Practical Obstetric Hematology PDF
Practical Obstetric Hematology PDF
Practical Obstetric Hematology PDF
Practical Obstetric
Hematology
Obstetric Prelims 20/10/05 3:09 pm Page ii
Obstetric Prelims 20/10/05 3:09 pm Page iii
Practical Obstetric
Hematology
This book contains information obtained from authentic and highly regarded sources. While all reasonable
efforts have been made to publish reliable data and information, neither the author[s] nor the publisher can
accept any legal responsibility or liability for any errors or omissions that may be made. The publishers wish
to make clear that any views or opinions expressed in this book by individual editors, authors or contributors
are personal to them and do not necessarily reflect the views/opinions of the publishers. The information or
guidance contained in this book is intended for use by medical, scientific or health-care professionals and is
provided strictly as a supplement to the medical or other professional’s own judgement, their knowledge of
the patient’s medical history, relevant manufacturer’s instructions and the appropriate best practice guide-
lines. Because of the rapid advances in medical science, any information or advice on dosages, procedures
or diagnoses should be independently verified. The reader is strongly urged to consult the drug companies’
printed instructions, and their websites, before administering any of the drugs recommended in this book.
This book does not indicate whether a particular treatment is appropriate or suitable for a particular indi-
vidual. Ultimately it is the sole responsibility of the medical professional to make his or her own professional
judgements, so as to advise and treat patients appropriately. The authors and publishers have also attempted
to trace the copyright holders of all material reproduced in this publication and apologize to copyright
holders if permission to publish in this form has not been obtained. If any copyright material has not been
acknowledged please write and let us know so we may rectify in any future reprint.
Except as permitted under U.S. Copyright Law, no part of this book may be reprinted, reproduced, transmit-
ted, or utilized in any form by any electronic, mechanical, or other means, now known or hereafter invented,
including photocopying, microfilming, and recording, or in any information storage or retrieval system,
without written permission from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copyright.
com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC), 222 Rosewood
Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides licenses and
registration for a variety of users. For organizations that have been granted a photocopy license by the CCC,
a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are used
only for identification and explanation without intent to infringe.
Visit the Taylor & Francis Web site at
http://www.taylorandfrancis.com
Contents
Preface vii
Glossary ix
Index 189
Obstetric Prelims 20/10/05 3:09 pm Page vi
Obstetric Prelims 20/10/05 3:09 pm Page vii
Preface
In the last 10 years there has been a rapid expansion in the clinical interaction
between hematologists and obstetricians. As a result there is an increasing
challenge to hematologists, obstetricians and midwifery staff to understand and
manage the clinical manifestations of a number of rapidly developing areas of
hemato-obstetric science.
This book is intended to serve practitioners involved in the management of
pregnancy, from trainees in obstetrics, hematology and vascular medicine to
general practitioners involved in the day-to-day management of maternity care, as
well as midwifery staff and specialists in hematology, obstetrics and vascular
medicine. The book is designed to assist the reader to diagnose and treat these
conditions, but also provides the reader with a user-friendly, but authoritative,
source of information on the pathophysiology of hemato-obstetric problems that
incorporates the best practice contained within internationally accepted
guidelines of obstetric and hematological care.
The book is intended to assist the reader rapidly assimilate the essential aspects
of the pathophysiology of these complex disorders and provide an aide memoir
to the management of these conditions. As well as a guide to the pitfalls associated
with investigation and treatment, each chapter has a similar format and employs
extensive use of tables and figures.
The first chapter covers the physiological changes in hematological indices
caused by normal pregnancy. The second provides a succinct review of the basic
science of transfusion medicine and appropriate blood component usage as it
relates to obstetric practice. The subsequent chapters deal with the impact,
diagnosis and management of adverse transfusion events, hemolytic disease of the
newborn, anemia, malignant hematology, bleeding disorders, antithrombotic
therapy, thrombophilia, thrombotic disorders and red cell disorders in
pregnancy.
To assist the reader, each of these chapters conforms to a similar style that
includes: pathophysiology; presentation; differential diagnosis; diagnostic tests;
diagnostic difficulties; maternal complications, fetal complications; and potential
management complications. Each chapter also includes tables summarizing the
key points in diagnosis and management, as well as information on the likely
impact of new therapies and a preview of the developing issues relating to each
condition.
Obstetric Prelims 20/10/05 3:09 pm Page viii
Obstetric Prelims 20/10/05 3:09 pm Page ix
Glossary
ACCP American College of Chest Physicians
ACE Angiotensin-converting enzyme
ADAMTS A disintegrin and metalloproteinase with thrombospondin domains
ADP Adenosine diphosphate
AF Atrial fibrillation
AFLP Acute fatty liver of pregnancy
AIHA Autoimmune hemolytic anemia
AIN Autoimmune neutropenia
AIP Acute intermittent porphyria
AML Acute myeloid leukemia
APAS Antiphospholipid antibody syndrome
aPC Activated protein C
aPCR Activated protein C resistance
APS Antiphospholipid syndrome
APTT Activated partial thromboplastin time
Art T Arterial thrombosis
AST Aspartate Transaminase
AT Antithrombin
ATP Adenosine triphosphate
BMI Body mass index
CD Cluster of differentiation
CEP Congenital erythropoietic porphyria
CI Confidence interval
CIAO Cerebral ischemia of arterial origin
CMV Cytomegalovirus
CNS Central nervous system
CT Computerized tomography
CTG Cardiotocography
CVP Chorionic villus sampling
DDAVP Demopressin analog
DIC Disseminated intravascular coagulation
Obstetric Prelims 20/10/05 3:09 pm Page x
Glossary
2,3-DPG 2,3-diphosphoglycerate
dRVVT Dilute Russell viper venom test
DVT Deep vein thrombosis
ECG Electrocardiogram
EDTA Ethylenediaminetetraacetic acid
ELISA Enzyme-linked immunosorbent assay
EPCR Endothelial protein C receptor
Epo Erythropoietin
EPP Erythropoietic protoporphyria
ET Essential thrombocythemia
F Factor
FBC Full blood count
FDP Fibrin degradation products
FFP Fresh frozen plasma
FMH Feto-maternal hemorrhage
FPA Fibrinopeptide A
G6PDH Glucose-6-phosphate dehydrogenase
G-CSF Granulocyte colony-stimulating factor
GI Gastrointestinal
GPI Glycophosphatidylinositol
GVHD Graph-versus host disease
Hb Hemoglobin
HCY Homocysteine
HDN Hemolytic disease of the newborn
HELLP Hemolysis, elevated liver enzymes, and low platelets
HHV Human herpes virus
HIT Heparin-induce thrombocytopenia
HITTS HIT thrombosis syndrome
HLA Human leukocyte antigen
HPA Human platelet antigen
HPFH Hereditary persistence of fetal hemoglobin
HPLC High-performance liquid chromatography
HTLV Human T-cell leukemia/lymphoma virus
HUS Hemolytic uremic syndrome
ICH Intracerebral hemorrhage
x
Obstetric Prelims 20/10/05 3:09 pm Page xi
Glossary
Glossary
CHAPTER 1
Hematology
measurements in
pregnancy
Pregnancy results in significant changes in metabolism, fluid balance, organ
function and circulation, which are driven, in part, by estrogen and the presence
of the feto-placental unit. These dramatic changes influence a wide variety of
hematological parameters. A knowledge of these changes is essential when inter-
preting the results of hematological investigations to diagnose, or monitor,
illness in pregnancy.
Blood count
The maternal plasma volume begins to increase from around 6 weeks gestation,
but this is not accompanied by an immediate increase in red cell mass. The effect
of this is a dilutional anemia, which may be evident from the 7–8th week of gesta-
tion. Although the mechanism of this increase in blood volume is not fully
understood, it may be more marked in multiple pregnancies and occurs even
when iron stores are poor. The physiological purpose of this change may be to
reduce maternal blood viscosity, and improve delivery of oxygen and nutrients to
the fetus. The increase in plasma volume may reach 140% of non-pregnant
values and usually becomes maximal in the late second trimester. By this stage in
pregnancy, a 15–25% increase in red cell mass, driven by an increase in maternal
erythropoietin production, will also be present. These changes should not,
however, reduce the hemoglobin to <11g/dl in the first trimester or <10g/dl in
the second and third trimesters, and should result in a normochromic, normo-
cytic red blood cell pattern on the blood film. This ‘physiological anemia’ of
pregnancy does not worsen during the third trimester, which may reflect a reduc-
tion in maternal plasma volume and a stable red cell mass. Consequently, a rela-
tive increase in hemoglobin and hematocrit may be observed in the late third
trimester.
With normal pregnancy there may also be a rise in the mean corpuscular
volume (MCV), and a normal MCV does not, therefore, exclude iron deficiency.
Similarly, a modestly elevated MCV does not diagnose folate (or vitamin B12) defi-
ciency in pregnancy (Table 1.1).
1
Obstetric CH01 18/10/05 10:20 am Page 2
Hemoglobin Falls, but >11g/dl in the first trimester and >10g/dl in the second and third
trimesters
Mean corpuscular No change, rise or fall all possible
volume (MCV)
Platelets >80 × 109/l
Neutrophils Variable rise (earlier forms seen)
Vitamin B12 <50% fall with increasing gestation
Serum folate <30% fall with increasing gestation
Red cell folate <20% rise, or no change with increasing gestation
Homocysteine No change, or fall
Vitamin B6 <20% fall with increasing gestation
Fe Variable fall with increasing gestation
Ferritin Variable fall; can be reduced to near non-pregnant iron-deficient ranges
Total iron-binding Variable rise with increasing gestation
capacity (TIBC)
Transferrin receptors (tfR) Increase after 20/40 weeks gestation
2
Obstetric CH01 18/10/05 10:20 am Page 3
Hematinic assessment
Normal pregnancy is associated with a progressive fall in serum iron levels and an
increase in serum total iron-binding capacity (TIBC: see Chapter 3). The TIBC
reflects the amount of the iron transport protein (transferrin) that is saturated
with iron; the level of TIBC fluctuates widely and is also influenced by recent
iron ingestion. A fall in serum ferritin levels also occurs in normal pregnancy,
although very low ferritin levels (<12ug/l) will still reliably indicate iron defi-
ciency. A number of newer tests of iron deficiency have been evaluated out-with
pregnancy. These include the plasma level of free transferrin receptors (tfR) and
the plasma level of free protoporphyrin. In the latter case, iron deficiency leads
to a reduction in the incorporation of iron into the prophyrin ring that forms
the heme moiety of hemoglobin. The level of the ‘unused’ protoporphyrin is
therefore a measure of deficiency. However, an increase in free protoporphyrin
occurs in normal pregnancy. The tfR is a transmembrane protein that mediates
delivery of iron to the developing erythroblast and is shed from the maturing red
cell. The serum tfr (stfR) level relates to the degree of iron deficiency and
increasing values precede changes in red cell morphology. stfR has been used
successfully to discriminate iron deficiency from the anemia of chronic disease,
as it does not increase in response to inflammation. However, stfR levels have
been shown to increase with gestation, returning to non-pregnant values 3
months postpartum. Although this could relate to undiagnosed iron deficiency,
it seems more likely to be due to an increase in maternal erythropoiesis.
Vitamin B12 levels may fall by 30–50% during pregnancy, with evidence of
reduced levels in the first trimester in some individuals. This occurs despite a diet
adequate in vitamin B12 and is likely to be due to a combination of increasing mater-
nal plasma volume, hormonal changes, increasing maternal requirement and
vitamin B12 transfer to the fetus. Overall, the bulk of current evidence suggests that
serum or plasma homocysteine falls with increasing gestation in most subjects. The
impact of folate repletion on these changes, however, requires further investigation.
Although vitamin B6 indices seem to decrease during pregnancy, it is not clear
whether normal pregnancy is associated with a vitamin-B6-deficient state or not,
and, even if it is, whether this has any clinical significance or not. In non-supple-
mented pregnancies there is a progressive fall in serum folate with increasing
gestation and either a rise, or no change, in red cell folate can both occur.
Hemostasis
Pregnancy is associated with significant changes in hemostasis. An understanding of
the normal hemostatic system and these physiological changes is essential to the diag-
nosis and management of bleeding and thrombotic disorders. It is also necessary to
understand the limitations of the D-dimer algorithms that are used in the screening
of non-pregnant subjects suspected of venous thromboembolism (VTE). Further-
more, it may provide an insight into the mechanisms of not only gestational venous
3
Obstetric CH01 18/10/05 10:20 am Page 4
thrombosis, but also pregnancy loss and complications such as pre-eclampsia, which
have recently been associated with both acquired and heritable thrombophilia.
Pregnancy results in an upregulation in the maternal coagulation cascade,
which leads to an overall increase in thrombin generation. This maternal alteration
in coagulation is clearly a physiological preparation for delivery. As with many bio-
logical factors, this change in coagulation factors may relate to alterations in:
hormone-influenced synthesis; the volume of distribution; lipid interaction; or
catabolism. The progressive increase in maternal estrogen levels may alter the
uptake of coagulation proteins into the uterus and may also result in an increase in
the uterine generation of tissue factor. It is possible that both placental and mater-
nal thrombin have an important influence on cellular growth, and thrombin gen-
eration results in fibrin formation, which is essential for placental implantation.
The ‘extrinsic’ initiation of coagulation with a combination of tissue factor (TF), Factor (F) VII and activated FVII
(FVIIa) is shown. Solid lines indicate activation and dotted lines indicate inhibition; the principal pathway is
shown in bold type. The FVII–FVlla–TF complex activates FX (X) to FXa (Xa), as well as FIX (IX) to FIXa (XIa):
this results in the activation of prothrombin (PT) in a prothrombinase complex of calcium (Ca2+), phospholipid
(Plipid) and activated FV (Va), resulting in a burst of thrombin. This burst of thrombin activates FV (V) to FVa
(Va), FVIII (VIII) to FVIIIa (VIIIa) and FXI (XI) to FXIa (XIa). The combination of thrombin’s action and activation of
the ‘intrinsic’ coagulation pathway by exposure to damaged endothelium (via activation of FXII (XII) to FXIIa
(XIIa), results in activation of FXI (XI); this leads to activation of FIX (IX). The further activation of FX (X) in the
Tenase complex of FIXa (IXa), FVIIIa (VIIIa), Plipid and Ca2+ amplifies the coagulation initiated by the thrombin
burst. Thrombin also activates FXIII (XIII), which cross-links and stabilizes formed fibrin.
4
Obstetric CH01 18/10/05 10:20 am Page 5
INTRINSIC
XII XIIa
XI
Ca2+
IX XIa
IXa
V
5
X VIIIa
TF/FVII/FVIIa
EXTRINSIC
Ca2+/Plipid PT Va
Tenase Fibrinogen
Xa
THROMBIN
Ca2+/Plipid burst Fibrin
Prothrombinase
and around 1% circulates as FVIIa, the activated form. Both FVIIc and FVIIa are
bound to TF expressed on the vessel wall and inflammatory cells, thereby localizing
the hemostatic response to the site of injury. Once FVIIc binds to TF it can be acti-
vated by TF itself. This complex can convert further FIXc to FIXa, FXc to FXa, and
FVc to FVa. Formed FXa binds to FVa to form a complex (the prothrombinase
complex) with calcium on a phospholipid membrane surface. In the presence of
these phospholipids, further FVc is converted to FVa and prothrombin is activated
leading to a ‘thrombin burst’. As prothrombin is cleaved by FXa to form thrombin,
a fragment is released. This fragment is called prothrombin fragment 1+2 (F1+2)
and, measured in plasma, can be used as a measure of thrombin generation. The
thrombin burst can then convert fibrinogen to fibrin, and activate parts of the
intrinsic pathway (FVIIIc and FXIc), as well as FVc, FXIIIc (which leads to cross-
linking and stabilization of formed fibrin), the endothelium, platelets and mono-
cytes. As thrombin is also involved in anticoagulation and fibrinolysis (see below), it
has a central role in orchestrating the hemostatic response.
In the intrinsic pathway, the binding of FXIIc to a negatively charged surface
leads to a local increase in FXIIc concentration, which results in its autoactiva-
tion. FXIIa causes the conversion of pre-kallikrein to kallikrein, and activates
FXIc to FXIa. FXIa, as well as the TF–FVIIa complex, activates FIX to FIXa. Acti-
vated FIXa forms a complex (the Tenase complex) with FVIIIa, calcium and
phospholipids. This results in activation of FXc to FXa, which contributes to the
prothrombinase complex and further thrombin generation.
Thrombin converts soluble fibrinogen to an insoluble fibrin polymer, which
seals the site of injury and protects damaged tissue during wound healing. Fib-
rinogen activation generates soluble fibrin monomers, fibrinopeptide A and fib-
rinopeptide B. The resultant fibrin mesh is stabilized by cross-linking catalyzed by
thrombin-activated coagulation FXIIIa.
Coagulation control
There are a number of controls of coagulation (Figure 1.2). TF pathway
inhibitor (TFPI) is released constitutively from endothelial cells, and, in associ-
The principal points of physiological inhibition and downregulation of the coagulation cascade are shown. Solid
lines indicate a positive effect and dotted lines indicate inhibition. The main inhibitors are antithrombin (AT –
previously called antithrombin III). AT inhibits almost all of the coagulation factors, but its principal targets are
Factor (F) Xa and thrombin. The interaction of AT with thrombin results in the formation of the
thrombin–antithrombin (TAT) complex. Thrombin, when it binds to endothelial thrombomodulin (TM), loses its
procoagulant activity and activates protein C (PC) to activated protein C (aPC). aPC, with its cofactor protein S
(PS), cleaves activated FV (Va) and activated FVIII (VIIIa). PC can also be activated independently by the endothe-
lial protein C receptor (EPCR). Other inhibitors of coagulation include tissue factor pathway inhibitor (TFPI),
which rapidly inactivates the FVII–FVIIa–tissue factor (TF) complex. In addition, antitrypsin and C1-esterase
inhibitor are capable of inhibiting FXIa and FXIIa respectively. Ca2+, Calcium; Plipid, phospholipid.
6
Obstetric CH01 18/10/05 10:20 am Page 7
INTRINSIC
C 1-esterase Inhibitor
XII XIIa
XIIi
XI
2+
Ca
IX XIa Antitrypsin
TM
IXa aPC PC
(EPCR)
V +PS
7
TF/FVII/FVIIA
EXTRINSIC
Ca2+/Plipid PT Va Vi
Tenase Fibrinogen
Xa
AT THROMBIN
TFPI Ca2+/Plipid burst Fibrin
Xi
Prothrombinase
TAT
ation with FXa, is able to rapidly inactivate the TF–FVIIa complex. Other con-
trols include antithrombin, which can inhibit many activated clotting factors
including thrombin and FXa. By binding to antithrombin, thrombin forms a
stable thrombin–antithrombin (TAT) complex, which is rapidly cleared from the
circulation. Another regulatory mechanism is protein C, which circulates as an
inactive zymogen. To become activated it binds to the endothelial protein C
receptor (EPCR), or the transmembrane protein thrombomodulin, which is
expressed on endothelial cells. The formation of a thrombin–thrombomodulin
complex on the endothelial surface accelerates thrombin’s activation of the
natural anticoagulant protein C by around 20 000-fold. Formation of this
complex directly inhibits the capacity of thrombin to cleave fibrinogen and acti-
vate platelets. To function as an inhibitor, activated protein C (aPC) dissociates
from EPCR and binds to its cofactor protein S. Protein S has no enzymatic activ-
ity of its own, but acts as a cofactor to aPC in the inactivation of FVa and FVIIIa.
This then results in a marked reduction in activity of the prothrombinase
complex.
Additional inhibitors include the complement component C1-esterase
inhibitor (which inhibits FXIIa) and antitrypsin (which can inhibit factor FXIa).
In addition, α2-macroglobulin acts as a broad-spectrum proteinase inhibitor,
which has a secondary function to other proteinases and may also be involved in
the regulation of the immune system.
Fibrinolysis
Additional clotting control comes from lysis of the fibrin clot (Figure 1.3). Fibri-
nolysis results from a series of enzymes that control the cleavage of fibrin. These
include plasminogen, which when activated by plasminogen activators results in
plasmin. This lyses fibrin to fibrin degradation products (including D-dimers).
The major activator of plasminogen is tissue plasminogen activator (tPA) – a
serine protease produced by endothelial cells – but FXIIa, FXIa, kallikrein and
The action and principal controls of fibrin breakdown (fibrinolysis) are shown. Solid lines indicate activation and
dotted lines indicate inhibition.Tissue plasminogen activator (t-PA) activates plasminogen to plasmin, which can
break down both fibrin and fibrinogen. This results in the generation of a number of fibrinogen and fibrin
degradation products, which can have an overall inhibitory effect on further fibrin clot generation. Products
released from fibrinogen include D and E fragments, and products released from formed, cross-linked fibrin
include D-dimer, Y-dimer and DY fragments. t-PA is, in turn, inhibited and controlled by plasminogen activator
inhibitor (PAI)-1. In pregnancy the placenta produces PAI-2, although it does not have a major function in the
control of fibrinolysis in the mother. Plasmin is principally inhibited by α-2 antiplasmin, although there is a
contribution from α-2 macroglobulin. Thrombin is also capable of the activation of thrombin activatable
fibrinolysis inhibitor (TAFI), which inhibits the action of plasmin on formed fibrin. In addition, urokinase (u-PA),
which is found in urine, can activate plasminogen. uPA itself is activated from pre-u-PA after limited hydrolysis
by kallikrien, which is derived from the intrinsic pathway of coagulation.
