low-grade tumours, 60 94 viscosity, 21 smoking, 15
immunosuppression, 86 complementary therapy, 95 volume expansion, in pregnancy, Polycythaemia vera (PV), 64–66,
lymphocytosis, 9 myeloma, 63, 95f 88 65f

tahir99-VRG & vip.persianss.ir


Index 123

Polymerase chain reaction (PCR), and warfarin, 81 high concentration in Senile purpura, 71f
73, 90, 100–101 see also Activated partial polycythaemia, 64 Sepsis, 14, 76
real-time quantitative thromboplastin time intrasplenic pooling, in Septicaemia, 86
polymerase chain reaction (APTT) hypersplenism, 11 Sequestration, 34, 68
(RQ-PCR), 45 Pseudomonas aeruginosa, 86 membrane, 4f Serial analysis of gene expression
Polymorphism, prothrombin gene, Pseudopolycythaemia, 64–65 disorders, 29 (SAGE), 100
78 Pseudoxanthoma elasticum, 71 ‘normochromic normocytic’ Severe aplastic anaemia (SAA),
Positron emission tomography Psoas muscle bleed, haemophilia indices, 28 53
(PET) scans, 59, 61 A, 72f premature destruction, in Sézary syndrome, 49
Posterior iliac crest, 106f–107f Pulmonary arteriovenous hypersplenism, 11 Shock, blood transfusion, 84
Post-transfusion purpura, malformations (PAVMs), 71 removal, where unwanted, 10 Sickle cell syndromes
thrombocytopenia, 69 Pulmonary embolism (PE) renal disease, increased counselling, 35
Precursor lymphoid neoplasms, 42 main pulmonary trunk, 80f production in, 96 dactylitis, 34f
Prednisolone in pregnancy, 89 structure, 4 doubly heterozygous sickling
diffuse large B-cell lymphoma, thrombophilia, 78 transfusion, 84–85, 84t disorders, 35
61 Pure red cell aplasia (PRCA), 91 Red pulp, spleen, 10, 11f inheritance, 34f
follicular lymphoma, 61 Purine analogues, hairy cell Reed–Sternberg cells, lymph node pathophysiology, 34
myeloma, 63 leukaemia, 48 biopsy, in HL, 58f sickle cell anaemia (HbSS),
thrombocytopenia, 69 Purpura fulminans, 79 Regulatory molecules see Growth 34–35, 34f
warm autoimmune haemolytic Purpuric rash factors ethnic origin of patient, 16
anaemia, 30 myelodysplastic syndrome, 50f Renal disease, 96 in pregnancy, 88
see also Steroid treatment thrombocytopenia, 68, 69f Renal failure splenic atrophy, 10–11
Pre-eclampsia, 88–89 Pyruvate kinase (pk) deficiency, 29 Burr cells, 96f sickle cell trait (HbAS), 35
Pregnancy chronic, 71, 96 Side-effects
acute obstetric haemorrhage, 89t
anaemia in, 22, 88
Q in elderly people, 92
haemolytic transfusion reactions,
bone marrow harvesting, 107
cytotoxic drugs, 54
antenatal detection of genetic 84 interferon alfa, 48
Quantitative immunoassay, vWF
disorders, 101 platelet function disorders, 71 possible haematological, 15t
antigen, 75
coagulation abnormalities in, 89 Resistance, multi-drug, 54 Signal transduction, 3
Quinine, 99
common haematological malaria, 98 Silver impregnation, trephine
Quinolone antibiotic, 87
changes, 88f Respiratory burst, phagocytosis, 6 biopsy, 19
disseminated intravascular Reticulocytes Sjögren’s syndrome, 96
coagulation in, 88–89 R haemolytic anaemia, 28 Skin disorders
essential thrombocythaemia, 66 increased, in warm AIHA, 30f infection, 86f
haematological changes, 88 Radioimmunoassay, erythropoietin Reticulocytopenia, aplastic sickle cell anaemia, 34