8
Obstetric CH01 18/10/05 10:20 am Page 9
Pre-u-PA
PAI-1
Coagulation cascade Kallikrien
u-PA t-PA
Fragments D/E
PAI-2
FIBRINOGEN
9
Plasmin Plasminogen
THROMBIN
burst
FPA
Fibrin
XIII XIIIa
CROSS-LINKED
FIBRIN
D-DIMER
Thrombomodulin DY/ YY FRAGMENTS
TAFI
Obstetric CH01 18/10/05 10:20 am Page 10
urokinase (uPA) are also capable of activating plasminogen. Release of tPA from
the endothelium near the site of injury may result from the action of fibrin, by
thrombin attached to the fibrin clot or by the effect of venous occlusion itself.
The primary inhibitor of plasmin is α2-antiplasmin. Other potential plasmin
inhibitors include α2-macroglobulin, antithrombin, α1-antitrypsin and C1-esterase
inhibitor. In addition, there are four plasminogen activator inhibitors of which
plasminogen activator inhibitor (PAI)-1 is the most important in vivo. PAI-1 is
released from the endothelium in response to agents such as thrombin and
endotoxin. PAI-2 is produced only in the placenta and so is only found in preg-
nant women. In addition to these controls, thrombin, when bound to thrombo-
modulin, can activate thrombin-activatable fibrinolysis inhibitor (TAFI). TAFI
cleaves the COOH terminal end of fibrin. This reduces the ability of fibrin to
facilitate plasminogen activation via tPA. TAFI also directly inhibits plasmin activ-
ity and, when cross-linked to fibrin, prevents premature lysis.
F, Factor; FPA, Fibrinopeptide A; TAT, Thrombin–antithrombin complex; vWF, von Willebrand Factor.
Pregnancy results in a fall in free and total protein S levels, with levels less
than those of non-pregnant subjects observed in the first trimester, and levels
<50% of those observed in the first trimester evident between 36 and 40 weeks
gestation.
Protein C activity 0 0
Protein C antigen 0 0
Antithrombin activity 0 0
Protein S total 9 15
Protein S free 20 30
Plasma thrombomodulin 10 41
APTT–APCR 13 20
Modified APTT–APCR 0 0
Fibrinolysis
The activation and inhibition of fibrinolysis occurs at a local level, so the study of
plasma can only give a limited insight into the balance of fibrinolysis during preg-
nancy. Increasing gestation is associated with a complex alteration in plasma
markers of plasminogen activation and inhibition. Plasminogen activity, and PAI-
1 and PAI-2 levels increase throughout pregnancy. This is accompanied by a
probable increase in tPA antigen but a reduction in its activity and release after
venous occlusion. No alteration in α2-antiplasmin levels has been reported in
normal pregnant subjects. A number of studies have shown that increasing gesta-
tion is associated with an overall reduction in systemic fibrinolytic activity.
Despite this, pregnancy is associated with an increase in circulating fibrin degra-
12
Obstetric CH01 18/10/05 10:20 am Page 13
dation products. The reason for this apparent disparity is not known, although it
may point to an increase in fibrin generation and degradation in the placenta
(despite a reduction in the fibrinolytic potential of the general circulation), or to
a reduction in the plasma clearance of these products.
References
Dahlback B, Carlsson M, Svensson PJ, Proc Natl Acad Sci USA 1993;
90(3):1004–8.
14
Obstetric CH02 18/10/05 10:21 am Page 15
CHAPTER 2
15
Obstetric CH02 18/10/05 10:21 am Page 16
Red cells
Although many transfusion triggers have been proposed, as a general rule, red
cells should be used to treat significant hypoxia due to anemia, i.e. anemia
causing cardiac or respiratory distress. In certain circumstances, red cell transfu-
sions may also be used for other purposes, such as the suppression of dyserythro-
poiesis in thalassemia, or the dilution of sickle cells in sickling disorders.
Component Indications
FFP
FFP contains all the labile clotting factors and is used to correct a coagulation
deficiency – identified by prolongation of the prothrombin or partial thrombo-
plastin times – when bleeding is ongoing, or is predictable, e.g. immediately
prior to surgery or closed procedures such as liver biopsy, lumbar puncture and
epidural anesthesia. As it is a human blood product (with the attendant risks of
immunological or infectious complications), it should not be used for fluid
replacement and, where possible, single clotting factor deficiencies should be
treated with specific virally inactivated fractionated products. It should be
remembered that provision of FFP to the patient will require at least 30
minutes to permit thawing of the product and transport from the transfusion
laboratory.
The exceptions to the indications noted above are the provision of the pro-
tease required in the treatment of thrombotic thrombocytopenic purpura
(TTP)/hemolytic uremic syndrome (HUS) (see Chapter 8), and the provision of
anticoagulant proteins such as protein S when a specific product is not available.
FFP has no role in the reversal of heparin, but can be used in the reversal of war-
farin. However, severe bleeding secondary to warfarin will require a prothrombin
concentrate, as FFP will not lead to an adequate restoration of plasma FIXc levels
in these circumstances.
also been used to maintain the placental integrity. Where a licensed virally inacti-
vated fibrinogen concentrate is available, this should be used when such fibrino-
gen treatment is required.
Platelets
Platelets are produced either by plasma exchange (where one adult dose is
obtained from one donor) or from the buffy coat of whole blood donations
(where four donations are required to provide an equivalent adult dose).
Platelets are stored at room temperature (specifically 22 +/–2°C) and have a shelf
life of 5 days if continually agitated. Platelet transfusions are indicated in the pre-
vention and treatment of bleeding due to thrombocytopenia (a reduced platelet
count) or thrombasthenia (a qualitative platelet defect). As with all plasma prod-
ucts, platelets carry the risks of transfusion outlined below. In addition, there is
the potential risk of maternal RhD alloimmunization.
Thrombocytopenia can occur for a number of reasons (see Chapter 8).
Although, in general, platelets should only be used where bleeding is attended by
thrombocytopenia, there is also a role, albeit more limited, for prophylactic use.
A threshold for prophylaxis is not evidence based, but there is consensus that a
threshold of 10 × 109/l is now acceptable if there are no additional risk factors
such as infection, vomiting or coagulopathy. This contrasts with the previous
accepted threshold of 20 ×109/l. An individual decision on dosing will be
required if there is a concern over platelet refractoriness, or if there is immune
thrombocytopenia (ITP: when donor platelets may be destroyed in the same way
as endogenous platelets), or when the patient has a stable chronic thrombocy-
topenia.
In non-pregnant subjects, a platelet count of 50 × 109/l has been adopted as
the safe minimum for surgical procedures, or when there is major bleeding. With
Cesarean sections, a level of at least 80 × 109/l has been recommended to allow
epidural anesthesia; for a vaginal delivery (without epidural anesthesia), a level
of 50 × 109/l is acceptable. Clearly, in the peripartum patient with thrombocy-
topenia, non-steroidal analgesics should be avoided. In each instance a check
should be made that the appropriate platelet count has been achieved before the
commencement of surgery.
In platelet function disorders, medication that interferes with platelet func-
tion should be avoided or withdrawn (such as non-steroidal analgesics and
aspirin), and the hematocrit should be optimized prior to platelet transfusion.
Desmopressin (trade name DDAVP: see Chapter 4) should be considered if there
is an inherited platelet defect. As DDAVP is very similar to oxytocin, there was
concern that DDAVP might induce uterine contractility. However, it is now
known that these agents operate through different receptors and that DDAVP
does not cause uterine contractions.
When RhD-positive platelets are transfused to an RhD-negative woman (who
is pregnant or in the reproductive age group), a dose of 250 IU of anti-D
18
Obstetric CH02 18/10/05 10:21 am Page 19
Immunoglobulin
Intravenous human normal Ig (IVIgG) is used in the management of a number
of conditions which can occur in pregnancy. These include ITP, neonatal alloim-
mune thrombocytopenia (NAIT), post-transfusional purpura (PTP: see Chapter
8), Guillain-Barré syndrome, myesthenia gravis and some cases of hemolytic
disease of the newborn (HDN). Although immunoglobulin has also been used in
the management of recurrent fetal loss, recent systematic reviews and clinical
trials have shown no evidence for this use. However, further information on
patient selection and the timing of infusions may yet indicate a subset of patients
who may benefit from it.
As with any blood component there is a risk of pathogen transmission,
although modern IVIgG should undergo at least two distinct viral inactivation
procedures; and furthermore, there is some evidence that the process of fraction-
ation itself leads to a reduction in prion contamination. The most commonly
reported significant side effects with IVIgG are fever, acute renal failure, jaun-
dice, thrombosis and a positive direct antiglobulin test (+/– autoimmune hemol-
ysis). However, these problems may also relate to the underlying condition.
Acute tubular necrosis appears more common in products containing sucrose,
and occurs more frequently in subjects with pre-existing renal disorders or those
on nephrotoxic drugs. In such cases a low sucrose product should be used. In all
cases, renal function should be checked prior to infusion and monitored in the
days after completion. In addition, the maximum dose and dose rate should
never be exceeded. Mild hypersensitivity to IVIgG is not uncommon, but severe
reactions are rare and may be associated with severe IgA deficiency. There are a
number of reports of venous thrombosis, perhaps related to increased plasma vis-
cosity or enhanced platelet aggregation; and there are also reports of cerebral
ischemia of arterial origin (CIAO) in association with IVIgG use. The exact risks
are, however, hard to define, as no substantial comparative trials of different
IVIgG products have ever been carried out.
19
Obstetric CH02 18/10/05 10:21 am Page 20
body is also present. When, as is more often the case, the patient’s serum reacts
equally against all donor cells, there are a number of strategies to determine if
there is also an underlying alloantibody.
If the patient has not received a transfusion within the previous 3 months,
then it can be assumed that there will not be significant levels of any previously
transfused donor red cells in the patient. In these circumstances, using a whole
blood sample, any autoantibody bound to the patient’s red cells is removed with
an enzyme (‘ZZAP’). These ‘stripped’ cells are then used to adsorb the free
autoantibody from a sample of the patient’s plasma at 37°C. This process
(autoadsorption) is repeated several times to remove as much of any autoanti-
body from the patient plasma as possible. The resultant plasma is then used
against donor cells to determine if any alloantibody is present. Such an autoad-
sorption procedure requires a considerable sample of the patient’s blood.
If there has been a history of transfusion within the previous 3 months then
any donor cells still in the patient may bind to the alloantibody, making detec-
tion of an alloantibody unreliable. If autoadsorption cannot be carried out, then
an attempt to elute the autoantibody from the patient’s serum using allogenic
red cells with a variety of antigens can be attempted. A combination of reactions
will often assist in detecting a significant hemolytic red cell antibody.
One further strategy is to phenotype the patient and select donor blood that
carries the same antigens. Ideally, this phenotyping should include the RhD, C, E,
c, e, Kell, Jka, Jkb, Fya, Fyb, S and s antigens. However, the presence of an autoanti-
body that is adherent to the patient’s red cells may make such extended pheno-
typing impossible, and in such cases autoadsorption will be required.
If the patient’s autoantibody is only cold acting, then the compatibility test
can be carried out strictly at 37°C. In such circumstances, the autoantibody will
have no effect on the compatibility procedure. Such patients should, of course,
be given their transfusion through a blood warmer.
In extremely urgent circumstances, the appropriate blood for transfusion may
have to be selected upon the basis of the ‘least reaction’ of donor cells against the
patient’s plasma. This is an extremely unreliable approach and every effort should
be made to formally exclude a significant alloantibody. Further, whether such a
selection of ‘least incompatible’ blood (based upon a slight variability in reactivity in
testing of the plasma against a variety of donor cells) improves the survival of donor
cells in the patient, when an alloantibody has already been excluded, is unknown.
In urgent circumstances, communication between the laboratory and the clin-
ician is vital, as there may often be time to carry out a limited phenotype to
improve the safety of the transfusion when time does not permit an auto-adsorp-
tion procedure.
Transfusion reactions
Reactions to transfusions can occur for a number of reasons (Table 2.2). Of these,
hemolytic reactions are the most clinically important given their potential fre-
21
Obstetric CH02 18/10/05 10:21 am Page 22
Febrile non-hemolytic Red cell reactions: from recipient white cell antibodies
Platelet reactions: from donor white cell cytokines
TRALI: Donor HLA antibodies
Allergy Urticaria/anaphylaxis
Infection Bacterial:
– Environmental: Staphylococci / Pseudomonas
– Syphilis (Treponema)
– Lyme disease (Borrelia)
– Yersinnia/Salmonella
– Q fever (Coxiella)
Viral
– Hepatitis A, B, C or D
– HIV
– HTLV
– EBV, CMV, HHV
– Parvovirus
Protozoal
– Malaria
– Chagas’ disease
– Toxoplasma
– Babesia
– Leishmania
Prion?
CMV, Cytomegalovirus; EBV, Epstein-Barr virus; HHV, Human herpes virus; HLA, Human leukocyte antigen; HPA,
Human platelet antigen; HTLV, Human T-cell leukemia/lymphoma virus; TRALI, Transfusion-related acute lung injury.
22
Obstetric CH02 18/10/05 10:21 am Page 23
Hemolytic reactions
These result from the binding of an antibody to a red cell antigen. This leads to
opsonization, with or without complement activation, red cell destruction and
the activation of inflammation. In acute reactions this occurs within 24 hours
whereas, in delayed reactions, this usually occurs within 5–7 days. Acute hemoly-
sis is often mediated by IgM, which efficiently activates complement leading to
intravascular destruction, with release of hemoglobin into the circulation and
urine. In the delayed reaction, IgG is often the mediator. As a result there may be
no, or incomplete, complement activation. In such cases, red cell breakdown
usually occurs in the spleen, leading to a fall in blood hemoglobin levels rather
than frank intravascular hemolysis. Cytokines may be released as a consequence
of both complement and macrophage/monocyte activation.
Antibodies against ABO are the most common cause of acute hemolytic reac-
tions. This most often results from clerical error leading to the misidentification
of the unit of red cells for donation, or misidentification of the recipient. Anti-
bodies against A, B, H (O), K (one of the Kell group), Jk a (one of the Kidd group)
and Le a (one of the Lewis antigens) are often associated with intravascular hemol-
ysis. Antibodies against the Rh, Duffy or M, N, or S blood group antigens are
most often associated with extravascular hemolysis.
In the acute reaction, even a small volume of red cells may be sufficient to
trigger hemolysis. The first signs are often patient apprehension, agitation, fever
and diffuse abdominal pain. This may lead to breathlessness, shock, disseminated
intravascular coagulation and acute renal failure. Immediately upon suspicion of
a reaction, the infusion of blood should be stopped. If possible, the unit of blood
should be taken down with the administration set still attached, which allows
examination of the unit for evidence of bacteriological contamination. Manage-
ment should follow the principles shown in Table 2.3.
Delayed hemolytic reactions are usually the result of an anamnestic response
occurring upon re-exposure to an antigen. Such re-exposure leads to an increase in
the level of antibody in the recipient which can result in red cell destruction. This
usually leads to asymptomatic coating of the transfused cells with antibody (a positive
direct antiglobulin test), but can result in clinically evident hemolysis with fever, jaun-
dice, a falling hemoglobin level and spherocytosis. This is usually clinically evident
some 5–7 days after the transfusion. Although multiple antibodies can be involved,
Rh group, Kell, Duffy and Kidd antibodies are most often implicated. Repeat testing
some days after the transfusion may be required to detect the antibody. Severe reac-
tions are uncommon and specific treatment, other than the provision of suitable
antigen-negative transfusion for continuing anemia, is seldom required.
Urticarial Chlorpheniramine
Allergy
Mild allergic reactions, such as urticaria and wheeze, are not uncommon with the
transfusion of blood components, and may occur after single or multiple expo-
sure. Although often difficult to disentangle from a reaction to concomitant ther-
apies, their immediate management is akin to that of other allergic reactions. Of
note, is the rare occurrence of severe IgA deficiency (IgA <0.05mg/dl) in the
recipient. This may trigger a reaction on exposure to donor IgA. The detection
of specific anti-IgA antibodies in the recipient may assist in making the diagnosis,
and such individuals may require washed red cells (which removes the IgA) or
consideration for blood-sparing modalities such as cell salvage or autologous
donation.
TA-GVHD
TA-GVHD is a very rare, but almost universally fatal, condition caused by the
infusion (in a transfusion component) of immune competent lymphocytes.
These cells are able to mount an immunological reaction against the recipient.
This most commonly occurs in those recipients who share an HLA haplotype
with the donor. However, immunodeficiency or immunosuppression in the
recipient also predispose to the condition. Patients often present with a rash,
diarrhea and abnormal liver function. This progresses to marrow failure and
sepsis within 4 weeks of diagnosis. Although there is no specific treatment for the
condition, it can be effectively prevented by gamma irradiation of cellular trans-
fusion components prior to transfusion.
PTP
Around 2% of Caucasians do not carry the platelet antigen HPA-1a on their
platelets and PTP usually occurs in parous women who do not carry this antigen
(see Chapter 8). It is assumed in such cases that the mother has carried an HPA-
1a positive fetus in a previous pregnancy that has resulted in immunization of the
mother, but has not led to the development of neonatal alloimmune thrombocy-
topenia (see Chapter 8). Subsequent exposure to the HPA-1a antigen, via a red
cell transfusion, results in an increase in the titer of anti-HPA-1a antibodies. This
leads, by an essentially unknown mechanism, to destruction of the women’s
platelets (despite the fact that they do not carry the HPA-1a antigen). This often
results in a rapid fall in platelet count presenting around 1 week after the trans-
fusion. The resultant thrombocytopenia results in widespread bruising and
bleeding from the gastrointestinal or renal tracts.
The condition is diagnosed by the history of recent transfusion and the detec-
tion of anti-HPA-1a antibodies in the recipient’s serum and exclusion of other
causes of thrombocytopenia (see Chapter 8).
Other HPA antibodies have also been reported in association with PTP,
including anti-HPA-1b, -3a, -3b, -4a or -5b. As further donor platelets (even if
antigen negative) will also be destroyed, platelet therapy may be ineffective.
Indeed, there is no evidence that the specific HPA antigen-negative platelets will
be more effective than random platelets. So therapy is geared towards suppres-
sion of the immune response with high-dose IVIgG therapy. A number of
successes have also been reported with plasma exchange. The provision of
blood component therapy after an episode can also be problematic, as a recur-
rence is unpredictable. It is usual practice to attempt to provide appropriate
HPA negative blood for future transfusions, although there may also be a role
for leukodepletion or autologous donation.
26
Obstetric CH02 18/10/05 10:21 am Page 27
Bacterial contamination
Bacterial contamination may be present in as many as 1 in 3000 cellular blood
component transfusions and, when severe, presents as an acute hemolytic trans-
fusion reaction. The true incidence is difficult to judge, as minor contamination
may not lead to clinical problems. Around 60% of fatalities are associated with
microbial contamination of platelets.
The primary source of bacterial contamination of blood products is the
donor. This can be due to poor cleaning technique at the venesection, or asymp-
tomatic bacteremia in the donor (such as occurs after dental treatment). On
account of this, bacterial contamination is also a problem with pre-deposit auto-
logous donation. Problems can be reduced by: taking an adequate donor history;
meticulous venesection technique; specific systems to detect microorganisms in
blood components; and inspection of the blood component prior to transfusion.
As red cell components are stored at 4°C, Gram-negative organisms such as
Yersinnia and Pseudomonas are often implicated. By contrast, as platelets are
stored at room temperature, Gram-positive organisms such as staphlyococci and
streptococci are often the cause of such contamination.
also not yet known. Although animals have a different tissue distribution of prion
proteins to humans, transmission of prion disease by transfusion has been
demonstrated in sheep. Furthermore, in 2004, a person who had donated blood
in the UK subsequently developed nvCJD. The recipient of his donation also
went on to develop nvCJD some time after the transfusion. Although a link
cannot be proven, there is the potential that direct introduction of the abnormal
prion by transfusion may bypass the protective effects of enzyme degradation that
take place if the prion is ingested in infected meat. It is likely, however, that the
absolute risk of prion transmission by transfusion is low. There is also likely to be
a reduction in blood product contamination with prions if the products have
undergone a fractionation process, such as is used in the manufacture of
immunoglobulin or clotting factor concentrates.
However, this theoretical risk of prion transmission by transfusion, allied to
the absence of a screening test (although many first generation tests are in devel-
opment) and evidence that the protein is not susceptible to standard viral inacti-
vation procedures, highlights the need to restrict blood product usage to those
indications where there is a clear, positive, benefit:risk ratio.
Cell saving
Cell saving encompasses a number of techniques which attempt to reduce expo-
sure to allogenic blood. These techniques include autologous pre-deposit, intra-
operative hemodilution and intra- or postoperative cell salvage. A number of
other therapies, such as recombinant erythropoietin and antifibrinolytic agents,
are also used to reduce allogenic exposure, and a further strategy is the develop-
ment of useful blood substitutes.
In view of the potential risks of allogenic transfusion there has been consider-
able interest in the use of cell-saving techniques in relation to delivery and, in
particular, Cesarean section. Although techniques such as pre-deposit of autolo-
gous blood and intraoperative cell salvage are well tolerated and considered safe
in the majority of instances, their use in pregnancy requires due consideration of
their potential effects on maternal and fetal wellbeing. In addition, the wide-
spread adoption of these techniques requires clarification of those in whom such
interventions would be clinically or cost-effective.
Autologous pre-deposit
There are many case series examining the feasibility of autologous pre-deposit in
relation to obstetrics. Most often pre-deposits (equivalent to 1–2 units of red
cells) are taken by standard venesection in the third trimester. Although mater-
nal side effects, such as faintness, are well tolerated, the effects of blood loss on
the feto-placental unit are not fully understood. Although it may be that there is
no effect on cord blood pressure and flow, it has been suggested that venesection
of 400ml of maternal blood in the third trimester is associated with a reduction
28
Obstetric CH02 18/10/05 10:21 am Page 29
in fetal middle cerebral artery pulsatility in the first 24 hours after the event. In
general, to be suitable, women require a hematocrit sufficient (usually >33%) to
permit safe venesection. In addition, there has to be a high probability that col-
lected units will be used. Given this, autologous pre-deposit can be justified in
those with multiple or difficult red cell alloantibodies (e.g. thalassemia), those in
whom bleeding is likely (e.g. where there is a severe bleeding disorder or pla-
centa previa) and those subjects who cannot receive transfusions (e.g. Jehovah’s
Witnesses, Bombay phenotypes). In most obstetric patients without placenta
previa, the need for peripartum transfusion cannot be predicted with sufficient
accuracy to justify antepartum pre-deposit, and in studies of feasibility, only
14–34% of subjects with placenta previa were suitable for pre-deposit. Indeed, as
with many cell-saving techniques, the use of pre-deposit leads to an overall
increase in the likelihood of transfusion. This increase in the likelihood of use of
the autologous pre-deposit means that this modality of therapy actually carries a
similar risk of both bacterial contamination and of clerical error to that found in
allogenic transfusion.
Erythropoietin
Erythropoietin (Epo) is a glycoprotein, which is synthesized in the kidney in
response to hypoxia. Recombinant human Epo is derived from Chinese hamster
ovary cells, and has been used widely in anemia associated with renal failure,
malignancy and chemotherapy. It is given subcutaneously, at a dose of
50–200IU/kg two–three times per week along with oral or parenteral iron, with
the aim of increasing the hemoglobin by 1 g/dl/month. It has been used in
Jehovah’s Witnesses and in the management of renal failure in pregnant subjects,
and appears to be well tolerated. Although it does not cross the placenta, it
29
Obstetric CH02 18/10/05 10:21 am Page 30
Blood substitutes
There is a demand to produce red cell and platelet substitutes to address a com-
bination of concerns. These include shortages in blood supply, the risks of conta-
mination and immunomodulation, the costs associated with blood collection, the
cost of temperature-regulated storage, the need for cross-match procedures, and
the need for strict patient identification. There has been a substantial advance in
the development of solutions able to achieve volume expansion and, although a
number of blood substitutes able to carry oxygen are in development, the major-
ity are not yet in clinical use. Such products are required to take up oxygen in
the lungs and give it up in the tissues. They are also required to be non-toxic, to
have a useful half-life in the body and a useful shelf life for storage.
There has been much attention on hemoglobin-based solutions. Unfortu-
nately, hemoglobin readily participates in redox reactions leading to oxidative
damage. As a consequence, free hemoglobin has a short half-life and is a potent
vasoconstrictor. A number of attempts to improve the stability of hemoglobin by
cross-linking have been attempted. Of these, bovine polymerized hemoglobin
has a licence for use in veterinary practice. The different hemoglobin products in
current development, however, have differing oxygen affinities, hemoglobin con-
centrations and stability. The major side effect of these products is hypertension,
which results from vasoconstriction (by an as yet poorly defined mechanism,
perhaps related to a local reduction in nitric oxide or alteration in viscosity),
which may in fact hamper oxygen supply.
One other group of products are the perfluorocarbon emulsions, which take
up oxygen to a greater degree than human plasma, but do not have the flexibility
of uptake and supply associated with hemoglobin. Perfluorocarbon emulsions
may also modulate the inflammatory response, and their use can be associated
with the development of flu-like symptoms.
At present, none of the available red cell substitutes offer the immediate
prospect of a useful oxygen supply without the risks and costs associated with
current transfusion therapies, and there is no specific evidence of their safety in
pregnancy.
these the most important Rh type in obstetric practice is RhD, which is around
50 times more immunogenic than other Rh types. Fifteen per cent of Cau-
casians, 5% of Black subjects and 2% of Asians are RhD negative. When an
RhD-negative mother is carrying an RhD-positive child, the transplacental
passage of both fetal blood and maternal IgG immunoglobulin may result in
the development of an anti-RhD antibody in the mother that is also active in
the fetus.
Whether the mother develops an antibody depends upon the amount of fetal
blood entering the maternal circulation and the presence of any feto-maternal
ABO mismatch, which reduces the development of an anti-RhD antibody (pre-
sumably by clearance of fetal cells by pre-formed maternal anti-A or anti-B anti-
bodies). The Rh haplotype carried by the fetus may also be important as the fetal
carriage of cDE/cde is more likely to induce antibodies than other RhD-positive
haplotypes. If an RhD-negative woman carries an ABO-compatible, RhD-positive
child there is a one in six chance of forming anti-RhD in the absence of prophy-
laxis. If an RhD-negative recipient receives an RhD-positive transfusion there is a
90% chance of forming anti-RhD. The majority of hemolytic disease cases occur
in the second pregnancy, but in some instances the fetal cells may result in a
brisk maternal response leading to significant fetal hemolysis in the first preg-
nancy.
The amount of RhD expressed on the red cell is dependent upon which
other members of the Rh haplotype are present. There are also many other
quantitative and qualitative variations in RhD expression. Where there is a
reduction in the expression of all RhD epitopes, this is termed ‘weak D’. The
definition of weak RhD is, however, dependent upon the sensitivity of the local
laboratory reagents used to detect RhD. On the other hand, the absence of
some of the RhD epitopes is termed ‘variant D’, and there are many classes of
variant D. As someone with a weak D has all the RhD epitopes present, exposure
to RhD-positive blood will not result in such an individual developing anti-RhD,
and such individuals should be classed as RhD-positive. The importance of
detecting weak RhD in blood donors is a matter of debate, but such weak D cells
could be capable of eliciting an antibody response in a recipient. On the other
hand, if someone has a variant D, they may be capable of developing anti-RhD
on exposure to RhD-positive cells (from a fetus or from transfusion). In particu-
lar, one type of variant RhD (called RhDVI) may, upon exposure to RhD, lead to
the development of both anti-RhD and HDN. On account of this, in obstetric
practice, the laboratory reagents used for antenatal typing should not detect the
RhDVI variant, and such mothers should be classed as RhD negative. As such,
they should receive RhD-negative blood products and prophylactic RhD
immunoglobulin if potential sensitizing events occur (Table 2.4). Whether a
transfusion of DVI donor blood carries a significant risk of sensitizing a recipient
is not clear. In any case, when a laboratory cannot reliably distinguish RhD vari-
ants from a weak RhD phenotype, it is safer for the mother to be considered to
be RhD-negative.
31
Obstetric CH02 18/10/05 10:21 am Page 32
There are three general classes of IgG immunoglobulin, each with differing
characteristics. Of these, HDN only occurs with IgG of the subclasses IgG1 or IgG3,
which are capable of transplacental passage. The transplacental passage of IgG1 may
occur as early as 20 weeks gestation, with the peak of IgG3 evident at around 28
weeks. Despite this, IgG3 is associated with greater postnatal jaundice than IgG1. As
IgG1 rarely binds complement, the mechanism of red cell destruction is not clear,
but may involve increased presentation of the IgG Fc component to macrophages.
The majority of affected fetal red cells are destroyed in the fetal spleen.
cord blood sample should be obtained to determine the ABO and RhD type of
the baby. At delivery, the presence of anti-RhD on the fetal cells should be exam-
ined by direct antiglobulin test, and the mother should have a blood sample
analyzed for an estimation of FMH.
There is a correlation between the gestation at which antibody is first detected
and the severity of any resultant HDN. Therefore, if antibodies are detected at
booking, they are likely to cause more problems than those detected for the first
time later in pregnancy. If an antibody associated with HDN is detected this
should be quantified. For most antibodies this will be achieved by the titer on an
indirect antiglobulin test. It has been recognized for some time that automated
quantification of anti-RhD antibody levels provides more reproducible and
meaningful values than manual titration, with the level of maternal anti-RhD
immunoglobulin used to plan further testing and the need for fetal blood sam-
pling or delivery. Indeed, in the case of RhD, it is recommended that the anti-
RhD titer should be reported in international units (IU). In general, the absolute
value is not as important as any rise in titer with increasing gestation. However,
when the levels remains at <4 IU/ml, HDN is unlikely and few fetuses suffer
major anemia. In such cases, serial anti-RhD monitoring and ultrasound determi-
nation of a combination of parameters (such as Doppler of umbilical vein or
middle cerebral artery flow, liver and spleen size, and the early detection of
ascites) may assist in management.
When the levels are 4–15IU/ml, there is a moderate risk of HDN and investi-
gation is intituted in some, but not all, areas of the UK. Early in pregnancy, geno-
typing techniques can be used to genotype the fetus (when the partner is
unknown, or it is an RhD heterozygote). Such polymerase chain reaction (PCR)
techniques do require due consideration of the ethnic origins of the partners,
and both false-positive and false-negative results can occur.
33
Obstetric CH02 18/10/05 10:21 am Page 34
In addition, it is now possible to determine if the fetus is RhD positive (as well
as the Kell and c status: see below) from free fetal DNA obtained from the mater-
nal circulation. Although this is in its infancy, very high sensitivity has been
reported in some studies using fluorescence-based PCR techniques.
After 27 weeks gestation, amniocentesis can be used to determine the level of
bilirubin in the amniotic fluid by its absorption at 450 nm. The level obtained is
interpreted by a Lilley graph – essentially relating the level to a gestation-specific
reference range to assist in determining the necessity for intrauterine transfusion
or delivery. This technique reflects the degree of fetal hemolysis but not the fetal
hemopoietic response, and significant discrepancies between the fetal hemat-
ocrit and the amniotic fluid optical density can occur. Analysis of amniotic fluid
anti-D concentrations has also been investigated. Although these correlate well
with maternal serum anti-D levels, they have no added value in predicting the
severity of fetal hemolysis.
As yet, in such cases, fetal assessment by most non-invasive methods, including
fetal movement counting, ultrasound appearance and fetal electocardiogram
waveforms, have not proved helpful in determining whether the fetus will
become anemic. Abnormal fetal heart rate patterns have been described in
severe disease, and although cardiotocography (CTG) will identify the seriously
anemic fetus it is not sensitive enough to predict mild to moderate anemia. Com-
puterized fetal heart rate analysis has identified a consistent relationship between
baseline variability and fetal hematocrit, and warrants further study.
Weekly velocimetry of the fetal middle cerebral artery has also been used to
monitor affected fetuses. In such cases, peak systolic velocities >1.5 multiples of
the median for the specific gestation have been shown to be predictive of moder-
ate or severe fetal anemia (with 100% sensitivity and a false-positive rate of
around 12%), and ultrasound studies correlate well with the bilirubin level in the
amniotic fluid. This technique has begun to replace serial amniocentesis and is
now being used to predict when fetal blood sampling is required.
The only direct method of assessing the severity of RhD disease, however, is by
measuring the hematocrit of fetal blood samples. When levels exceed 15IU/ml
or if indicated by velocimetry, fetal blood sampling at >18 weeks gestation may be
required. This is not without complications (from <2% mortality in non-affected
fetuses to 5–15% fetal mortality in affected cases), and the procedure can result
in a marked increase in the maternal antibody levels. Other reported complica-
tions include fetal exsanguination, bradycardia, failure to obtain a sample and
cord constriction. Fetal blood sampling can be achieved by ultrasound-guided
sampling of the cord vessels, the hepatic vein, or the fetal heart, which will also
permit an assessment of the fetal RhD group. Fetal blood sampling should only
be carried out, however, if there is the expertise and facilities to carry out an
intrauterine transfusion, if required.
In summary, when the maternal anti-RhD level is <0.5IU/ml, four-weekly
monitoring of anti-RhD levels is all that is required. If anti-RhD levels are
>0.5IU/ml, then two-weekly anti-RhD assessment is required. If levels are
34
Obstetric CH02 18/10/05 10:21 am Page 35
between 10 and 20IU/ml, fetal blood sampling will usually be required, but the
fetus does not often require an intrauterine transfusion. If the maternal anti-RhD
level is >20IU/ml, fetal blood sampling will be required and the fetus will prob-
ably require intrauterine transfusion. In general, if the anti-RhD level is >4IU/ml
but <10IU/ml, then early delivery may be required (however, usually at >36
weeks). For levels of >10IU/ml, 33–38 week delivery may be necessary.
Fetal management
If fetal maturity permits, delivery of the fetus is indicated when there is evidence
of a high level of bilirubin in the amniotic fluid. When fetal immaturity does not
make delivery feasible, fetal transfusion should be carried out. The introduction
of intrauterine fetal transfusion has reduced the perinatal mortality rate in severe
cases of HDN from 95 to 50%. Intrauterine transfusion is indicated if the fetal
hematocrit is <25% between 18 and 26 weeks gestation or <30% after 34 weeks
gestation. The blood used should be cross-matched against maternal serum and
should have a hematocrit of 75–90%, be sero-negative for cytomegalovirus and
gamma irradiated. The aim of the transfusion is to increase the fetal hematocrit
to around 45%, and the volume required is calculated from the pre-transfusion
hematocrit, the donor hematocrit and the estimated fetal blood volume. In some
units, pre-medication is given to the mother for maternal and fetal sedation,
whilst others have employed tocolytic agents, antibiotics and corticosteroids. The
transfusion may be given directly into the fetal vasculature (umbilical vein, umbil-
ical artery, or inferior vena cava), into the fetal heart, or into the fetal peritoneal
cavity. Further transfusions may be required at 1–3-weekly intervals. The compli-
cations of intrauterine transfusion include transfusion site trauma and
hematoma formation, spasm of the cord, umbilical arterial thrombosis, fetal
thrombocytopenia, an increase in maternal antibody levels, fetal leukoen-
cephalopathy and fetal bradycardia.
35
Obstetric CH02 18/10/05 10:21 am Page 36
Other modalities of therapy have been used when there has been a history of
previous fetal loss, or where the likely father is homozygous for RhD. These
include repeated, maternal plasma exchange from early in pregnancy to remove
anti-RhD from the maternal circulation until fetal transfusion therapy can be
achieved. In such circumstances, maternal procedures, which could cause allo-
immunization, should be avoided. Similarly, regular maternal treatment with
high-dose human normal immunoglobulin (2g/kg on a two-weekly basis) has
also been used in this circumstance.
After delivery, neonatal treatment consists of phototherapy to oxidize biliru-
bin and, when required, exchange or top-up transfusion.
Prevention
In the 1960s around 46 deaths/100 000 live births were attributed to the RhD
immunization in the UK. The routine use of post-delivery anti-RhD immunoglob-
ulin immunoprophylaxis was introduced in 1969. By 1990, the number of fetal
deaths attributed to RhD immunization in the UK had fallen to 1.6/100 000.
Despite this, there continue to be cases of HDN. This may be due to maternal
sensitization in a previous pregnancy, previous transfusion of red cells or
platelets, inadequate treatment of potential sensitizing events, sensitization from
unrecognized events, or reactions to red cell antigens other than RhD.
Prevention is based upon the suppression of the maternal immune response
to the RhD antigen by the timely administration of a passive antibody, the
mechanism of which remains poorly understood. Intramuscular anti-RhD should
be administered into the deltoid muscle to all RhD-negative women as soon as is
practical, but certainly within 72 hours of delivery. If it is not given before 72
hours, every effort should be made to administer it within 9–10 (or even 28)
days, as this may still offer some protection. The dose should be determined by
the level of FMH. Given intramuscularly, 125IU of anti-RhD is considered suffi-
cient to protect against exposure to 1ml of RhD-positive red cells. In the UK,
250IU is routinely given for any potential sensitizing event that occurs before 20
weeks gestation and 500IU for any event after 20 weeks gestation (see Table 2.4).
If a further sensitizing event occurs >7 days after a prophylactic dose of anti-RhD,
it is common practice to give a further dose of anti-RhD. In addition, after 20
weeks gestation an FMH assessment should be carried out to determine if the
bleed exceeds 4 ml, when a further appropriate dose of anti-RhD may be
required (see Table 2.5). It is routine practice in the UK not to give anti-RhD to a
threatened miscarriage where the bleeding has stopped before 12 weeks gesta-
tion and there is still a continuing pregnancy.
As noted above, some at-risk women may become sensitized in the last trimester
by unrecognized events. In fact, given the success of the prophylaxis for sensitizing
events, this has become the most important cause of maternal sensitization in many
developed countries. One strategy designed to cope with these ‘silent’ FMH is the
routine administration of anti-RhD immunoglobulin to all RhD-negative women
36
Obstetric CH02 18/10/05 10:21 am Page 37
who have no detectable anti-RhD antibodies in their blood. This has been shown to
further reduce the risk of antenatal immunization. At present, however, a variety of
dosages and dosage schedules for prophylaxis have been employed. Of these, two
options, which both seem effective, include a dose of 500IU at 28 and 34 weeks ges-
tation or a single larger dose (1500IU) early in the third trimester. Enthusiasm for
this method of HDN prevention, however, may be tempered by problems of anti-
RhD availability and, in Europe, the continuing concern about the possibility of
transmission of prions by blood products, although, as noted above, the process of
fractionation is thought to reduce this risk.
Non-RhD antibodies
In addition to RhD, at least 40 red cell antigens have been associated with HDN.
These include Rhc, RhC, RhE, Kell, Duffy, MNS, Lutheran, Kidd and U. After
anti-RhD, antibodies against c, Kell (K1) or E are the most frequently encoun-
tered antibodies requiring treatment.
Kell
The gene coding for Kell antigens is found on chromosome 7, although expres-
sion is also dependent upon genes on the X chromosome. Of the 22 Kell anti-
gens, antibodies to Kell (K1), k (cellano), Kpa (Penny), Kpb (Rautenberg), Jsa
(Sutter) and Jsb (Matthews) have all been implicated in HDN. Antibodies to K1
are the most commonly encountered antibodies after ABO or RhD, and are
usually IgG and occasionally complement binding. The K1 antigen is found in
9% of Caucasians (who are virtually all heterozygous). Kpb and Jsb antigens have a
very high frequency in Caucasians, and Jsb is almost universal in Black subjects. In
addition, a reduction in all Kell antigen expression can occur. This is called the
Mcleod phenotype and it arises from inheritance of Kx and leads to a chronic
mild hemolytic state.
Between 8 and 18% of pregnancies with detectable maternal anti-K1 result in a
K1-positive fetus, and hydrops has been reported in around 30% of such cases. In
recent years this incidence may have been reduced by the use of intrauterine
transfusion. In anti-K1 HDN, fetal anemia results from a combination of hemolysis
in the fetal blood as well as destruction in the marrow. This marrow suppression
leads to a lower bilirubin level in the amniotic fluid than would be expected by
the degree of anemia. The management of a sensitized women requires a combi-
nation of ultrasound and fetal blood sampling. One potential strategy in anti-K1-
positive pregnancies is to genotype the father (when paternity can be assured). If
the father is kk, no further action may be appropriate. If the father is K1k, then
anti-K1 titers <4 can be monitored monthly. If a level of 8 is reached, amniocente-
sis and fetal K1 typing may be appropriate, as 50% would be expected to be kk. If
the fetus is K1 positive, serial middle cerebral artery Doppler and cordocentesis
(with the potential for intrauterine transfusion) should be considered.
37
Obstetric CH02 18/10/05 10:21 am Page 38
MNS and U
The MNS system is coded on chromosome 4 by two linked genes: GYP A (M and
N) and GYP B (S, s and U alleles). Anti-M is relatively common and can be IgG or
IgM. Anti M can react at 37°C and very occasionally has been associated with HDN.
There is insufficient data to give specific guidance, but the presence of IgG should
be determined, as IgM anti-M will not cross the placenta. High IgG titers should
lead to further investigation with amniocentesis, and possibly fetal and paternal
genotyping. Anti-N occurs rarely and is, most often, of no clinical significance.
Both anti-S and anti-s are usually IgG and have rarely been implicated in HTR
or HDN, with the majority of cases of HDN reported resulting in mild hemolysis
only. The U antigen is essentially only found in Black subjects, and anti U is very
rare and usually of immune origin. It can be associated, however, with HDN, and
geographically where this is a problem, provision of U-negative blood for fetal
transfusion can be problematic.
Duffy
The Duffy gene consists of co-dominant alleles including Fya and Fyb. The Duffy
antigen functions as a receptor for Plasmodium vivax and carriage of the Fy(a-b-)
phenotype has been associated with an increased risk of pre-eclampsia, perhaps
due to a reduction in Duffy-mediated clearance of interleukin-8.
Antibodies to Fya are more common than Fyb, are predominantly IgG and can
bind complement. Anti-Fyb is not associated with HDN; but HDN associated with
anti-Fya has been reported to carry significant morbidity (with one third of cases
requiring intrauterine transfusion) and fetal mortality (18% of cases), with
significant fetal disease possible at relatively low maternal antibody titers.