HELLP syndrome, 89 estimation by, 64 anaemia, 52 Smear cells, chronic lymphocytic
sickle cell anaemia (HbSS), 35 Radioimmunotherapy, follicular Rhesus (Rh) blood group system, leukaemia, 46
thrombocytopenia in, 88–89 lymphoma, 61 82–83 Smears, marrow aspirate, 106
thrombophilia in, 79 Radiotherapy clinical practice, 85 Smoking, history taking, 15
see also Newborns; Paediatric chronic lymphocytic leukaemia, paediatric haematology, 90 Social history, 15
haematology 47 Rheumatoid arthritis, 37, 96 Southern blotting, factor VIII gene
Prematurity, anaemia of, 90 follicular lymphoma, 61 Richter syndrome, 46 inversion in haemophilia,
Prenatal diagnosis, 33, 35 Hodgkin’s lymphoma, 59 Ristocetin, failure to aggregate 73f
Procoagulant action, platelets, 12 myeloma, 63 with, 70 Spectrin, 4
Proguanil, malaria, 99 palliative, 94 Rituximab, 55 Spectrophotometer, automated
Prolymphocytes, 48 stem cell transplantation, 56–57 chronic lymphocytic leukaemia, haematology counters, 18
Prolymphocytic leukaemia (PLL), Rai system, Binet adaptation, 46 47 Spindle poisons, 54
48 Rashes, purpuric diffuse large B-cell lymphoma, Spleen
Prostatic carcinoma, bone marrow myelodysplastic syndrome, 50f 61 abnormal states, 10–11
invasion, 97f thrombocytopenia, 68, 69f follicular lymphoma, 61 absent, 10–11
Protein C Raynaud’s phenomenon, 30 prolymphocytic leukaemia, and B-lymphocytes, 9
coagulation regulation, 13 Real-time quantitative polymerase 48 enlargement, schematic view,
deficiency, familial chain reaction (RQ-PCR), 45 treatment advances, 102 17f
thrombophilia, 78–79 Recombinant activated protein C, warm autoimmune haemolytic examination, 17, 17f
Protein S acquired coagulation anaemia, 30 function, 10
coagulation regulation, 13 disorders, 76 Rivaroxaban, 81, 102–103 Howell–Jolly bodies, 10–11
deficiency, familial Recombinant vWF, 75 Romanowsky’s stain, 18–19, 28 light microscopy, 11f
thrombophilia, 78–79 Rectal examination, 16 Ruxolitinib, 55, 67 location of lymphocytes in, 8
Proteolytic enzymes, 12 Red cell aplasia, 91 red and white pulp, 10, 11f
Proteasome inhibitors, 55 Red cell distribution width (RDW), structure, 10
Prothrombin, 12–13 25 S Splenectomy, 10–11
Pro-thrombinase complex, Red cell indices, 22 chronic lymphocytic leukaemia,
platelets, 12 Red cell metabolism, 5 Schilling test, vitamin B12 47
Prothrombin complex concentrate, abnormalities, 29 absorption, 27 hairy cell leukaemia, 48
77 schematic diagram, 29f Schizonts, 98 hereditary elliptocytosis, 29
Prothrombin G20210A, familial Red cells, 4–5 Scintigraphy, 11 pyruvate kinase (pk) deficiency,
thrombophilia, 78 ageing and death, 5 Screening strategies, sickle cell 29
Prothrombin time (PT) blood count, 18 syndromes, 35 thrombocytopenia, 69
disseminated intravascular characteristic biconcave shape, 4f SDS–PAGE multimer analysis, vW warm autoimmune haemolytic
coagulation, 76 enlarged (macrocytosis), 15, 24 factor, 74f anaemia, 30
haemostasis, 12–13 erythropoietin, 4 Sedation, bone marrow aspiration Splenic artery, 10
laboratory haematology, 20 haemoglobin and oxygen and trephine biopsy, Splenic cords, 10
vitamin K deficiency, 77 transport, 5 106–107 Splenic sinuses, 10