Kidd
The Kidd antigens are coded on chromosome 18 and consist of the co-dominant
Jka and Jkb. Antibodies to Jka are more common than Jkb. They usually bind com-
plement and there is often a high percentage of IgG3. Detection of the antibody
in maternal plasma can be problematic and often requires the use of homozy-
gous Jka or Jkb for detection (i.e. a cell that has homozygote expression of the
antigen is required for binding of the maternal antibody to occur). Detection of
antibodies may also be complicated by the rapid fall in titers which often occurs
soon after an immunization that has arisen as a consequence of transfusion. Anti-
Kidd antibodies have been occasionally associated with HDN, and fetal genotyp-
ing may assist in determining appropriate care.
38
Obstetric CH02 18/10/05 10:21 am Page 39
DIC, Disseminated intravascular coagulation; HELLP, Hemolysis, elevated liver enzymes, and low platelets;
PCV, Packed cell volume; PET, Pre-eclampsia; PMH, Past medical history; PPH, Post partum hemorrhage
39
Obstetric CH02 18/10/05 10:21 am Page 40
Placental abruption
Placental abruption, or retroplacental hemorrhage, is the premature separation
of a normally implanted placenta from the uterine wall, resulting in hemorrhage
before the delivery of the fetus. It occurs in around 1 in 80 deliveries. Although
maternal mortality is rare, maternal morbidity from hemorrhage, shock, dissemi-
nated intravascular coagulation and renal failure is not uncommon. The risk to
the fetus depends on the severity of the abruption, the gestation, the birthweight
and the amount of concealed hemorrhage. The perinatal mortality with placen-
tal abruption is high, and may be >300/1000.
The bleeding, however, may be in whole, or in part, concealed if the
hematoma does not reach the margin of the placenta so that the blood loss is
revealed. In view of this, the amount of revealed blood loss reflects poorly the
degree of hemorrhage, as an enlarged hematoma may be concealed within the
uterus. The hematoma may also result in bleeding into the amniotic cavity, with
subsequent blood-stained liquor being noted when the membranes rupture. Fur-
thermore, the bleeding may infiltrate the myometrium resulting in so-called Cou-
velaire uterus. This infiltration of blood into the myometrium is associated with
sustained uterine contraction, making the uterus feel ‘solid’ on examination,
provoking labor and reducing utero-placental blood flow with serious compro-
mise to the fetus. Placental abruption may occur at any stage of pregnancy. In
severe abruption there may be heavy vaginal bleeding, or increasing abdominal
circumference if the bleeding is concealed and retained within the uterus. The
woman is usually in severe pain and may be in hypovolemic shock. The uterus
may be tender and irritable, with palpable contractions or uterine hypertonus,
often resulting in labor. Fetal distress or intrauterine fetal death may be present.
In less severe cases, the diagnosis of placental abruption may not be obvious,
particularly if the hemorrhage is largely concealed, and such cases may be mis-
diagnosed as idiopathic preterm labor. Thus, abruption should be considered in
any patient presenting with unexplained vaginal bleeding or possible preterm
labor, particularly if fetal distress is present.
40
Obstetric CH02 18/10/05 10:21 am Page 41
Summon staff
(obstetrician, midwife, anesthetist, porter)
Inform
(blood transfusion and hematology)
Access
(<2 peripheral venous cannulae, chorionic villus sampling (CVP) – if possible, arterial line advised)
Samples
(cross-match ≥6 units, coagulation screen, full blood count)
Plasma expanders
(hemacel, albumin)
Consider need for uncross-matched blood
(O RhD negative – pressure cuff, blood warmer – if possible)
Monitor
(blood pressure, pulse, electrocardiogram, blood gas)
Monitor
(hemoglobin, platelets, coagulation – including fibrinogen)
Replace
(coagulation factors with fresh frozen plasma (FFP)/cryoprecipitate)
Early intervention
(artery ligation, Cesarean hysterectomy)
Assess
(need for intensive therapy unit)
41
Obstetric CH02 18/10/05 10:21 am Page 42
42
Obstetric CH03 18/10/05 10:21 am Page 43
CHAPTER 3
Iron deficiency
Iron has a wide distribution in the body, with around 70% located in hemoglobin
and myoglobin. Most non-heme iron is found in ferritin or hemosiderin, in
either hepatocytes or macrophages. There is a small amount of iron associated
with the iron-transport protein transferrin; and iron is also required for the func-
tion of enzymes, such as catalases and peroxidases. When iron is required for
incorporation into these proteins, it is most often derived from recycled iron that
has been released from red cell breakdown.
Iron absorption takes place in the first part of the duodenum, and iron
balance is achieved by the regulation of this absorption. Iron is absorbed from
heme, or from ferrous (non-heme) iron by two distinct pathways. Non-heme iron
absorption can be enhanced by ascorbic acid, which assists in the conversion of
ferric iron to the ferrous form (the state that is required for absorption). By con-
trast, phytates (found in cereals) bind non-heme iron and inhibit its absorption,
and dietary phosphates also inhibit iron absorption from eggs and milk.
Iron is released from heme on the cell border, and inside the mucosal cell is
added to the pool of absorbed iron. Some of the iron is transported into the
plasma within a few hours of absorption, but much remains within the mucosal
cell. Loss of the mucosal cell (and its iron), by sloughing, appears to have a
major role in the regulation of iron balance.
It is likely that iron is transported from the mucosal cell by the action of a
transmembrane protein, whose mRNA synthesis is stimulated by iron deficiency.
Iron leaving the cell must be oxidized to the ferric state to allow binding to the
transport plasma protein (apo-) transferrin. Iron absorption is increased when
body iron stores are depleted and reduced when excessive body iron is present.
Iron absorption is also increased when there is an increase in erythropoiesis,
hypoxia or inflammation. Iron is then transported in the plasma by the glycopro-
tein transferrin. This is synthesized predominantly in the liver in response to iron
deficiency: the total amount of iron that the available plasma transferrin can
bind is called the total iron binding capacity (TIBC). In normal non-pregnant
subjects around one third of the TIBC is used. Transferrin carries iron to specific
transferrin receptors, which are prominent on the developing red cell but are
lost as the red cell nears maturity. When transferrin saturation falls to <16%
there is insufficient iron delivery to the bone marrow. This results in an increase
in free red-cell-derived protophorphyrin, due to the lack of iron leading to a
reduction of the incorporation of protophorphyrin into heme. As there is a
reduction in the red cell hemoglobin, the red cell continues to divide, resulting
in smaller red cells (i.e. microcytosis). Iron deficiency also results in a degree of
ineffective erythropoiesis, which results in a reduced red cell half-life (Table 3.2).
may also be symptoms and signs associated with the underlying cause. Severe
iron deficiency anemia (IDA) is associated with atrophy of the papillae, a painful
tongue, angular chelitis, nail ridging and, when severe, spooning and (with the
development of a post-cricoid web) dysphagia. There may also be an increased
risk of infection due to the effect of iron deficiency on cellular immunity and
phagocytosis. These changes, however, are extreme and are unlikely to be seen
in iron-deficient pregnancies in developed countries. Common causes of iron
deficiency in women of childbearing age are shown (Table 3.3).
As noted above, the physiological anemia of pregnancy is associated with a
normochromic normocytic picture. It should be noted, however, that a reduction
in hemoglobin concentration is a relatively late feature of iron deficiency, and is
preceded by depletion in iron stores and serum iron. Furthermore, the reduc-
tion in mean corpuscular volume (MCV), which is seen at an early stage with iron
deficiency in the non-pregnant, is not so reliable in pregnancy because of the
physiological changes in red cell mass and plasma volume. In particular, there
may be a rise in the MCV and, thus, a normal MCV does not exclude iron defi-
ciency. In pregnancy the initial appearance of IDA may therefore be nor-
mochromic and normocytic, and when there is a hypochromic microcytic
picture, the possibility of thalassemia should be considered and excluded.
Although serum ferritin contains little iron, its circulating concentration cor-
relates well with body iron stores, but the use of ferritin to detect iron deficiency
may be complicated, as it is an acute phase protein and the lower limit of the ref-
erence range is increased in the presence of inflammatory disorders. However, in
early pregnancy, reduced serum ferritin concentrations usually provide a reliable
indication of iron deficiency, but hemodilution in the second and third
trimesters of pregnancy reduces the concentrations of all measures of iron status,
and thus the ranges used in non-pregnant women are not always reliable.
Nonetheless, a low result still indicates iron deficiency in most pregnant subjects.
Dietary Vegetarian/vegan
Malabsorption Celiac
Gastrectomy
46
Obstetric CH03 18/10/05 10:21 am Page 47
With regard to other tests of iron deficiency, normal pregnancy (as noted in
Chapter 1) is associated with a progressive fall in serum iron, and an increase in
both serum TIBC and free protoporphyrin levels. Serum transferrin receptor
levels may also increase with gestational age (especially after 20 weeks gestation),
and may not fall in response to iron given at a supplemental dose. As noted
above, it appears most likely that any increase in serum transferrin receptor levels
in later pregnancy relates to increased maternal erythropoiesis. From this it is dif-
ficult to know how reliable serum transferrin receptors are in diagnosing IDA in
pregnancy. Thus, a combination of assessments should be used to assess iron
status when the diagnosis is not clear cut.
advance of any change in the red cell indices, which may reflect improvements in
the intracellular metabolic processes where iron is required.
Iron therapy may fail to replete iron stores when there is malabsorption, or
when iron lost in bleeding exceeds intake. Occasionally, no improvement is seen,
as the diagnosis of iron deficiency is incorrect. However, in the vast majority of
instances, failure to improve iron status results from poor compliance with the
iron therapy. Vitamin C can enhance absorption of ferrous iron, and this can be
achieved by taking the iron supplement with a glass of fresh orange juice or a
50mg tablet of ascorbic acid. It is the authors’ practice to routinely prescribe
vitamin C with iron supplements for treatment of IDA in pregnancy. For women
who have difficulty with swallowing tablets, liquid preparations are available, but
these can cause staining of the teeth.
There are a number of forms of parenteral iron therapy, which can be given
as a total dose intravenously (iron dextran and iron sucrose compounds) or, in
some instances (iron dextran), as multiple doses intramuscularly. The indica-
tions for parenteral therapy include malabsorption and failure of compliance
with oral therapy, and there may also be requirement when erythropoietin is
being used.
With the exception of renal failure requiring dialysis or malabsorption, par-
enteral iron probably does not produce a faster hemoglobin response than oral
iron. Despite this, parenteral iron therapy is often used when severe IDA is diag-
nosed at an advanced stage in pregnancy.
With intravenous dextran (and to some extent iron sucrose) compounds
there is a small risk of an anaphylactoid reaction (estimated at around 0.6% of
recipients), and it is recommended that a test dose of 25mg of iron should be
given over 15 minutes before a therapeutic dose is given. With intramuscular
preparations there is a risk of pain or abscess formation, skin pigmentation and
local sarcoma at the injection site. Overall, some side effects are reported in
around 25% of subjects receiving parenteral iron therapy. The majority of these
are mild, and include headache, urticaria and nausea, but serious adverse events
have been reported to occur in as many as 5% of subjects. In around 1% of recip-
ients of either preparation there is also the possibility of a delayed reaction with
myalgia, arthralgia, lymphadenopathy and fever.
through the skin and there is also an enterohepatic circulation through the
bile. Folic acid deficiency is most commonly due to dietary insufficiency associ-
ated with alcoholism and poverty. Malabsorption, due to gluten-induced
enteropathy, partial gastrectomy or Crohn’s disease may also exacerbate dietary
insufficiency, and such patients should routinely receive folate supplements
throughout their pregnancy. Other subjects at risk include those with an
increased usage of folate. This includes those with chronic hemolytic con-
ditions (e.g. hemoglobinopathy, sickle-cell syndromes, red cell membrane and
enzyme disorders, myeloproliferative disorders, and autoimmune hemolytic
anemia), those taking antiepileptic drugs (e.g. carbamazepine), and those
requiring dialysis (where there may be an increase in loss through dialysis due
to loose plasma protein binding) (Table 3.4).
Polyglutamated folate is an essential coenzyme in the transfer of single carbon
units required for the manufacture of DNA and RNA. By this, and other means,
folic acid may function to reduce oxidative potential. Folic acid, along with
vitamin B12 and vitamin B6, assists in the homocysteine (HCY)–methionine
pathway (Figure 3.1), where the presence of folic acid drives the production of
methionine from HCY, leading to a reduction in the plasma levels of HCY.
Circulating HCY is readily oxidized, and as a result may increase the genera-
–
tion of superoxide (O 2) and hydrogen peroxide (H2O2) species. These free radi-
cals can induce oxidation of low-density lipoproteins (LDL) in the endothelium,
and it has been proposed that prolonged exposure of the endothelium to HCY
results in exhaustion of the mechanisms of endothelial protection against HCY.
HCY also has an effect on a number of aspects of coagulation, which includes
upregulation of Factor (F) V generation and downregulation of the anticoagu-
lants activated protein C (aPC) and thrombomodulin.
Although most subjects are diagnosed with deficiency because of an incidental
finding of a raised MCV, folate and vitamin B12 deficiency may present with
Dietary Alcoholism
Poverty
Adolescence
Loss Dialysis
Miscellaneous Anticonvulsants
49
Obstetric CH03 18/10/05 10:21 am Page 50
B6 CBS
5 METHYL
HCY
THF
MTHFR
SAH
THF METHIONINE
SERINE GLYCINE
B6
PROTEIN
50
Obstetric CH03 18/10/05 10:21 am Page 51
Vitamin B12 Animals 3µg/day Serum vitamin B12 Gestation effect ?3µg/day 1mg
Urinary methyl- Poor correlation
malonic acid with vitamin B12
(MMA)
a neural tube defect, folic acid therapy (at a dose of at least 5mg/day) should be
given when pregnancy is planned and for the first trimester. As noted above, pro-
phylaxis is also required for subjects with chronic inherited or acquired red cell
disorders. In some such cases, there may also be a role for adding iron prophy-
laxis.
Proven folic acid deficiency anemia should be treated with folic acid therapy
at a dose of 5mg × 3/day. In all such cases of anemia, vitamin B12 deficiency must
also be excluded, as folate supplementation may improve the anemia of vitamin
B12 deficiency but exacerbate the neurological deterioration associated with it
(see below). In general, it is also good practice to check the vitamin B12 status of
all those presenting with significant folic acid deficiency. Folinic acid – a form of
folic acid which can bypass a block in the methylene tetrahydrofolate reductase
enzyme induced by methotrexate, or an excess dose of trimethoprim – can be
used to reverse folate inhibitory drugs, and is occasionally used when there is
severe folate deficient anemia from other causes.
In all women with evidence of folic acid deficiency it is important to take a
dietary history, and if the diet is lacking in folate-rich foods to provide dietary
advice.
A correlation between maternal and neonatal vitamin B12 levels has, however,
been reported. Thus, although mild vitamin B12 deficiency appears compatible with
a normal pregnancy outcome, it could result in a low level of vitamin B12 in the off-
spring (particularly if the baby is subsequently breastfed). Lower levels of vitamin
B12 have also been found in women who have had a child with a neural tube defect
and in those with early recurrent abortions. Indeed, it is thought that persisting
vitamin B12 deficiency can result in infertility, but the role of lesser deficiency in
fetal cognitive development or birthweight is not yet resolved. In countries where a
vegetarian diet is the norm, folate deficiency is less common and vitamin B12 defi-
ciency may play a more important role in the development of neural tube defects.
Macrocytosis
Reticulocytosis
Spherocytosis
Serum haptoglobin
Plasma hemoglobin
Hemosiderinuria/hemoglobinuria
Lactate dehydrogenase
Positive direct antiglobulin (Coombs’) test
55
Obstetric CH03 18/10/05 10:21 am Page 56
red cells, contain detectable RNA. If there is brisk intravascular breakdown there
will be a reduction in the level of the serum protein haptoglobin, which com-
plexes with hemoglobin to remove it from the circulation. There may also be
detectable plasma hemoglobin and evidence of hemoglobinuria. When hemoly-
sis is less marked, free hemoglobin is catabolized by the renal tubular cells to
hemosiderin, which is detectable in the urine. The enzyme lactate dehydroge-
nase is also released from damaged red cells and the hemoglobin metabolite
bilirubin is released from the reticulo-endothelial system to be conjugated in the
liver and excreted in bile into the gut. This fecal urobilinogen is reabsorbed from
the gut into the circulation and is then excreted in the kidneys, and is also
detectable in the urine.
The presence of an antibody bound to a red cell antigen can be demonstrated
by the direct antiglobulin test (the Coombs’ test) if it is IgG. If it is IgM or IgA, its
presence can be inferred by a positive antiglobulin test that has detected the
presence of complement on the red cell surface. The presence of bound anti-
body or complement does not itself indicate hemolysis, but simply that an
autoantibody is present. Antibody-mediated damage results in the loss of red cell
membrane and this leads to the formation of detectable spherocytes within the
circulation.
Warm-acting autoantibodies are most often IgG and, although they occasion-
ally have a specificity for RhC, e, Kell or RhD, they most often react equally with a
number of donor red cells in cross-matching procedures (see Chapter 2). The
consequences of such antibodies can vary from an incidental positive direct
antiglobulin test to fulminant hemolysis. The majority of clinically affected sub-
jects present with a chronic stable anemia.
More than 90% of cold antibodies have the I red cell antigen as the target,
with the remainder aimed at the i antigen. In the case of the condition known as
paroxysmal cold hemoglobinuria (classically associated with syphilis, but also
associated with viral infections), the antibodies are most likely to be of polyclonal
IgG type and specific for the P red cell antigen. In this disorder the IgG binds in
the patient’s periphery (at cooler temperatures), but then results in intravascular
lysis when it binds complement in the warmer central circulation.
AIHA in pregnancy
In women of childbearing age, cold-acting IgM antibodies are now most often
idiopathic, or are found in association with the presence of autoimmune dis-
orders or infections, such as infectious mononucleosis, measles or mumps. If
these antibodies are only active at <30°C, they are unlikely to cause significant
hemolysis.
Warm antibodies in this age group are often idiopathic, but can also be associ-
ated with autoimmune disorders, some therapeutic drugs (classically penicillin,
quinidine or methydopa), viral infections, immunodeficiency (including HIV),
lymphoproliferative disorders and other malignancies (Table 3.8). The rate of
56
Obstetric CH03 18/10/05 10:21 am Page 57
Treatment of AIHA
Drug triggers for hemolysis should be sought and any potential implicated drugs
excluded, if possible. No treatment is required for asymptomatic direct antiglob-
ulin test positivity. When treatment is required for an idiopathic AIHA, the first-
line therapy should be corticosteroids. Most often this is given as oral
prednisolone at a dose of 1–2mg/kg body weight. When a stable hemoglobin is
achieved, the steroids are reduced at a rate of 5–10mg/week until a maintenance
dose is achieved. With high doses, the mother will be at risk of steroid-related
side effects, but at maintenance doses this is not usually a problem. However, any
mother on steroid supplements should receive 150mg of hydrocortisone 8 hourly
at the time of delivery. Fetal problems are not associated with steroid therapy,
although with very prolonged use of high doses (>30mg/day of prednisolone),
fetal adrenal suppression can occur, and the pediatricians should be alerted to
this possibility.
Where steroids cannot be used, or fail, regular infusions of intravenous
human normal immunoglobulin may be used to maintain hemoglobin levels. In
57
Obstetric CH03 18/10/05 10:21 am Page 58
58
Obstetric CH03 18/10/05 10:21 am Page 59
109/l, and especially those with evidence of a platelet function defect (or
acquired von Willebrand’s disease), may be at particular risk of bleeding. Those
with a past history of thrombosis or bleeding should be considered at high risk of
another thrombotic or bleeding event. Whether there is any impact of co-inheri-
tance of thrombophilia on the occurrence of thrombosis in ET is unknown.
ET in pregnancy
Although, as noted above, the clinical course of ET is most often benign in the
young, there are reports of an association with an adverse pregnancy outcome.
The incidence of adverse effects comes predominantly from case reports or small
case series. From these, the most common complication is miscarriage (from 25
to 40%), with around a third occurring in the first trimester. The occurrence of
late fetal loss may also be increased, with stillbirth/intrauterine death reported to
occur in 5–11% of pregnancies, and there may also be an increased risk of
growth restriction and pre-term delivery. The consequences to the mother
appears more favorable, with the incidence of thrombosis at <3.5%, and most
probably less <1%, for significant vascular occlusion (cerebral venous thrombosis
or transient cerebral ischemia of arterial origin). The presence of placental
thrombosis has been reported in association with late fetal loss and bleeding has
been reported in <10% of cases, with the majority of events of a minor nature.
As with normal subjects, pregnancy is associated with a reduction in the
platelet count in subjects with ET. Whether the degree of fall in platelet count is
related to pregnancy outcome is not clear, and, in general, experience in a previ-
ous pregnancy, or the maternal pre-morbid condition, do not clearly indicate the
prognosis in the current pregnancy.
A platelet level of 1000–1500 × 109/l has been suggested as the level at which
cytoreduction is required. Despite this, it has been suggested that chemothera-
peutic agents can be safely withheld in pregnancy in subjects <30 years of age. At
present there is insufficient evidence, however, to recommend that therapy can
be safely withheld in asymptomatic individuals when a diagnosis is first made
during pregnancy. However, cytoreduction should be avoided in the first
trimester if possible.
Hydroxycarbamide (otherwise known as hydroxyurea, a ribonucleotide
reductase inhibitor) is widely used in non-pregnant subjects and has been
shown to reduce the risk of thrombosis. There is, however, the potential for fetal
teratogenicity and its safety in pregnancy, especially in the first trimester, is not
known.
Although anagrelide has been used to treat ET, it appears to be less effective
in the prevention of thrombotic events and possibly carries an increased risk of
myelofibrosis. It therefore has a very limited role in the management of ET
generally. As it also crosses the placenta, as well as the breast, and there is
limited information on its safety in pregnancy, it should not be used in this
circumstance.
59
Obstetric CH03 18/10/05 10:21 am Page 60
In the main, the decision for therapy is an individual one. In those where
chemotherapeutic agents are required to achieve cytoreduction, α-interferon
may be the drug of choice as it does not cross the placenta, although it can be
present in breast milk.
In those subjects taking hydroxycarbamide, when pregnancy is planned, α-
interferon should be substituted 3–6 months prior to a planned conception. It
has been claimed that α-interferon improves pregnancy outcome in those with a
previous poor pregnancy, but this is by no means proven.
The role of platelet-pheresis is poorly defined, as it is not clear whether it influ-
ences outcome, but it is well tolerated and may be useful when an acute reduction
in platelet numbers is required. However, multiple (often twice weekly) treat-
ments are usually required to sustain a reduction in the platelet count.
Antiplatelet agents may be used to reduce the risk of placental thrombosis
and may also assist in the prevention of maternal thrombosis. This may be
particularly useful when there are symptoms of microvascular thrombosis, but
may exacerbate any platelet defect or acquired von Willebrand’s deficiency.
Heparin may be employed as thromboprophylaxis in those subjects with a pre-
vious vascular occlusion, but its role in the prolongation of pregnancy is not
known. In addition, the necessity and timing of thromboprophylaxis in asympto-
matic subjects remains unknown.
Intravascular hemolysis
Thrombocytopenia
Leukopenia
Median survival 10–15 years
Thrombosis/bleeding/infection/leukemia/aplasia/remission
GPI anchor CD 59 on RBC/PMN
Sensitivity to complement
Washed cellular products for transfusion
Significant fetal and maternal morbidity/mortality
CD, Cluster of differentiation; GPI, Glycophosphortidylinositol; PMN, Polymorphonuclear cell; RBC; Red blood cell
60
Obstetric CH03 18/10/05 10:21 am Page 61
PNH in pregnancy
The management of PNH in pregnancy requires consideration of a number of
aspects of the disease. The thrombotic effect is likely to be due to an interaction
between white cells or platelets with complement, but may also, at least in part,
be mediated through circulating pro-coagulant microparticles, which are also
found in antiphospholipid syndromes. Thus, there may be similar processes and
consequences in both disorders.
Therapeutic anticoagulation is needed in those who have previously experi-
enced thrombosis. The role of thromboprophylaxis in those who have not yet
had a thrombosis is not known, but we would recommend this in all cases. As
with other disorders requiring anticoagulation, this is best achieved in pregnancy
with therapeutic or prophylactic dose, low-molecular-weight heparin (LMWH)
antepartum, and with warfarin or continued LMWH (avoiding the need for inter-
national normalized ratio (INR) monitoring) in the puerperium.
PNH is also associated with thrombocytopenia, and there may be an exaggera-
tion of the normal thrombocytopenia of pregnancy. It has been suggested that
the platelet count should be maintained at >30 × 109/l throughout pregnancy.
61
Obstetric CH03 18/10/05 10:21 am Page 62
This may be difficult to achieve and will require washed platelet component
therapy to reduce the risk of exposure to exogenous complement. There is little
evidence that this is required and in the absence of bleeding problems and with a
platelet count >10–20 × 109/l, the present authors would manage the patient con-
servatively. As with other conditions (see Chapter 8), an adequate platelet count
is, however, required for delivery or Cesarean section, thus washed platelet
preparations are best reserved for delivery.
Epidural analgesia may be problematic from both thrombocytopenia and anti-
coagulant viewpoints, and so may be unavailable to such women. In all events, a
planned delivery will most often be required to ensure adequate hemostasis.
Anemia is common, and also requires the provision of washed red cell com-
ponents. The development of severe anemia may also have a bearing on fetal
oxygenation and development. Following delivery, the combined oral contracep-
tive must be avoided because of the thrombotic risk.
The maternal mortality may be of the order of 20% in cases of PNH during
pregnancy. Pregnancy may be associated with a substantial requirement for
maternal platelet and red cell component therapy, and an increased risk of both
venous thrombosis and infection. Although successful pregnancies have been
reported, the fetal mortality may be as high as 10%, with 50% delivering pre-
term, and there is also an increased risk of fetal growth restriction in those
mothers with severe anemia.
There may be few late effects on children exposed to chemo- or radiotherapy in utero
AML, Acute myeloid leukemia; IUGR, Intrauterine growth restriction; NHL, Non-Hodgkin’s lymphoma.
Lymphoma in pregnancy
The co-occurrence of lymphoma and pregnancy is relatively rare (Table 3.11). As
with the treatment of acute leukemia, management should consider the maternal
wishes, the type of disease, the likelihood of cure, the gestation at diagnosis, the
possibility of fetal effects and the possibility of collection of ovarian material for
storage. In the vast majority of lymphomas there is no evidence that the lym-
phoma itself influences pregnancy, or that the pregnancy has an adverse influ-
ence on the lymphoma.
When Hodgkin’s disease presents in the first trimester, a number of options
should be considered. Where there is aggressive disease, therapy may be required
immediately and the possibility of termination should be discussed. When, as is
more common, there is a lower grade of disease there are a number of possible
63
Obstetric CH03 18/10/05 10:21 am Page 64
therapeutic options. These include monitoring the disease without therapy, the
use of single agent therapy (such as vinblastine) to control the disease until
definitive treatment is considered appropriate, use of a modified chemothera-
peutic regimen, full therapeutic combinations, or radiotherapy (where appropri-
ate) with fetal shielding. Non-Hodgkin’s lymphoma (NHL) usually presents later
and often with more aggressive disease. In most cases, immediate standard-dose
therapy is required and should not be delayed. However, if either NHL or
Hodgkin’s disease presents in late pregnancy, there may be scope to withhold
treatment until safe delivery can be achieved.
There is no suggestion that diagnostic radiation or nuclear diagnostic tests
themselves should be an indication for termination. Indeed supradiaphragmatic
irradiation, including spiral computerized tomography (CT) scanning, does not
expose the fetus to a significant radiation dose and is considered safe for the
fetus. Indeed, despite the caution expressed about chemotherapy (especially
when given in the first trimester), long-term follow-up of offspring of those who
received treatment for acute leukemia, Hodgkin’s disease or NHL, does not show
any evidence of an adverse effect on intellectual development, growth or the
occurrence of malignancy.
In subjects who have been previously treated for lymphoma there is an
increased risk of ovarian failure and premature menopause. This is particularly
the case in women treated after the age of 25 years with a combination of
chemotherapy and infradiaphragmatic radiotherapy. However, even those who
have undergone high-dose regimens may conceive (with hormone replace-
ment) with a successful outcome. Abdominal radiation affecting only a part
of the uterus does not preclude pregnancy, but when radiotherapy has
involved the uterus, early delivery and growth restriction may occur in sub-
sequent preg-nancies. At present, the significance of these observations is not
clear. After radical radiotherapy the uterus is unlikely to support conception,
even if there is satisfactory ovarian function. In general, after treatment for
lymphoma, patients are advised to wait 1–2 years before attempting pregnancy.
Whether this reduces risk is not clear, but appears sensible in the context of
possible relapse.
Ethics/consent
Viral safety
Genetic disorder exclusion
Microbial safety
Traceability
Storage safety and records
Dose suitable for pediatric cases
? Graft rejection
? GVHD
The criteria for the selection of maternal donors for allogenic donation are
similar to those employed by most blood banking organizations, but with addi-
tional information required regarding past obstetric history and the possibility of
transmission of genetic defects. A number of practical issues, such as the safety
and yield of cells obtained when the cord is clamped early, or when the collec-
tion is taken prior to delivery of the placenta, are yet to be resolved. In addition,
the effect of Cesarean, rather than vaginal, delivery on the quality of cells also
requires further study. Overall, the collection appears to have little impact on
staff time, with often only an additional 10 minutes required for such collections.
The testing of such donations covers similar areas to that of blood donation,
including statutory virological screening (including hepatitis A, B and C, and
HIV, human T-cell leukemia/lymphoma virus (HTLV) and cytomegalovirus
(CMV)) and microbiological culture. The problem of window period infections
and the absence of the safety net that is presumed to come from those who have
successfully donated before, may all have an impact on the implementation of
such placental banking programs.
In addition, there is the requirement (as with hematopoietic stem cells derived
from adults) to determine the viability (from progenitor cell culture) and stem cell
component of each collection. It has been supposed that the human leukocyte
antigens (HLA) match may be less stringent with cord stem cells, although further
work on this, and the exclusion of maternal cells from the collection, is required.
At present, cord banking remains an expensive source of hematopoietic stem
cells, which, like cells derived from adults, requires a substantial infrastructure to
maintain safety and traceability. To date, a substantial number of pediatric trans-
plants have been carried out using cord stem cells, although much work is
required to allow these collections to form the basis of transplants for adults.
Ultimately, the expense of the collection and storage procedures may be offset by
the greater amount of transplant material available from this source and (if con-
firmed) the requirement for less stringent HLA matching and lesser likelihood
of graft-versus host disease with such transplants.
65
Obstetric CH04 18/10/05 10:22 am Page 66
CHAPTER 4
Bleeding disorders in
pregnancy
Hemophilia A
The gene coding for Factor (F) VIII is found on the X chromosome at position
q28, and heritable deficiency of FVIII occurs as an X-linked recessive disorder.
The frequency of hemophilia A is between 1 in 10 000 and 1 in 20 000 of the
population. Although it has a wide distribution amongst different ethnic groups,
it is rare in Chinese populations. The disorder results from a variety of genetic
mutations, although 40% of severely affected individuals have an inversion
involving intron 22 of the gene. By contrast, the majority of mild disease results
from point mutations.
Clearly, this is a disorder predominantly affecting males; females will usually
be heterozygous and therefore can only be carriers. For a karyotypically normal
woman to have a hemorrhagic problem with hemophilia A, she would have to be
either homozygous for the mutation (with inheritance of affected X chromo-
somes from both an affected father and a carrier mother – an exceptionally rare
occurrence), or demonstrate significant lyonization (see below), with resulting
low FVIII levels. The latter is not unusual in carriers, but virtually all carriers in
this situation have levels of FVIII above that which would result in spontaneous
bleeding problems. Moreover (see Chapter 1), the vast majority of such reduced
FVIIIc levels will normalize during pregnancy. Other mechanisms of clinical
FVIII deficiency in a female include an acquired antibody to FVIII, a hemophilia
carrier with Turner’s syndrome (XO), and autosomal dominant hemophilia A
(due to a somatic gene affecting FVIII synthesis).
Hemophilia A tends to follow a similar pattern in affected family members,
reflecting the particular genetic mutation associated with hemophilia A in their
family, although in 30% of cases there may be no known family history. Daugh-
ters of an affected male are obligate carriers of the disease. Most of these will be
unaffected unless there is extreme lyonization. Lyonization is the random inacti-
vation of the X chromosome that occurs in all cells. If, by chance, this inactiva-
tion affects more of the normal X chromosome in FVIII-producing cells, this will
produce a mild hemophilia state in the affected female carriers. A carrier female
has a 50:50 chance of passing hemophilia onto her son and a 50:50 chance of
passing the carrier state onto her daughter.
66
Obstetric CH04 18/10/05 10:22 am Page 67
PT √ √ √ √ √/
APTT /√ /√ /√ Mild /√
TCT √ √ √ √ √
FVIIIc √ √ Mild –
FIXc √ √ √ –
FXIc √ √ √ –
vWF √/ √ √ –
APTT, Activated partial thromboplastin time; F, Factor; PT, Prothrombin time; TCT, Thrombin clotting time; vWF,
von Willebrand’s Factor.
67
Obstetric CH04 18/10/05 10:22 am Page 68
be a disparity between antigen and activity (i.e. activity that is less than antigen
level), indicating the presence of a functional genetic defect. In most severe cases
the only likely alternative diagnosis is either hemophilia B (FIX deficiency) or C
(FXI deficiency). Thus, the initial diagnostic work-up should include an assay of
FVIIIc, FIXc and FXIc. In milder cases, von Willebrand’s disease (vWD) may also
have to be excluded (Table 4.1). When the disease presents with the incidental
finding of a prolonged APTT, a specific factor inhibitor (see below) or a lupus
inhibitor (lupus anticoagulant) may have to be excluded.
Factor replacement
Replacement with factor products in hemophilia A should be achieved with
recombinant or high purity FVIII preparations where possible, with a dosage
based upon the plasma concentration of FVIII required and upon the body
weight or plasma volume. To calculate the required dose by weight the following
formula can be used: (body weight [kg] × desired rise in FVIII [IU/100ml])/2.
The amount of FVIII administered is expressed in IU: 1IU of FVIIIc equals the
quantity of FVIIIc in 1ml of normal human plasma. Empirically, 1IU of FVIII/kg
of body weight increases the plasma FVIIIc level by 1.5–2.0IU/100ml. After each
dose a peak level should be checked and further doses given, if required, to
maintain the target level (Table 4.2). When there has been bleeding, dose calcu-
lation by plasma volume may be more appropriate. When required, the plasma
volume can be derived from the patient’s hematocrit and their blood volume
(which can be estimated as 7% of the body weight). For most clotting factors, the
decline of therapeutic levels occurs in two phases: a rapid loss followed by a
slower decline. For FVIII infusions, around 80% of the dose can be recovered ini-
tially in the plasma, leading to an initial and subsequent half-life of infused FVIII
of 6 and 12 hours, respectively. This leads to a requirement to dose every 8–12
hours.
Because of repeated exposure to allogenic FVIII, the development of acquired
inhibitors to FVIII is not uncommon in hemophilia A. In such circumstances,
cross-reactivity of the antibody with porcine FVIII should be determined. Where
there is little or no cross-reaction, porcine FVIII products can be used. This
68
Obstetric CH04 18/10/05 10:22 am Page 69
Table 4.2 – Factor (F) VIII dosing in hemophilia A (HA) and hemophilia B (HB)
Antifibrinolytic agents
For minor surgery, antifibrinolytic agents (such as tranexamic acid) can be a
useful adjunct to other therapy, and may be particularly useful in dental surgery.
Tranexamic acid binds to plasminogen, inhibiting the binding of plasminogen to
fibrin. This leads to a reduction in fibrinolysis, and has a plasma half-life of
around 2 hours. It should be given in advance of any procedure and can be given
as an oral dose, or as a mouthwash for dental procedures.
Renal tract bleeding and/or renal failure are contraindications to such thera-
pies, as thrombosis in the renal tract may further impair renal function. Tranex-
amic acid should also be used with caution in pregnancy, as fibrinolysis is already
shut down by the physiological increase in plasminogen activator inhibitor (PAI)-
1 and PAI-2 (see Chapter 1).
The technology of assisted conception now offers the opportunity for pre-
implantation genetic diagnosis, so avoiding the need for termination. Following
ovulation induction and transvaginal ultrasound-guided oocyte retrieval, in vitro
fertilization is performed, and single-cell biopsy at the 4–8 cell stage allows deter-
mination of the embryonic sex (usually by fluorescent in situ hybridization). This
allows the potentially affected developing male embryos from a carrier mother,
or the potential carrier female embryos of an affected male father, to be dis-
carded and only unaffected embryos replaced, so avoiding the birth of an
affected male or carrier female. The success rate for pregnancies following this
procedure is consistent with other assisted reproduction techniques, and cur-
rently is of the order of 25–30%. There appears to be no developmental problem
with the embryo when biopsied at this stage. This technique is now available in a
limited number of centers in Europe and North America. In the future, more
precise genetic testing on single cells may become more readily available, which
will allow determination of whether the embryo carries the particular hemophilia
gene for that family.
73
Obstetric CH04 18/10/05 10:22 am Page 74
Hemophilia B
Hemophilia B is another X-linked bleeding disorder, which, although consider-
ably less common than hemophilia A, is more often associated with a family
history. Hemophilia B may result from mutations that lead to a reduction in FIX
function, accompanied by either detectable, or reduced, FIX antigen levels. The
wide variety of mutations of the FIX gene includes those that alter carboxyl
group formation (carboxyl groups on vitamin K-dependent factors lead to
binding of the coagulation factor to activated phospholipid at the site of coagula-
tion, accelerating the coagulation response) and those that alter the activation of
FIXc by FXIa. In addition, a mutation in the FIX promoter region may lead to a
marked reduction in antigen at birth, but increasing production occurs with
increasing age (hemophilia B Leyden).
Severe FIX deficiency is considerably less common than severe hemophilia A,
but presents with similar problems as hemophilia A subjects with comparable
factor levels. The diagnosis of hemophilia B can be achieved by determination of
FIXc in an APTT-based assay.
Pregnancy management
There is substantial genetic heterogeneity in cases of hemophilia B, with many
mutations being peculiar to an individual family. Confirmation of female car-
riage requires knowledge of the mutation within the family to confirm the diag-
nosis. In general, to be certain that an identified mutation causes hemophilia, it
is recommended that the clinician confirms that the mutation is already included
in one of the international databases of hemophilia mutations. This is necessary,
as a number of mis-sense mutations result in no observable change in the func-
tion of the FIX molecule. Furthermore, where there is a sporadic case, the
mother may have a mosaic (a mixture of native and mutated DNA in the same
germ cell) and carrier status cannot be confirmed by assessment of her somatic
cells.
Female carriage is likely if there is a reduced level of FIX activity (which often
results in a more clear-cut distinction from non-carriers than FVIII carriage in
hemophilia A), but assessment of FIX antigen levels is more problematic, as
there may be a considerable overlap with non-hemophilic subjects. Although FIX
activity is higher in women on the combined oral contraceptive pill, it does not
change substantially with increasing gestation, and those carriers with a low level
tend not to ‘nomalize’ in pregnancy, in contrast to hemophilia A carriers.
All carriers should have an assessment of FIX levels at their first visit in preg-
nancy and in the third trimester. In more markedly affected carriers, hemostatic
support is more likely to be required during pregnancy than with equivalent
hemophilia A carriers. Such support should be with recombinant factor prod-
ucts, as DDAVP is usually ineffective in FIX deficiency. As with FVIII carriers, a
FIX level of at least 40 U/dl is required for a vaginal delivery, and a level of at
least 50 U/dl for Cesarean section or epidural. Levels should be maintained for
the first 4–5 days after delivery.
The methods of prenatal diagnosis of FIX deficiency, and the management of
the delivery and the postpartum phase, follow the same principles and tech-
niques as those used for cases of FVIII deficiency. To facilitate restriction frag-
ment length polymorphism (RFLP) detection, Taq1, Xmn1, Dde1, Hha1, Mnl1
and Mse1 are informative in around 90% of cases in Caucasians.
Hemophilia C
Hemophilia C is characterized by a reduction in coagulation FXIc. Unlike hemo-
philia A and B, it is inherited as an autosomal recessive trait. In homozygotes, the
FXIc level may be <20 U/dl: in heterozygous subjects the levels range between 20
75
Obstetric CH04 18/10/05 10:22 am Page 76
and 70 U/dl. The disorder has a prevalence in the general population of around
one in one million, but occurs at a higher frequency in the Ashkenazi and Iraqi
Jew populations, where up to 0.3% of subjects are homozygous for the deficiency
and there is a gene prevalence of up to 11%.
The gene coding for FXI is found on chromosome 4, with at least 28 known
mutations. These can result in an alteration of gene splicing (Type I mutations),
the production of a stop codon (which leads to FXIc of <1U/dl), or a minor
amino acid substitution (Type III mutations). Of these, mutations associated with
the production of a stop codon (so-called Type II mutations) are most often asso-
ciated with bleeding. In the Ashkenazi Jew population, two mutations are
responsible for most cases, and compound Type II/Type III heterozygotes are
the commonest cause of moderate to severe disease. In other populations there
is a greater variation in the mutations involved.
Unlike hemophilia A and B, bleeding is more difficult to predict in subjects
with hemophilia C. Generally, hemarthrosis is uncommon and spontaneous
bleeding is rare. Furthermore, bleeding does not correlate well with residual FXI
activity and around 50% of heterozygous subjects may have a bleeding tendency.
By contrast, subjects with severe deficiency may not have a severe bleeding tend-
ency. In many affected individuals bleeding only occurs following surgery or
trauma. The possibility of delayed hemorrhage should be considered, especially
in oral, ear, nose and throat (ENT), gynecological and urological surgery, where
there is high local fibrinolytic activity.
FXI deficiency may prolong the APTT, although this is only likely to detect
homozygous subjects. In all suspected cases, a FXIc assay should be performed.
Management in pregnancy
As with other heritable bleeding disorders, pregnant subjects should be managed
in a center with expertise in the assessment and treatment of hemophilia.
There are a variety of studies that show no change in FXIc levels with gesta-
tion in normal individuals, although many show a gradual reduction. As there is
no good correlation between FXIc levels and bleeding in hemophilia C, close
monitoring of FXIc levels in pregnancy in hemophilia C subjects is of doubtful
value in any case.