tahir99-VRG & vip.persianss.ir


124 INDEX

Splenomegaly Submicroscopic mutations, alloimmune, 91 Transfusion associated circulation


chronic lymphocytic leukaemia, leukaemia, 38 autoimmune, 46 overload (TACO), 84
46 Sudan black stain, 40–42 bleeding, 14, 68 Transfusion-related acute lung
chronic myeloid leukaemia, 44 Superior vena cava syndrome, 60 bone marrow aspiration, 106 injury (TRALI), 84
common causes, 17t Supplementation causes, 68 Transient erythroblastopenia of
hypersplenism, 11 folic acid, 27 clinical presentation, 68 childhood, 91
myelodysplastic syndromes, 50 iron, 88 clinical syndromes, 68–69 Treatment of conditions
myelofibrosis, 66–67, 67f vitamin K, 77, 81 heparin use, 80 advances in, 102–103
pain in, 14 Supportive care hypersplenism, 11 bleeding, in haemophilia A,
polycythaemia, 64 acute myeloid leukaemia, 41 incidental (gestational), 88 72–73
prolymphocytic leukaemia, 48 aplastic anaemia, 53 myelodysplastic syndromes, 50 chronic lymphocytic leukaemia,
thrombocytopenia, 68 haematopoietic growth factor myeloma, 62 46–47, 47f
tropical splenomegaly syndrome, therapy, 55 in newborn, 91 chronic myeloid leukaemia, 45,
99 myelodysplastic syndromes, 51 in pregnancy, 88–89 55
Staging of malignant disease Supravital stain, blood film, 18–19 symptoms attributable to, 14 infection, 87
chronic lymphocytic leukaemia, Surgery T-cell large granular lymphocyte megaloblastic anaemia, 27
46 sickle cell anaemia (HbSS), 35 leukaemia, 48–49 myelodysplastic syndromes, 51
Hodgkin’s lymphoma, 58–59, splenectomy see Splenectomy see also Immune viral, 73
59f 5q-syndrome, 50 thrombocytopenia (ITP) see also Chemotherapy; Drug
myeloma, 62–63 Syngeneic stem cell Thrombocytosis, systemic enquiry, treatment; Surgery
non-Hodgkin’s lymphoma, transplantation, and 15 Trephine biopsy, bone marrow
60–61 allogeneic SCT, 56 Thromboembolism, anticoagulant acute lymphoblastic leukaemia,
Stains and dyes, blood films, 18–19 Syringe drivers, continuous drug therapy, 89 42
see also specific stains, such as infusions, 95 Thrombolytic therapy, 81 acute myeloid leukaemia, 40
Sudan black stain Systemic disease Thrombomodulin inhibitors, 102 aplastic anaemia, 52–53
Staphylococcus epidermidis, 86 basophilia, 7 Thrombophilia chronic lymphocytic leukaemia,
Stem cell hierarchy, blood count, 18 acquired forms, 79 46
haematopoiesis, 2, 3f connective tissue, 71, 96 acute venous thrombosis, 79 examination procedure, 19
Stem cell mobilisation, 55 infection, 97 familial, 78–79 hairy cell leukaemia, 48
Stem cell transplantation (SCT) liver, 96 genetic counselling, 79 human T-cell leukaemic
acute myeloid leukaemia, 41 malignancy, 96 patients to be investigated, 78 lymphoma, 39f
allogeneic see Allogeneic stem non-Hodgkin’s lymphoma, 60 in pregnancy, 79 myelofibrosis, 67f
cell transplantation renal, 96 Thromboplastin, prothrombin polycythaemia, 64
aplastic anaemia, 53 and thrombophilia, 78 time test, 20 procedure, 107
autologous see Autologous stem see also Infection; Liver disease; Thrombopoeitin receptor (TPO-R), staining, 19
cell transplantation Malignant disease second generation agonists, and structure of bone marrow, 2
chronic myeloid leukaemia, 45 Systemic lupus erythematosus 69 systemic disease, 96
graft-versus-host disease, 56, 57f (SLE), 79, 96 Thrombosis thrombocytopenia, 69
haematological malignancy, 101 in elderly people, 92, 92f uses, 19
immunosuppression, 86–87 large vessel, in DIC, 76 Tricyclic antidepressants, pain
leukapheresis, peripheral blood T polycythaemia, risk in, 64 management, 94
stem cells harvested by, 56, Thrombotic thrombocytopenic Trilaminar platelet membrane, 12
57f Tazobactam, 87 purpura (TTP), 31 Trophozoites, 98–99
non-myeloablative (‘reduced T-cell large granular lymphocyte Thymus Tropical splenomegaly syndrome,
intensity’) allogeneic, 57 leukaemia (T-LGL), 48–49 destruction of T-lymphocytes by, 99
sickle cell anaemia (HbSS), 35 T-cell receptor (TCR), 8 8 Twin (allogeneic and syngeneic)
syngeneic, 56 Tear-drop poikilocytes, location of lymphocytes in, 8 stem cell transplantation, 56
twin (allogeneic and syngeneic), myelofibrosis, 67 Tissue factor pathway inhibitor Tyrosine kinase inhibitors (TKIs),
56 Telangiectasia, 71 (TFPI), 13 55
umbilical cord blood, 57 Telomeres, in aplastic anaemia, 52 Tissue hypoxia, anaemia, 22 acute myeloid leukaemia, 55
see also Blood transfusion Thalassaemias Tissue necrosis, haemorrhagic, 76 chronic myeloid leukaemia, 45
Stercobilinogen, faecal, 28 classification, 32 Tissue plasminogen activator Tyrosine kinase receptor gene
Steroid treatment clinical syndromes, 32–33 (t-PA), 13, 81 mutation, acute myeloid
chronic lymphocytic leukaemia, gene therapy, 103 T-lymphocytes, 8 leukaemia, 41
47 prenatal diagnosis, 33 acute lymphoblastic leukaemia,
myeloma, 63
thrombocytopenia, 69
spleen, absent, 11
α-Thalassaemias, 32
42
adult T-cell leukaemia
U
warm autoimmune haemolytic β-Thalassaemia intermedia, 33 lymphoma, 49
Ulceration, cold autoimmune
anaemia, 30 β-Thalassaemia major, 32–33 interaction with antigen-
haemolytic anaemia, 30
see also Prednisolone β-Thalassaemia trait (minor), 33 presenting cells, 8f
Ultrastructural studies, 2
Stomach abnormalities, vitamin Thalidomide, 55 removal in stem cell
Umbilical cord blood (UCB)
B12 deficiency, 27 myelofibrosis, 67 transplantation, 56
transplantation, 57
Storage pool disorders, 70 myeloma, 63 splenic structure, 10
Unfractionated heparin, 80, 89
Streptococcus faecalis, skin Third-generation recombinant Topoisomerase poisons/inhibitors,
Uraemia, 71
infection, 86f factor VIII, 72–73 54
Urobilin, 5
Streptococcus pneumoniae, 11 3-D confocal imaging, 102 Tourniquets, 104–105
Urobilinogen, 5, 28
Streptokinase, 81 Thrombin, 12–13 Tranexamic acid, bleeding, 73
Urokinase, 81
Stromal cells, 2 inhibitors, 102–103 Transcranial Doppler
Stylet, bone marrow aspiration Thrombocytes see Platelets ultrasonography, sickle cell
and trephine biopsy, Thrombocytopenia syndromes, 35 V
106–107 acute lymphoblastic leukaemia, Transcription factors, 2
‘Subacute combined degeneration’, 42 Transforming growth factor-β, 66 Variant Creutzfeldt-Jakob disease
26 alcohol misuse, 96 Transfusion see Blood transfusion (vCJD), 84