Postpartum hemorrhage occurs in 20% of homozygotes, but excessive bleed-
ing may also occur in heterozygotes. In general, the majority of subjects with a
non-pregnant FXIc level of <15U/dl are likely to suffer excessive bleeding after
trauma or surgery, and any subject who has a history of excessive blood loss will
require hemostatic support for delivery. Even if specific hemostatic support is
thought not to be required, products should be available.
When therapy is needed, daily use of a FXI concentrate is probably justified,
with the aim of achieving a target FXIc level of no >70IU/dl (maximum dose of
30IU/kg). This level should be achieved during labor and maintained for 3–4
days after vaginal delivery and 4–5 days after Cesarean section.
76
Obstetric CH04 18/10/05 10:22 am Page 77
The target level of FXIc should not exceed 100U/dl, as this has been associ-
ated with thrombosis, although it is not known whether there is an excessive risk
of thrombosis whilst using FXI concentrates in late pregnancy and the puer-
perium. Although many manufacturers have added heparin to FXI concentrates,
since the early reports of thrombosis, it is not yet clear that this has reduced the
thrombotic potential of these products. In subjects with a history of thrombosis
or atherosclerosis, FXI concentrate should probably be avoided. If hemostatic
support is required, then fresh frozen plasma (FFP) should be used (FFP should
be virally inactivated if at all possible).
If there has been no history of bleeding, tranexamic acid could be used to
cover delivery in the first instance. However, as noted earlier, fibrinolysis is
already impaired in pregnancy and tranexamic acid should be used with
caution in view of the potential for thrombosis. There should be early recourse
to FXI concentrate, if available, when treatment is needed. While FFP is an
alternative, the use of FFP in such circumstances is complicated by the relat-
ively large volume required and the variable half-life of the coagulation factors
within it. Similarly, if, in an emergency, FXI products are not available, then
FFP should be used if hemostatic support is required. The role of recombinant
FVIIa is not yet certain, although it has been used effectively in subjects with
FXI inhibitors, and it may be preferable to the use of human blood products in
pregnancy.
If regional anesthesia is required, correction of the APTT has been advocated,
although this remains controversial. As with any other hemophilia, the possibility
of fetal carriage should be considered when planning fetal monitoring, forceps
and ventouse extraction at delivery.
Acquired hemophilia
Acquired hemophilia results from the development of an autoantibody, most
often against FVIII. Although classically associated with repeated treatment with
allogenic factor products in subjects with hemophilia, it can also occur in normal
healthy individuals, in subjects with autoimmune disorders (such as systemic
lupus erythematosis (SLE) and rheumatoid arthritis (RA)), in those with lympho-
proliferative and solid tumors, as well as in association with pregnancy. In non-
hemophilia subjects it is a rare and sporadic disorder, and the incidence has
been estimated at between 0.2 and 1 per million person-years. Of these, around
10% occur in relation to pregnancy.
Presentation in pregnancy
Pregnancy-related cases are often diagnosed within 3 months of delivery,
although presentation up to 1 year after delivery has been reported. Presentation
varies from bleeding after surgical procedures to spontaneous bruising, wide-
spread hematomas and GI bleeding. If untreated, acquired hemophilia carries a
77
Obstetric CH04 18/10/05 10:22 am Page 78
Treatment in pregnancy
The management of these patients requires consideration of both the level of
inhibitor and the clinical problem. In those with a high level of inhibitor
(Bethesda >10U), the therapeutic options include porcine FVIII, recombinant
FVIIa (rFVIIa), or activated clotting factor concentrates (e.g. FEIBA®) (Table
4.3). The potential value of porcine FVIII can be assessed by laboratory determi-
nation of cross-reactivity of the inhibitor with porcine FVIII. With low-level
inhibitors, spontaneous resolution without immunosuppression often occurs. In
some cases there may be no response to high-dose FVIII, even when a low titer of
inhibitor is present: indeed, even high-level inhibitors can sometimes respond to
normal doses of FVIII.
The 2-year mortality has been estimated at 3%, with complete resolution of
the antibody in almost all survivors by this time. Indeed, commonly, resolution
occurs 6 weeks to 6 months after presentation. The use of immunosuppression
may shorten the duration of the disease, but it is not yet clear whether it influ-
ences mortality. From the little information that is available, it appears that there
is unlikely to be a recurrence in a future pregnancy. If a recurrence does occur,
antepartum bleeding appears to be uncommon. In the vast majority of such cases
the fetus is not affected, although sporadic case reports of low FVIII levels in the
infant of an affected mother and of intracranial hemorrhage have been
reported.
hours, leaving FVIIIc levels of between 1 and 10IU/dl. vWD results in a reduction
of vWF and is the most common heritable bleeding disorder, with a frequency
that has been estimated at around 1% of many populations. Many mild cases
remain undiagnosed, as subjects are often asymptomatic.
The gene coding for vWF is found on chromosome 12, and the vWF molecule
is produced both in the megakaryocyte (the precursor of platelets) and in the
endothelium. Upon manufacture, it is stored in the Weibel-Palade bodies of the
endothelium and in the α granules of platelets. Upon release into the circula-
tion, vWF is cleaved by a proteinase of the A disintegrin and metalloprotease with
thrombospondin domain (ADAMTS) family to achieve the correct balance of
vWF multimers (see Chapter 8).
A brief description of the various types of vWD is shown in Table 4.4. The vari-
ants range from a mild/partial deficiency to a severe deficiency; between these
two extremes are other variants. Some reduce the interaction of vWF with
platelets (associated with either reduced or normal levels of high-molecular-
weight vWF multimers). One produces a variant vWF that has an increased affin-
ity for platelets. This, somewhat paradoxically, results in bleeding associated with
thrombocytopenia and increased clearance of high-molecular-weight vWF multi-
mers. There is also a vWD variant that has a reduced interaction with FVIII: in
this variant the reduction in the half-life of FVIII contributes to the bleeding phe-
notype.
79
Obstetric CH04 19/10/05 11:08 am Page 80
Table 4.4 – Classification of von Willebrand’s disease (vWD)
vWF affinity to platelet Aut dom √/ √/ √/ High molecular weight
glycoprotein 1b High- ( Platelet
80
Aut dom, Autosomal dominant; Aut rec, Autosomal recessive; F, Factor; RiCoF, Ristocetin cofactor; vWF, von Willebrand Factor.
Obstetric CH04 18/10/05 10:22 am Page 81
cytopenia, which often worsens in pregnancy and may fall in response to DDAVP.
In such cases DDAVP should probably be avoided. In subjects who do not
respond to DDAVP, treatment with a plasma-derived factor concentrate that
contains both FVIII and vWF is required (an ‘intermediate purity’ FVIII concen-
trate). A rough guide to dosing is shown in Table 4.5. The adequacy of the dose
can be assessed by monitoring either FVIIIc or vWF RiCoF levels. In some Type 2
vWD, RiCoF assessment is more useful, as the baseline FVIIIc is within reference
limits. There is no practical role for monitoring the bleeding time, as it reflects
platelet vWF rather than vWF in the plasma.
As noted above, tranexamic acid may be a useful adjunct to other therapy,
and in particular has a role in dental procedures. Although cryoprecipitate con-
tains up to 10 times more FVIII and vWF than FFP, unlike concentrates it under-
goes no virucidal procedures, and the exact amounts of factor contained in each
dose is unknown. Correspondingly, it should only be used in an emergency when
a suitable intermediate FVIII concentrate cannot be obtained.
Where methylene blue (MB)-treated cryoprecipitate is available, this should
be used in preference to an untreated version. MB is a cross-linking agent that
destroys some of the known blood-borne viruses. In most recently developed
products, >95% of the added MB is removed during product manufacture. The
potential for MB treatment to reduce the level of FVIII and vWF in the product
should, however, be borne in mind when dosing this product. In cases of Type 3
vWD that do not respond to vWF concentrates, a combination of vWF concen-
trate and platelet component therapy may prove useful.
Antenatal management
All women with vWD should be managed in a unit with rapid access to specialized
hemophilia care. If the fetus is at risk of Type 3 vWD, prenatal diagnosis may be
Table 4.5 – Intermediate Factor (F) VIII dosing in von Willebrand’s disease (vWD)
required. This requires planning to determine the mutation or RFLP, which may
be informative. Although vWF levels increase with gestation, it is important to
assess vWF and FVIIIc levels between 34 and 36 weeks gestation in all women with
vWD, to confirm that the levels have increased sufficiently to allow labor and deliv-
ery without the need for DDAVP or blood products. In subjects with mild–moder-
ate vWD (Types 1 and 2), vWF levels usually increase from the 6th to 10th weeks
of gestation, increasing 3–4-fold by the third trimester, making antenatal therapy
(as with hemophilia A carriers) unnecessary in most cases. Indeed, it appears that
significant antepartum hemorrhages and miscarriages are no more common in
such subjects. However, as with hemophilia A, early pregnancy complications may
require specific treatment to improve vWF levels.
As discussed above, DDAVP, if required, has no significant oxytocic effects, is
not associated with teratogenicity in animal studies and has been used in early
pregnancy to permit CVS or amniocentesis. In Type 3 vWD there is no rise in
vWF with gestation or DDAVP, and factor therapy may be required to cover inter-
ventional procedures or spontaneous bleeding. As noted above (page 70),
tranexamic acid is not absolutely contraindicated in pregnancy and is not associ-
ated with teratogenicity.
Management of delivery
A vWF or FVIII activity level of >40IU/dl or >50IU/dl, respectively, is con-
sidered safe for vaginal delivery or Cesarean section (or other procedures such
as amniocentesis or evacuation of the uterus). After delivery, the levels of vWF
rapidly return to pre-pregnancy levels and so, in general, vWD is associated
with a higher risk of primary and secondary postpartum hemorrhage. However,
in most Type 1 and 2 vWD, prophylactic therapy for vaginal delivery is not
needed, although treatment may be required for episiotomy or perineal tears
in Type 2 disease. In Type 1 vWD, epidural analgesia should not be undertaken
lightly, but with due regard to the risks of spinal hematoma. In those who are
known to respond to DDAVP, this can be used to cover Cesarean section.
Although there maybe a theoretical risk, it is not clear whether DDAVP use at
delivery carries any increased risk of fetal hyponatremia. If this is a concern,
then restricting use until after clamping the placental cord will avoid such a
problem. In Type 1 vWD there will be a fall in vWF after delivery, and discharge
planning in women with symptomatic disease should take account of the possi-
bility of secondary postpartum hemorrhage (PPH) at 4–5 days after delivery.
In any case, vWF and FVIIIc levels should be monitored in all cases in the first
days after delivery. After Cesarean section, DDAVP or factor products (if they
have been used) should be continued for at least the first 2–3 days of the
puerperium.
Type 3 vWD is, however, often associated with intrapartum and postpartum
hemorrhage. FVIIIc levels at delivery have been used as a guide to the necessity
for therapy, although the prediction of bleeding (as with any delivery) is
83
Obstetric CH04 18/10/05 10:22 am Page 84
Afibrinogenemia
Although afibrinogenemia, usually an autosomal recessive trait, can be associated
with life-threatening hemorrhage, bleeding is usually less than that observed in
hemophilia A and B. Bleeding may occur, however, from the umbilical stump
and bleeding into joints is not uncommon, but bleeding from the GI tract and
the central nervous system (CNS) is infrequently seen.
In affected mothers, there may be an increase in the risk of first trimester mis-
carriage, placental abruption and postpartum hemorrhage. In such circum-
stances, maternal fibrinogen replacement is often required for the pregnancy to
proceed to term.
As fibrinogen is the final component of hemostasis, congenital afibrinogene-
mia can be suspected from a failure to clot in the prothrombin time (PT), APTT
and thrombin clotting time (TCT). This is accompanied by an absence of fib-
rinogen antigen and, in some cases, a prolonged bleeding time.
When treatment is required, this should be achieved by a virus-inactivated fib-
rinogen concentrate where it is available. Where it is not, cryoprecipitate may
have to be used (with the caveats noted above). Prophylaxis is indicated in preg-
nancy and fibrinogen levels should be restored to at least 1g/l. In afibrinogene-
mia, exposure to allogenic fibrinogen may result in allergy or anaphylaxis, and
restoration of fibrinogen levels has also been associated with the development of
thrombosis.
84
Obstetric CH04 18/10/05 10:22 am Page 85
delay in wound healing and has been associated with a high risk of recurrent mis-
carriage.
Diagnosis
In suspected cases, the PT, APTT and bleeding time will be normal. If suspected,
a test to detect an abnormal solubility of fibrin can be used as a screening test for
the disorder. This is carried out by clotting an anticoagulated patient sample with
thrombin and calcium. The clot is then exposed to a chemical such as urea or
acetic acid. Of these, acetic acid may have a greater sensitivity to milder defects
(FXIII <10IU/dl) than urea (FXIII <5IU/dl), and is probably the more useful
screening test. Such tests should be confirmed by a specific assay of FXIIIc levels.
There are a number of FXIIIc assays available, although there is a great variation
in their performance between different laboratories; an FXIII antigen assessment
by ELISA is also available in some areas. Despite these variations, such tests will
be of use in monitoring the response to therapy.
Management in pregnancy
It has been reported that up to 50% of pregnancies in those with severe FXIII
deficiency will result in miscarriage without therapy. It is, therefore, usual prac-
tice for all severely affected women who conceive to receive monthly infusions to
maintain a FXIII trough level at >3IU/dl from the diagnosis of pregnancy.
As affected neonates may present with persistent bleeding from the umbilical
cord, and are at risk of intracerebral bleeding, close collaboration is required
with neonatologists to rapidly diagnose and treat infants presenting with unex-
plained hemorrhage in the presence of a normal coagulation screen and platelet
count. This is particularly relevant if there is known to be consanguinity in the
parents.
86
Obstetric CH04 18/10/05 10:22 am Page 87
87
Obstetric CH05 18/10/05 10:22 am Page 88
CHAPTER 5
Antithrombotic therapy
in pregnancy
In pregnancy, it is critical to consider the effects of any drug, not only on the
mother but also on the developing baby, e.g. teratogenesis in the first trimester,
developmental problems in the second and third trimesters, delivery problems,
and also the potential for longer term effects on childhood growth and develop-
ment. In this regard, particular concerns exist in relation to anticoagulants and
antithrombotic therapies in pregnancy, and a sound understanding of the effi-
cacy and safety of these agents is essential in order to provide the optimal man-
agement of thrombotic problems in pregnancy.
Coumarins
Of the coumarin group of drugs, warfarin is commonly employed in a large
number of countries. Warfarin is an orally active 4-hydroxycoumarin, which
inhibits the synthesis of the vitamin K-dependent clotting and anticoagulant
factors (see Table 5.1). This includes the coagulation factors (F) II, VII, IX and
X, and the natural endogenous anticoagulants protein C and protein S. War-
AF, Atrial fibrillation; ArtT, Arterial thrombisis; DVT, Deep vein thrombosis; F, Factor; ICH, Intracerebral
hemorrhage; INR, International normalized ratio; IQ, Intelligence quotient; ISI, International sensitivity index;
PC, Protein C; PS, Protein S; PT, Prothrombin time; PTc, Control PT; VTE, Venous thromboembolism.
88
Obstetric CH05 18/10/05 10:22 am Page 89
farin reduces the carboxylation of these factors, which renders them unable to
bind to catalytic phospholipid, which in turn is required for successful coagula-
tion (see Chapter 1). Warfarin has a long half-life, but its effect reflects the
half-life of the vitamin K factors. When warfarin therapy is commenced there is
an initial reduction in the levels of both FVIIc and protein S that is evident
within around 6 hours, reflecting the short half-life of these factors. Indeed, it
is this rapid fall in FVIIc that makes the prothrombin time (PT) useful in pre-
dicting the outcome of acute liver injury. However, on account of the rapid fall
in protein S, warfarin, when used alone, may induce a pro-coagulant imbalance
in the first days of therapy. This may lead to microvascular thrombosis and skin
necrosis. The risk, however, appears to apply only to those subjects who have
suffered thrombosis (when there may be consumption of coagulation and anti-
coagulant factors, and perhaps an underlying thrombophilia), but does not
extend to those receiving warfarin as prophylaxis for atrial fibrillation. For this
reason, heparin therapy should overlap with warfarin in the treatment of
thrombosis until the international normalized ratio (INR: see below) is >2. The
other vitamin K-dependent coagulation factors have considerably longer half-
lives.
Warfarin is highly bound to albumin and is itself metabolized in the liver.
The dose of warfarin given depends upon its measured effect on the PT (see
Chapter 1), which is often reported as an INR. The INR compares the patient’s
PT (in seconds) with that of a normal pool of plasma. These clotting times are
examined simultaneously by clotting the anticoagulated plasma samples with
tissue factor (TF) and calcium. A ratio of the patient:normal pool plasma clot-
ting time is generated. To allow inter-laboratory and international compar-
isons, the TF used should have a known sensitivity (called the international
sensitivity index or (ISI)). To generate the INR, the patient:control ratio is
then logged to the power of the ISI, e.g. for a thromboplastin with an ISI of
1.1, the INR would be equal to the (patient PT:control PT)1.1. This allows the
comparison of PT generated in different laboratories using different TF. In
general, the nearer the ISI is to one, the more precise and safe the INR assess-
ment will be.
A number of factors can interact with warfarin, resulting in an enhanced or
reduced effect (Table 5.2). As a general rule, an INR of 2–3 is the aim for treat-
ment of deep vein thrombosis (DVT), or for the prophylaxis of atrial fibrillation
(often with a target INR of 2.5), and an INR of 3–4 (often with a target of 3.5) is
the aim for the management of arterial thrombosis, mechanical heart valves or a
venous thrombosis that has occurred despite an INR of 2–3.
On account of the individual variability of warfarin dosage, long-term war-
farin carries a not insubstantial risk of bleeding, which after 6 months of treat-
ment for a first DVT can be equivalent to the risk of DVT recurrence. This is
why anticoagulation for DVT is restricted to 3–6 months of therapy, as after this
time, in most cases, the risk of anticoagulation outweighs the risk of recurrent
thrombosis.
89
Obstetric CH05 18/10/05 10:22 am Page 90
Warfarin in pregnancy
Warfarin crosses the placenta and inhibits coagulation factor production by the
fetal liver. It is also teratogenic, but is not found, to any significant degree, in
breast milk and is therefore safe to use in breastfeeding mothers. In the first
trimester, exposure to warfarin may result in warfarin embryopathy. The fetus is
vulnerable between 6 and 9 weeks gestation, and the incidence has been esti-
mated at around 6% of pregnancies exposed to warfarin in the first trimester.
The effect appears to be dose (not INR) dependent, with an increased risk
evident with does >5mg/day. Warfarin embryopathy can be avoided when war-
farin is substituted by heparin between 6 and 12 weeks gestation. There is also,
however, an association between warfarin usage and miscarriage, and around
25% of women taking warfarin throughout pregnancy will miscarry. Similar
figures are seen even when heparin is substituted in the first trimester. However,
if warfarin is discontinued and heparin employed at/or prior to 6 weeks gesta-
tion, the miscarriage rate drops to 15%. This emphasizes the critical nature of
switching to heparin before 6 weeks gestation to obtain the maximal benefit.
Warfarin embryopathy results in chondrodysplasia punctata (nasal and mid-
face hypoplasia, frontal bossing, short proximal limbs, scoliosis, short phalanges
90
Obstetric CH05 18/10/05 10:22 am Page 91
and short stature). There is also a characteristic facial appearance with nasal
hypoplasia. This is due to warfarin-induced premature calcification of the nasal
cartilage, which results in failure of growth of the nose during childhood devel-
opment. This defect is, however, amenable to corrective plastic surgery. In addi-
tion, when warfarin has been used between 12 and 36 weeks gestation, an
association has been reported between its use and neurological and develop-
mental problems in the infant. It is supposed that this is due to small intracere-
bral bleeds in the fetus, consequent upon excessive anticoagulation (due to the
immature status of the fetal liver). Indeed, intracerebral bleeds have been
reported in the fetus of women treated with warfarin in the second and third
trimesters. Between 36 weeks gestation and delivery, there may be an increased
risk of abruption and of intracerebral bleeding in the fetus (particularly during
delivery), as well as a risk of severe maternal bleeding at delivery.
Thus, although warfarin may be preferred to heparin in the maternal interest for
some conditions (such as mechanical heart valves or severe recurrent thrombosis),
due to the better efficacy data for preventing maternal thrombosis, it carries signific-
ant risks for the fetus. In each case it is critical to establish the optimal anticoagula-
tion regime, balancing the risks between the mother and the fetus, and taking into
account the mother’s view of these risks. Such decisions are clearly best made prior
to pregnancy. All women on oral anticoagulants who could become pregnant must
be aware of the risks of conceiving whilst on these drugs, and of the need to report
to their hematologist and/or high-risk obstetrician immediately they become preg-
nant to ensure that optimal anticoagulation and pregnancy management can be
provided. In particular, as noted above, a switch to heparin should be made before
6 weeks gestation if this is the therapeutic strategy that is to be employed.
Heparin
The heparins are glycosaminoglycans with a high affinity for the endogenous anti-
coagulant antithrombin (see Chapter 1), altering both its structure and anticoagu-
Around 3% bone loss in normal pregnancy Around 5% bone loss with LMWH in pregnancy
lant function. Two classes of heparin are used clinically: unfractionated heparin
(UFH), which contains a mixture of molecular weights, and the fractionated low-
molecular-weight heparins (LMWH: see Table 5.3). Binding of UFH to antithrom-
bin changes the conformational structure of antithrombin, resulting in a 1000-fold
increase in its ability to inactivate factor (F) Xa and FIIa. The smaller fragments
contained in LMWH preferentially bind the antithrombin–FXa complex.
The heparins currently available are inactive orally and have to given by an
intravenous or subcutaneous route. The half-life of intravenous UFH is variable,
but can be as short as 30–45 minutes. The therapeutic level of UFH can be
assessed by an activated partial thromboplastin time (APTT or PTT: see Chapter
1). This is achieved by determining a ratio of the APTT in the patient sample to
the APTT of a normal pooled plasma: this gives a therapeutic range of 1.5–2.5.
Unlike warfarin, there is no internationally accepted standardized ratio. Indeed,
when used in pregnancy, it must be remembered that the APTT may be short-
ened in pregnancy (as a result of an increase in the levels of key proteins such as
FVIIIc, fibrinogen and the heparin-binding proteins), giving the impression of
heparin resistance. When given subcutaneously, UFHs have a half-life of around
1.5 hours, and dose monitoring can be achieved by mid-interval APTT assessment
(4–6 hours after dosing). The rate of major bleeding in pregnancies treated with
UFH is 2%, which is consistent with the bleeding risk associated with both UFH
and warfarin therapy in non-pregnant subjects treated for DVT. Furthermore it
should be noted that dose-adjusted subcutaneous UFH can cause a persistent anti-
coagulant effect up to 28 hours after the last injection, which could be problem-
atic at the time of delivery. However, the mechanism for this effect is unclear.
LMWHs are given subcutaneously and have more predictable kinetics and
>90% bioavailability. This results in a half-life of 4–6 hours, which allows once, or
twice, a day dosing, and monitoring is seldom required. The APTT is insensitive
to the presence of LMWH (even when present in excess) and is likely to remain
within reference limits. If monitoring of these drugs is needed, this can be
achieved by assessment of the effect of the LMWH on the inactivation of coagula-
tion FXa (an anti-Xa assay).
Reversal of intravenous UFH can be achieved by protamine sulfate injection:
the amount to be given can be titrated to the predicted residual amount of UFH
in the circulation, which routinely results in a protamine dose of 25–50mg. The
downside of the long half-life of LMWHs is the difficulty in reversing their effect
if bleeding occurs, or an excess dose is given accidentally. Protamine will inhibit
only the LMWH currently in the circulation. In these circumstances, if bleeding
is ongoing, it has been suggested that further protamine dosing should be deter-
mined by clinical response alone, although an initial anti-Xa assessment is often
carried out in these circumstances. The anti-Xa level does not, however, predict
bleeding, and the frequent monitoring which would be required to determine
protamine dosing is often impractical. This problem can be significant, as an
excess of protamine can also act as an anticoagulant.
92
Obstetric CH05 18/10/05 10:22 am Page 93
Heparin-induced thrombocytopenia
There are two types of thrombocytopenia associated with heparin (see also
Chapter 8). The first is non-immune heparin-induced thrombocytopenia (HIT),
which is sometimes less accurately described as Type 1 HIT. This is a benign non-
immune condition that results in a mild degree of thrombocytopenia, which
appears to be due to a direct effect of heparin on the platelet membrane and is
not associated with clinical symptoms, or thrombosis.
The more important type of thrombocytopenia associated with heparin is
Type 2 HIT, also known as HIT thrombosis syndrome (HITTS), reflecting the
clinical expression of the syndrome with thrombosis as well as thrombocytopenia.
This immune condition arises from the development of specific heparin-depend-
ent immunoglobulin (IgG) antibodies against the platelet factor (PF) 4–heparin
complex. It commonly has its onset 5–14 days after the commencement of
therapy, although it can occur after a brief exposure if there has been prior
heparin exposure within the previous 3 months. This leads to platelet activation,
generation of pro-coagulant microparticles from the platelet membrane and
platelet consumption. Platelet levels can fall to a median of around 60 × 109/ l,
but levels <20 × 109/l are encountered in 10–15% of cases. Furthermore, a
significant fall which does not result in a level less than the lower limit of the ref-
erence range can still be consistent with the diagnosis of Type 2 HIT. In addition
to platelet consumption, Type 2 HIT is also associated with thrombin generation.
This occurs via the generation of TF on endothelial cells activated by HIT anti-
bodies binding to PF4 found on endothelial heparan sulfate, as well as increased
expression of TF on monocytes upon activation by HIT antibodies. Clinically
severe arterial or venous thrombotic complications can ensue. In 5–10% of sub-
jects there may be venous thromboembolism, but myocardial infarction, limb
ischemia, mesenteric artery thrombosis and cerebrovascular thrombosis also
occur. The occurrence of thrombosis in affected patients is associated with a
significant mortality. Interestingly, despite the thromboctyopenia, petechiae are
absent, but skin reactions at injection sites and systemic inflammatory reactions
can also occur. Thus, if a patient develops a skin reaction whilst on heparin it is
prudent to monitor the platelet count for HIT.
The incidence of HIT has been estimated at 2.7% with UFH, and there is
anecdotal evidence that HIT also occurs with LMWH, although the incidence is
less easy to define. The incidence of HIT with LMWHs could be as low as 0% and
the overall relative risk is likely to be at least 8-fold greater with UFH than
LMWH. This is likely to relate to the heparin polysaccharide chain length, which
needs to be at least 12–14 polysaccharides long to form an HIT antigen with PF4.
The risk of both HIT and thrombosis varies with the patient type, the duration of
use and the type of heparin. With UFH, cardiac surgery patients are most at risk
of developing antibodies (around 50%), but few (<5% of those who become anti-
body positive) develop HIT, while orthopedic surgical patients have a lower rate
of antibody development (15%), but as many as one third will go on to develop
93
Obstetric CH05 18/10/05 10:22 am Page 94
HIT. It is important to note that HIT does not always lead to a clinical throm-
botic event. Indeed, in cardiac surgery with UFH, 1% or less will develop a
thrombotic problem.
There are occasional reports of thrombocytopenia or heparin-dependent
platelet-activating antibodies in pregnant patients treated with LMWH. However,
even in pregnant patients diagnosed with HIT associated with LMWH, there is
often a previous sensitization or HIT episode related to UFH exposure. In
general terms, medical and obstetric patients are considered least at risk, and
recent recommendations do not consider that routine monitoring of the platelet
count is required with LMWH use in pregnancy (the risk with LMWH in preg-
nancy is considered to be <0.1%). Thus, platelet count monitoring should be
restricted to special situations (see Table 5.4).
HIT is a clinicopathologic syndrome, although the diagnosis is principally a
clinical one, supported by the detection of serologic factors. Key factors to con-
sider in making the diagnosis are shown in Table 5.4. HIT antibodies can be
detected by either platelet-activation (functional) assays or by antigen assays
directed against the heparin–PF4 complex. The diagnostic specificity of a sensi-
tive washed platelet activation functional assay is greater than that of an antigen
assay. The diagnosis of HIT is made when there is an otherwise unexplained fall
in the platelet count (usually >50%, even if the platelet count remains
>150 × 109/l), when there are skin lesions at heparin injection sites, or when an
acute systemic reaction such as cardiorespiratory distress after intravenous
heparin administration has occurred. The diagnosis is supported by the detec-
tion of HIT antibodies, but can be made even when there is a failure to confirm
the presence of such antibodies. HIT antibody seroconversion on its own (i.e.
without thrombocytopenia or other clinical sequelae) is not, however, con-
sidered sufficient to constitute a diagnosis of HIT.
Treatment centers on the discontinuation of any heparin preparation and the
use of alternative thrombin inhibitors, usually in therapeutic doses. These
alternatives include: the heparinoid, danaparoid; recombinant hirudin; and the
direct thrombin inhibitor argatroban. Fondaparinux has also been employed in
this context. While warfarin is best for chronic therapy, it should be avoided in the
acute stage, as (noted above) it may result in a precipitous fall in the endogenous
vitamin K-dependent anticoagulant protein S. This may result in skin necrosis and
venous limb gangrene: those patients with protein C, protein S or antithrombin
deficiency, or FV Leiden carriage are perhaps most at risk. Warfarin should there-
fore be delayed until the platelet count is >100 × 109/l and the HIT is resolving
with the use of other antithrombotic agents. Adjunctive treatments that have been
employed in the management of HIT include: embolectomy; plasmapheresis;
thrombolysis; and high-dose intravenous normal immunoglobulin (to inhibit the
antibody-mediated platelet activation). It should be noted that platelet compo-
nent therapy should be avoided in HIT patients, as it is thought that this may
increase platelet consumption and precipitate thrombosis.
Osteoporosis
Of all the potential complications of heparin, perhaps the side effect that con-
cerns obstetricians most is osteoporosis. Pregnancy itself is associated with a
reduction in bone mineral density of the order of 3%. With prolonged UFH use,
>30% of women on UFH will lose significant (>10%) bone mass. This is related
to the dose and duration of therapy, and is more common with prolonged high-
95
Obstetric CH05 18/10/05 10:22 am Page 96
dose treatment. However, problems have been reported with doses as low as
15 000 day and with therapy given for only 7 weeks. In one study of long-term
prophylactic UFH therapy in pregnancy, a 2.2% incidence of vertebral fractures
was reported. Although, in another study, spinal fractures were reported in as
many as 15% of patients receiving UFH at a dose of 10 000IU twice daily for a
period of 3–6 months.
Experimental model and animal studies provide some insight into the
mechanism of heparin-induced osteoporosis. A dose-dependent decrease in can-
cellous bone in the femur (resulting from decreased bone formation and
increased bone resorption) occurs in animals treated with UFH for 1 month. In
one animal study, UFH resulted in a 30% loss in cancellous bone during the first
month of treatment. In this study, heparin was shown to accumulate in the bone
and was retained there for at least a further 1 month after the cessation of
therapy. This prolonged accumulation would suggest that heparin-induced osteo-
porosis may not be rapidly reversible.
LMWH carry a substantially lower risk of osteoporosis than UFH. In one study
of non-pregnant venous thromboembolism (VTE) patients treated for 3–6
months, only 2.5% of subjects receiving LMWH experienced a vertebral fracture
compared to 15% in the UFH group. One randomized trial of LMWH versus
UFH in pregnancy measured bone mineral density in the lumbar spine for up to
3 years after delivery. In this study, bone density did not differ between healthy
controls and the dalteparin group, but was significantly lower in the UFH group
when compared to both controls and dalteparin-treated women. Multiple logistic
regression found that the type of heparin therapy was the only independent
factor associated with reduced bone mass. This is consistent with several observa-
tional pregnancy studies that have shown no difference in lumbar spine density
in subjects on prolonged LMWH therapy and no greater bone loss than that
which occurs in pregnancy naturally. In a review of LMWH (largely enoxaparin
and dalteparin) therapy in over 2500 pregnancies, only one case of heparin-
induced ostepenia with fracture (vertebral) was reported in a woman who had
received a high dose (15 000IU/day) of dalteparin for a total of 36 weeks.
Recently, however, three cases of osteoporotic fractures in association with tinza-
parin use in pregnancy in one center have been reported. Whether this finding is
causally related to tinzaparin (as additional therapies and underlying problems
can also influence the development of osteoporosis) and whether this risk
applies to other LMWH is unclear. In any event, the risk associated with LMWH
remains very low in comparison to UFH. This difference between UFH and
LMWH is consistent with animal studies, which show greater cancellous bone loss
with UFH than LMWH.
Bleeding risk
LMWH appear to carry a lower risk of bleeding than UFH: severe bleeding prob-
lems have not been attributed to either treatment or prophylactic-dose LMWH
96
Obstetric CH05 18/10/05 10:22 am Page 97
Antithrombotic efficacy
The effectiveness of LMWH in the management of thrombotic conditions in
pregnancy is supported by the low VTE recurrence rate (just over 1%) reported
during treatment in both large cohort studies and systematic reviews. This com-
pares favorably with the recurrence rates of 5–8% reported in trials of non-preg-
nant patients treated with either LMWH or UFH (followed by coumarin therapy)
after 3–6 month follow-up. LMWH thromboprophylaxis is also associated with a
VTE rate of <1%.
Fetal outcome
With regard to fetal outcome, in general terms, the evidence supports a benefi-
cial effect of LMWH on pregnancy loss rates. The successful pregnancy rate,
reported in women receiving LMWH for previous adverse pregnancy outcomes
(such as recurrent fetal loss), is >80%. This rate is consistent with that found in
randomized trials of antithrombotic therapy (UFH or LMWH) in women with
previous pregnancy loss associated with antiphospholipid syndrome or heritable
thrombophilia, where such intervention resulted in a significant improvement in
pregnancy outcome.
Danaparoid
Danaparoid is a heparinoid (a mixture of heparan, dermatan and chondroitin
sulfates), that inactivates FXa and FIIa. It has a relatively long half-life of 25 hours
in non-pregnant subjects. Danaparoid has been used successfully in pregnancy-
associated HIT and, out-with pregnancy, danaparoid is successful in >90% of
cases. However, a positive cross-reaction can occur in 15% of cases of HIT. Dana-
paroid is also useful in pregnant patients with an allergy to LMWH. The throm-
97
Obstetric CH05 18/10/05 10:22 am Page 98
boprophylactic dose for an average pregnant patient is 750IU given twice daily by
subcutaneous injection. At a prophylactic dose, it has no effect on platelet func-
tion and should not be associated with excess blood loss at delivery. When a ther-
apeutic dose is required, danaparoid can be monitored by an anti-Xa assay using
a danaparoid control. The recommended dosing with danaparoid for HIT out-
with pregnancy is an intravenous bolus of 2250IU, followed by an intravenous
infusion at 400IU/hour for 4 hours, then 300IU/hour for 4 hours, then
200IU/hour adjusted by anti-Xa levels. It does not cross the placenta and
although it is not yet clear whether it crosses the breast, this appears unlikely, as
no anti-Xa activity occurs in breast milk. In any event, it should be destroyed in
the infant GI tract. It is exclusively excreted by the kidney and should be used
with extreme caution in subjects with renal failure.
Lepirudin
Lepirudin, a recombinant hirudin (hirudin is derived from the medicinal leech
Hirudo medicinalis), binds tightly to thrombin with slow dissociation, making it
effectively an irreversible inhibitor. There is very little information on its use in
pregnancy, but it may have a role as an alternative to danaparoid in the manage-
ment of pregnancy-related HIT. The dose used for HIT is an initial infusion of
0.15mg/kg/hour intravenously, with the aim to achieve a target APTT of 1.5–2.0
times either the patient’s baseline APTT, or the mean of APTT laboratory refer-
ence range. It has a relatively short half-life of about 80 minutes and is renally
excreted. At higher doses the ecarin clotting time has a more linear correlation
with plasma hirudin levels and may be more useful for monitoring. In animal
studies, hirudin has low placental transit and it does not appear to cross the
breast. When considering the use of hirudin, it should be borne in mind that it
currently has no antidote and it should be used with caution in those with renal
impairment. In practice, the use of hirudin should be limited to those subjects
where the HIT antibody is shown to cross-react with danaparoid, or in those who
have a cutaneous allergy to danaparoid. There is a high rate of antibody develop-
ment to hirudin of 40–60% and anaphylaxis can occur, although this is rare.
Argatroban
Argatroban is a synthetic thrombin inhibitor derived from L-arginine. It binds
tightly, yet reversibly, to thrombin. Individuals receiving prolonged, or repeated,
administration show no generation of antibodies and no enhancement or sup-
98
Obstetric CH05 18/10/05 10:22 am Page 99
pression of the anticoagulant response. It is metabolized in the liver and its effect
can be monitored with the APTT (aiming to achieve 1.5–3 times control values).
Its action is quickly reversed by stopping the infusion (<60 minutes), but clear-
ance is reduced 3–4-fold if there is moderate hepatic impairment. It is used in
the treatment of HIT where its effectiveness is clearly demonstrated. In this situ-
ation the recommended dose is an initial infusion rate of 2µg/kg/minute, with
no initial bolus required. There is no significant experience of the use of this
agent in pregnancy.
Aspirin
Aspirin irreversibly inhibits (through acetylation) cyclo-oxygenase, which is
pivotal in the generation of thromboxane A2 in the platelet. The effect of aspirin
lasts between 7 and 10 days, as restoration of platelet function requires new
platelet synthesis. Aspirin only partially inhibits adenosine diphosphate (ADP)
and collagen-induced platelet activation, and has no effect on thrombin-medi-
ated platelet activation. Aspirin may be as effective as an antithrombotic agent at
modest doses (75mg/day) as it is at higher doses (1200mg/day). The principal
side effects of aspirin are bleeding and idiosyncratic or allergic reactions. In chil-
dren, ingestion of aspirin has been associated with Reye’s syndrome.
Aspirin has been widely assessed for the prevention of pregnancy-related com-
plications such as intrauterine growth restriction and pre-eclampsia. The use of
low dose aspirin (60–75mg/day) to prevent pre-eclampsia is based upon the
rationale that pre-eclampsia is associated with alterations in the production of
prostacyclin and thromboxane, secondary to activation of the clotting system and
changes in platelet function. A recent meta-analysis reported on 42 randomized
trials involving over 32 000 women comparing antiplatelet agents to placebo or
no treatment. Trials were subgrouped by maternal risk. Taking the cohort as a
whole, there was a 15% reduction (relative risk 0.85, 95% confidence interval
(CI) 0.78–0.92) in the risk of pre-eclampsia associated with the use of antiplatelet
agents. These assessments have shown that low-dose aspirin (around 75mg/day)
appears safe for use in pregnant women. At higher doses (>150mg/day) its
safety, especially in the first trimester, is controversial.
Although aspirin has been shown to reduce the risk of venous thrombosis in
the context of orthopedic surgery, its role in the context of pregnancy-related
thrombosis is not yet defined. However, it does not appear to offer the same level
of protection as UFH or LMWH for thromboprophylaxis. When aspirin has been
used as a substitute to coumarins (in subjects with mechanical heart valves), as
would be expected, it confers insufficient anticoagulation and results in an unac-
ceptable (and often fatal) 25% incidence of systemic embolism and 4% inci-
dence of valve thrombosis, although it appears safe from a fetal perspective.
Aspirin has a role in combination with other therapies, particularly in combi-
nation with LMWH, in the antiphospholipid syndrome. It is also of value where
specific antiplatelet therapy is required in pregnancy, such as in women with a
99
Obstetric CH05 18/10/05 10:22 am Page 100
Dextran
Dextran has been used for peripartum thromboprophylaxis, particularly during
Cesarean section. It does, however, carry a significant risk of anaphylactic and
anaphylactoid reactions, which have been associated with uterine hypertonus,
profound fetal distress, and a high incidence of fetal death or profound neuro-
logical damage. Thus, dextran for thromboprophylaxis should be avoided prior
to delivery.
leads to FXa inhibition, but has a prolonged half-life (80 hours), which could
allow once weekly dosing. There are currently no data on its use in pregnancy.
Protamine sulfate has no action on these drugs, but two studies have shown that
recombinant FVIIa (rFVIIa) may be able to reverse the biochemical changes
induced by these pentassaccharides in healthy volunteers. In future, heparinases
may also have a role in their inactivation.
Ximelagatran
Ximelagatran is the prodrug of melagatran, which is an active site-directed
thrombin inhibitor. Ximelagatran is well absorbed from the gut and has a plasma
half-life of 3–4 hours, allowing it to be administered twice daily. Thus, it has the
potential of being an alternative oral anticoagulant to warfarin. It may require a
dose adjustment when there is renal impairment, and in 6% of subjects a mild
rise in transaminases has been noted, although more severe reactions have also
occurred. This has led to concerns for the safety of this agent and it remains unli-
censed in both the USA and Europe at present. It appears most likely that xime-
lagatran will have a role in the prophylaxis of atrial fibrillation in the first
instance. At present, however, there is insufficient information on the safety of
this class of compounds in those who are pregnant or breastfeeding.
101
Obstetric CH06 18/10/05 10:22 am Page 102
CHAPTER 6
Management of
thrombotic disorders
Pregnancy places a substantial demand on the cardiovascular system. With
increasing gestation there is a 30–50% increase in cardiac output, which is
accompanied by an increase in prothrombotic coagulation factors, a decrease in
some natural anticoagulants and an alteration in systemic fibrinolysis. This prepa-
ration for the hemostatic challenge of delivery poses a particular threat to those
women at risk of thrombosis.
More and more, obstetricians and hematologists are faced with the manage-
ment of pregnant subjects who have risk factors for either arterial or venous
thrombosis. This most commonly includes those with a previous thrombosis or
those with prosthetic heart valves, but there is an evolving awareness of genetic
and acquired risk factors that may identify those at particular risk of vascular
occlusion, not only in pregnancy but also in later life.