tahir99-VRG & vip.persianss.ir


Index 125

Varicella zoster, 87 Viral infection diagnosis, 74f Warm autoimmune haemolytic


Vascular-occlusive crises, sickle cell adult T-cell leukaemia laboratory diagnosis, 75 anaemia, 30, 30f
anaemia, 34–35 lymphoma, 49 management, 75 White cell count (WBC)
Vascular purpuras, 71 aplastic anaemia, 52 types 1 and 2, 74 acute myeloid leukaemia, 40
Velocimetry, fetal middle cerebral haemophilia A, 73 Von Willebrand factor (vWF) chronic myeloid leukaemia, 44
artery, 90 hepatitis see Hepatitis platelet function disorders, laboratory haematology, 18
Venepuncture, 104–105 immunosuppression, 86–87 70 reference values, 90
Venesection, polycythaemia, 65 systemic disease, 97 range of levels, 74 see also Leukocytes
Venous access, 105 thrombocytopenia, 68 role in platelet adhesion, 74f White cells, low count see
Venous thrombosis Visceral leishmaniasis (kala-azar), thrombotic thrombocytopenic Leucopenia
acute, 79 99 purpura, 31 White pulp, spleen, 10, 11f
in elderly people, 92f Vitamin B12 deficiency Voriconazole, 87 World Health Organization
Factor V Leiden (FVR506Q), 78 intramuscular injection, 24–25 (WHO) classification
idiopathic, 81 megaloblastic anaemia, 26–27 systems
malignant disease, 96 in pregnancy, 88 W acute lymphoblastic leukaemia,
see also Deep vein thrombosis Vitamin K 42
(DVT) deficiency, 76–77 Waldenström’s acute myeloid leukaemia, 40
Vertebroplasty, myeloma, 63 intravenous, 81 macroglobulinaemia, 63 Hodgkin’s lymphoma, 58
Very severe aplastic anaemia platelets, 12 Warfarin non-Hodgkin’s lymphoma, 60
(VSAA), 53 supplementation, 77, 81, 92 indications for use, 80–81 Wright’s stain, 18–19
Vinblastine, 54 and warfarin, 76, 80f method of action, 80
Hodgkin’s lymphoma, 59 Vitamin K derived clotting factor, in pregnancy, 89
Vincristine, 54 73 protein C and S deficiencies, Z
diffuse large B-cell lymphoma, Von Willebrand disease (vWD) 78–79
61 classification, 74 side-effects, 79 ZAP-70 (signalling molecule), 46
follicular lymphoma, 61 clinical features, 74–75 and vitamin K, 76, 80f Zidovudine, 49

tahir99-VRG & vip.persianss.ir

Вам также может понравиться