Hypercholesterolemia
Family history of ischemic heart disease
Smoking
Hypertension
Diabetes mellitus
Obesity
Essential thrombocythemia
Polcythemia rubra vera
Sickle-cell disease
Prosthetic heart valve
Mitral stenosis and atrial fibrillation
Mitral bioprothesis and atrial fibrillation
Mechanical mitral valve
Antiphospholipid syndrome
? Smoking with Factor V Leiden or prothrombin 20210A
Hyperhomocysteinemia
Cocaine
Ergot
?Marijuana
Previous cerebral ischemia of arterial origin
Previous myocardial infarction
Hypotension
Pre-eclampsia
puted, but, if confirmed, will contrast with more severe heritable thrombophilias,
such as antithrombin, or protein C or S deficiency, where there has never been
convincing evidence of a link with arterial thrombosis.
fetal and maternal complications with the currently available therapies – warfarin
and heparin (see Chapter 5) – is also unknown. In particular, in view of the con-
cerns with oral coumarin therapy, there is a need to find alternative strategies
that are will be safe for both the mother and the fetus.
A recent review of maternal and fetal outcomes in women with prosthetic
heart valves reported that commonly used anticoagulant regimes were: (1) use of
coumarin throughout pregnancy; (2) replacement of coumarin with unfraction-
ated heparin (UFH) from 6 to 12 weeks gestation; and (3) UFH use throughout
pregnancy. Overall, fetal wastage was 33.6% in the first group, 26.5% in the
second and 19.6% in the third. In the coumarin regimes, heparin was used at
term to avoid delivery with an anticoagulated mother and fetus. Coumarin
embryopathy occurred in 6.4% of live births where a coumarin was used through
pregnancy: this was eliminated if heparin was substituted by 6 weeks gestation.
In this review (see Table 6.2), coumarin use through pregnancy was associated
with a lower risk of thromboembolic problems (3.9%) when compared with preg-
nancies where heparin was substituted for the coumarin. UFH used between 6
and 12 weeks gestation was associated with a risk of valve thrombosis of 9.2%, and
UFH used throughout pregnancy was associated with a 25% rate of valve throm-
bosis with adjusted-dose heparin and a 60% rate with low-dose UFH. However,
the numbers of patients exposed to UFH included in the review was modest. In
addition, many thromboembolic events were associated with an inadequate
heparin dosage. Moreover, modern bi-leaflet valves are substantially less throm-
bogenic than earlier mechanical valves such as the Bjork-Shiley and Starr-
Edwards valves, which were predominant in most of the work reviewed. These
limitations make it difficult to extrapolate early work directly to contemporary
practice with modern valves and well-controlled anticoagulation with UFH (or
indeed low-molecular-weight heparin (LMWH)) with regard to thrombotic risk.
Table 6.2 – Maternal outcomes from women with mechanical valves receiving anticoagulation
*Up to 50% of complications occurred on low-dose heparin and others with inadequate dosing based on the
activated partial thromboplastin time. There is a 2.5% incidence of severe bleeding at delivery and the rate with
low-molecular-weight heparin is unclear with an estimate of 13%.
105
Obstetric CH06 18/10/05 10:22 am Page 106
Thus, on the basis of the limited data available, coumarin appears more effect-
ive than UFH for thromboembolic prophylaxis in women with mechanical heart
valves in pregnancy (Table 6.2), but UFH is associated with a better fetal
outcome (see Table 6.3).
The situation is changing with the increasing use of LMWH in pregnant
women with prosthetic heart valves, which has attractions over UFH in terms of
monitoring and maternal side effects, although concerns still exist over how
effective LMWH is for thrombrophylaxis in this situation. In a review of 67 preg-
nancies treated with LMWH in women with prosthetic valves, valve thrombosis
occurred in seven cases, giving an incidence of 10.4% (95% confidence interval
(CI), 3.1–17.7%) and a mortality rate of 1.49% (95% CI, 0.03–8.34%). The
overall thromboembolic complication rate was 13.4% (9 in 67; 95% CI,
5.4–21.4%). However, eight of the nine patients with a thromboembolic compli-
cation had received a fixed dose of LMWH, and in two of these patients, a fixed
low-dose of LMWH. In 42 pregnancies with anti-Xa levels monitored, only one
thromboembolic complication occurred. The live birth rate was 89.5% (95% CI,
82.5–96.5%), the fetal wastage was 8.9% (95% CI, 2.1–15.7%) and there were no
reported congenital anomalies. Thus, adjusted-dose LMWH (anti-Xa level at a
minimum of 1.0IU/ml) compares favorably with UFH in this situation.
Because of the limited data, management remains controversial and several
approaches remain acceptable, including substitution of LMWH for UFH.
However, it is clearly critical to ensure adequate doses of UFH or LMWH. There
is still a need for consensus among experts to systematically gather the best avail-
able evidence, identify unresolved issues and generate studies designed to answer
these questions. Meanwhile, the American College of Chest Physicians (ACCP)
recommends either adjusted-dose twice-daily LMWH throughout pregnancy, in
doses adjusted either to keep a 4 hourly post-injection anti-Xa level at around
1.0–1.2IU/ml (preferably) or in doses adjusted by weight. Alternatively, aggres-
Table 6.3 – Fetal outcome in women with mechanical valves receiving anticoagulation
APTT, Activated partial thromboplastin time; LMWH, Low-molecular-weight heparin; UFH, Unfractionated heparin.
Most CIAO occur in the puerperium or the third trimester and, in >50% of
cases there may be evidence of a major arterial occlusion. In a substantial
number of cases, however, no evidence of major occlusion is found either at
angiogram or post-mortem. A number of common risk factors relate to CIAO in
pregnancy as they do in the non-pregnant. These include hypertension, smoking,
pre-existing premature atherosclerosis and valvular heart disease. In pregnancy,
eclampsia is also associated with small-vessel cerebral complications. CIAO is
likely to be more common in older women, in those with sepsis and in those who
have undergone Cesarean section delivery. However, in many studies, a clear dis-
tinction between ischemic and other cerebral events is not made. In addition,
rarer causes – such as choriocarcinoma, paradoxical embolism, sickle-cell disease,
lupus inhibitor-related thrombosis, ergot alkaloid usage and homocystinuria –
have also been reported in the etiology of pregnancy-related CIAO.
Table 6.5 – Management of those with cerebral ischemia of arterial origin (CIAO) or myocardial infarction (MI) in
pregnancy
ACE, Angiotensin-converting enzyme; IHD, Ischaemic heart disease; LMWH, Low-molecular-weight heparin;
LVF, Left ventricular function; VTE, Venous thromboembolism.
Although infarction may occur at any gestation, the majority of deaths occur
at, or within 2 weeks of, the infarction, and often during labor or delivery. In sub-
jects in whom a post-mortem was carried out, coronary atherosclerosis was
evident in 43% of subjects and probable coronary thrombosis, without evidence
of atherosclerotic disease, was evident in 21%. Atherosclerosis was more com-
monly found in subjects with an antipartum myocardial infarction than in those
with a postpartum event. Dissection of the coronary artery was found in 16% of
subjects and was more often associated with infarction in the puerperium. No
detectable evidence of coronary occlusion was found in around 30% of antepar-
tum and 75% of postpartum events. The absence of detectable occlusive disease
may indicate a significant role of transient coronary artery spasm in these sub-
jects.
It is likely that the prothrombotic changes in coagulation, which occur even in
normal pregnancy, increase the risk for arterial thrombosis. These include an
increase in coagulation factors and a reduction in the coagulation inhibitor
protein S (although heritable protein S deficiency is not associated with adult
109
Obstetric CH06 18/10/05 10:22 am Page 110
option for the mother, as this avoids the cardiovascular stress of labor and the
possibility of an emergency Cesarean section, which carries a substantially greater
risk of complications. There may also be a role for continuing LMWH, due to the
increased risk of venous thrombosis that may occur as a consequence of cardiac
failure, immobility or concurrent inflammation. There may be a requirement to
avoid neuraxial anesthesia if a combination of aspirin and clopidogrel is used, as
it may be associated with a more marked increase in the bleeding time than
aspirin alone. When a maternal death has occurred there should be a rapid deliv-
ery if a viable fetus is to be secured.
The risk associated with future pregnancy is dependent on the etiology of the
event, the presence of persisting myocardial ischemia and the degree of residual
impairment of left ventricular function.
DVT
The absence of elevated D-dimer levels (detected by enzyme-linked immunosor-
bent assay (ELISA) or whole blood agglutination assay) is useful to exclude VTE
in non-pregnant subjects, where they have a high negative predictive value. As
discussed in Chapter 1, elevated D-dimers are seen in around 50% of females in
the first trimester, and may be universal in later pregnancy and the puerperium.
On account of this, their ability to exclude VTE in pregnancy is not yet reliable.
Although a ‘normal’ result may be helpful, D-dimer results in pregnancy do not
remove the need for an objective diagnosis.
Although contrast venography is the gold-standard investigation for DVT diag-
nosis, compression ultrasound assessment is more likely to be used as the initial
assessment. Ultrasound has a high sensitivity and specificity for proximal thrombo-
sis in non-pregnant subjects, but has its greatest utility when used between the
inguinal ligament and the bifurcation of the popliteal vein. If, on compression, the
venous lumen collapses then there is unlikely to be thrombus present. When there
is the possibility of an isolated iliac or calf DVT, compression ultrasound may be of
less value in the exclusion of disease. In general, if there is a clinical suspicion,
treatment should be started and ultrasound scanning of the lower limbs should be
performed. If the scan is negative, and there is a low clinical suspicion, then treat-
ment can be stopped. If, despite a negative scan, there is still a high clinical suspi-
cion then treatment should be continued for 7 days and the scan should be
repeated. When there is a suspicion of isolated calf disease, serial ultrasound assess-
ment over the next 7–14 days, without treatment, has been proposed by some. This
proposal is based upon the low risk of extension to popliteal and femoral vessels
112
Obstetric CH06 18/10/05 10:22 am Page 113
(of the order of 20–25%), and the low risk of embolization of isolated calf DVT,
although the general applicability of this approach is not clear. There may also be a
problem in the diagnosis of an isolated iliac thrombosis in pregnancy, as there may
be difficulty in assessing the vein as it passes behind the pregnant uterus. This is,
thankfully, an uncommon diagnostic problem and requires an individual approach
to minimize the risk of fetal radiation exposure from venography or CT scanning.
As a guide, bilateral venography may result in fetal exposure of around 0.6rad
without abdominal shielding, although more limited venography will markedly
reduce exposure (perhaps to <0.05rad). It is thought that in utero exposure to
<5rad does not result in fetal loss or congenital abnormality, but may be associ-
ated with a 2-fold increase in the risk of childhood malignancy. An alternative
technique may be MRI, which appears to give no immediate problems, but its
long-term effects on the fetus are unknown.
PTE
Patients with acute PTE in pregnancy often present with dysponea, but wheeze,
unexplained fever, pleuritc pain, as well as central chest pain, cyanosis and col-
lapse (associated with more massive thrombosis) may all be presenting features.
In such patients, other potential diagnoses such as infection, effusion and pneu-
mothorax, should be excluded by X-ray. Electrocardiogram (ECG) analysis does
not produce a sufficiently specific pattern to diagnose PTE, and blood gas analy-
sis interpretation is complicated by the presence of pregnancy-related respiratory
alkalosis. The sampling procedure itself may also result in bleeding from the
puncture site. Overall, blood gas analysis is probably not warranted in the routine
diagnosis or exclusion of PTE in pregnancy. Indeed, X-ray examinations, blood
gas analysis and ECG are, on their own, insufficient to exclude or diagnose PTE.
To investigate PTE in pregnancy, one potential strategy is to investigate the
lower limbs for evidence of DVT by compression ultrasound. If the results are
positive, treat the patient with a presumptive diagnosis of PTE.
When there is no alternative diagnosis that accounts for the symptoms and
signs, and there is no evidence of a peripheral DVT, V/Q scanning should be
performed. As this may result in fetal exposure of <0.5rad, useful information
can be obtained from a perfusion scan alone in some cases. Avoidance of the
ventilation scan can reduce the fetal exposure to <0.01rad. A positive result will,
however, require a ventilation scan to exclude conditions such as pneumonia.
As in non-pregnant subjects, a V/Q scan will lead to three possible outcomes:
a high probability of PTE (a large ventilation/perfusion mismatch); an interme-
diate probability (small, multiple defects); or a normal result (no perfusion
defect seen). Although, as in non-pregnant subjects, a negative result does not
entirely exclude thrombosis, a normal result will often be considered as evidence
of no PTE. If, however, there is continuing clinical suspicion, or when an inter-
mediate result is obtained, spiral CT should be considered. It may lead to less
radiation to the fetus than standard angiography by the femoral route, and even
113
Obstetric CH06 18/10/05 10:22 am Page 114
V/Q scanning, but results in a substantial dose to the maternal breasts of around
2–3rads. In addition, although spiral CT has a high utility for central thrombosis,
it may not reliably detect small peripheral events and has no proven track record
in pregnancy. If there is sufficient diagnostic doubt, pulmonary angiography may
be required. In such cases the brachial route should be preferred, as this results
in a substantial reduction in the potential radiation exposure to the fetus. Many
of the known side effects attributed to pulmonary angiography predate the intro-
duction of low-ionic, low-osmolar contrast medium. Furthermore, pregnant
women are less likely to have major cardiac, respiratory or renal disease, and are
probably less susceptible to such side effects.
114
Obstetric CH06 18/10/05 10:22 am Page 115
Table 6.7 – Rough guide to thrombophilia and venous thromboembolism (VTE) risk related to pregnancy
*Assumes 1 VTE in 1500 deliveries and an unselected population. AT, Antithrombin; F, Factor; PT, Prothrombin.
115
Obstetric CH06 18/10/05 10:22 am Page 116
Monitor for clinical signs of VTE in the first week of the puerperium
Early mobilization and hydration
2+ risk factors: Compression stockings
3+ risk factors: LMWH/UFH for 3–5 days
Table 6.10 – Low-molecular-weight heparin (LMWH) prophylaxis against venous thromboembolism (VTE) in
pregnancy
AN, Antenatal; APA, Antiphospholipid antibody; APAS, Antiphospholipid syndrome; AT, Antithrombin;
FH, Family history; LT, Long term; PMH, Past medical history; PN, Postnatal
For those on treatment or high prophylactic doses of heparin, the dose should
be reduced to a standard prophylactic dose the day before a planned delivery and
continued throughout delivery at this dose. If the woman presents in labor, the
treatment dose should be withheld and protamine sulfate used if required.
117
Obstetric CH06 18/10/05 10:22 am Page 118
Treatment should be continued for 3–6 months after the event and should
include the puerperium in all cases. Whether treatment should be continued for
6 or 12 weeks after delivery is not clear, but may depend on whether the VTE
occurred early or late in pregnancy. In any case, it is sensible to review all
patients at 3 months to determine if there are persisting risk factors. Whether it is
safe to reduce the dose of heparin after the first 2–3 weeks of therapy is also not
clear, but a change to LMWH with, or without, a dose reduction is widely prac-
ticed, especially if there is a high risk of osteoporosis or hemorrhage. In all cases,
the platelet count should be measured at least after 7 days of therapy, and
perhaps more frequently if there is to be prolonged use of UFH. In those with an
antenatal VTE, an individual decision is required as to whether heparin or war-
farin should be used for continuing treatment in the puerperium.
References
Chan et al. 2000. Anticoagulation of pregnant women with mechanical heart
valves: a systematic review of the literature. Arch Int Med, 160, 191–6.
121
Obstetric CH07 18/10/05 10:23 am Page 122
CHAPTER 7
Thrombophilia and
pregnancy outcome
Serious pregnancy complications, including pre-eclampsia (PET), fetal loss,
intrauterine growth restriction (IUGR) and placental abruption, occur in up to
5% of women of reproductive age, and may be related to alterations in placental
perfusion and development. In the past few years maternal carriage of a number
of heritable thrombophilias, or thrombophilias with a mixture of heritable and
environmental influences, have been linked with such pregnancy complications.
Although a link between the thrombophilias and pregnancy-associated venous
thromboembolism (VTE: Table 7.1) is consistent with their potential to generate
an excess of thrombin and fibrin. The mechanism for their role in other
Pregnancy 4
Puerperium 14
Cancer 10
Medical immobilization 11
Postoperative 6–10
COCP 4–6
HRT 2–4
FVIIIC 4–6
HCY 2–4
PC/PS/AT deficiency Around 10
Family history Around 10
Heterozygous FV Leiden 5–8
Homozygous FV Leiden 50–80
Heterozygous PT 202010A 2–4
Homozygous PT 202010A 10
Antitphospholipid syndrome 7–9
AT, Antithrombin; COCP, Combined oral contraceptive pill; F, Factor; HCY, Homocysteine; HRT, Hormone
replacement therapy; PC, Protein C; PS, Protein S; PT, Prothrombin.
122
Obstetric CH07 18/10/05 10:23 am Page 123
pregnancy disorders is not so obvious, but could also be linked with alterations in
both thrombin generation and endothelial function.
As detailed in Chapter 1, thrombin is the pivotal protein in the coagulation
process. In addition to a wide variety of pro-coagulant functions (including
platelet activation, conversion of fibrinogen to fibrin and activation of Factors
(F) Vc and VIIIc), it also has an anticoagulant function via thrombomodulin and
the generation of activated protein C (aPC). Thus, via this negative feedback,
thrombin ultimately regulates its own generation. Adequate anticoagulant func-
tioning of thrombin via aPC depends upon intact functioning of the endothe-
lium, adequate levels of the components of the protein C/protein S system and a
normal sensitivity to the effects of aPC. Thrombin also participates directly in
tissue remodeling, wound repair, leukocyte chemotaxis, leukocyte adhesion, vas-
cular contraction and vascular permeability, via interactions with specific
endothelial and leukocyte thrombin receptors.
Although endothelial activation is a feature of normal pregnancy, heightened
activation has been linked with disorders such as PET. Any condition, such as a
thrombophilia, that has the potential to increase thrombin generation, or alter
endothelial function, may be capable, therefore, of influencing placental func-
tion and thereby fetal development. Maternal thrombophilia may, by excessive
thrombin generation, also alter the maternal response to the feto-placental unit.
This may, for example, contribute to the maternal presentation of PET. Whether
fetal inheritance of paternal thrombophilia contributes to these conditions has
not been fully examined, but could also be important in the generation of these
fetal and maternal disorders.
Heritable thrombophilias
Protein S deficiency
Protein S is a vitamin K-dependent, single-chain glycoprotein, which is synthe-
sized in the liver and vascular endothelium, and acts mainly as a cofactor to aPC
in the inactivation of FVIIIa and FVa (Figure 7.1). The plasma level of protein S
depends upon age, sex, lipid levels, estrogen, oral anticoagulant usage and the
presence of acute thrombosis (Table 7.2). In the plasma, around 60% of circulat-
ing protein S is bound to C4b binding protein, and only free protein S can func-
tion as a cofactor to aPC. The binding protein itself may also function as a link
between thrombosis and complement activation. Indeed, as C4b increases in
pregnancy, the binding protein also contributes to the physiological reduction in
protein S levels seen with increasing gestation.
Heritable protein S deficiency is transmitted as an autosomal dominant trait.
Those with heterozygous deficiency are at increased risk of VTE, as well as war-
farin-induced skin necrosis. Homozygote deficiency is extremely rare, as it is
usually associated with neonatal purpura fulminans and fetal or perinatal death.
However, there are reports of homozygous individuals who have survived long
123
Obstetric CH07 18/10/05 10:23 am Page 124
Protein C deficiency
Like protein S, protein C is also a vitamin K-dependent glycoprotein. It is synthe-
sized in the liver and, when activated, inactivates FVa and FVIIIa in the presence
of protein S. Its plasma levels are influenced by age, sex, lipid levels, and the
presence of liver disease, renal disease, acute thrombosis, disseminated intravas-
cular coagulation (DIC) or warfarin use. Higher levels can be seen in the puer-
perium and in subjects on oral contraceptives (see Table 7.2).
As with protein S, heritable deficiency is transmitted as an autosomal trait and
heterozygous deficiency increases the risk of thrombosis around 10-fold. The
The principal points of the coagulation cascade that are associated with thrombophilia are shown. The actions of
antithrombin (AT – previously called antithrombin III), which are reduced by AT deficiency are shown. Protein C
(PC), via activated protein C (aPC) and its cofactor protein S (PS), acts to inactivate Factor (F) Va (Va) and FVIIIa
(VIIIa). The action on FVa is impaired when there is the FV Leiden (FVL) mutation, and the action on FVIIIa is
impaired when there is an elevation of FVIIIc. This occurs in pregnancy, during the acute phase reaction and also
in association with the combined oral contraceptive or hormone replacement therapy. Elevated FIXc levels are
also an independent risk factor for venous thromboembolism (VTE). Elevated fibrinogen levels and antibodies to
phospholipid are associated with an increased risk of VTE and arterial thrombosis. Ca2+, Calcium; Plipid,
phospholipid; PTG2021OA, prothrombin G20210A mutation; TAT, thrombin–antithrombin complex; TF, tissue
factor.
124
Obstetric CH07 18/10/05 10:23 am Page 125
INTRINSIC
XII XIIa
XI
Ca2+
IXc XIa
TM
IXa aPC PC
(EPCR)
FVL +PS
125
TF–FVII–FVIIA
EXTRINSIC
Ca2+/Plipid PTG20210A Va Vi
Tenase Fibrinogen
Xa
AT THROMBIN
Ca2+/Plipid burst Fibrin
Xi
Prothrombinase
TAT
126
Obstetric CH07 18/10/05 10:23 am Page 127
homozygous state is also associated with neonatal purpura fulminans. In such cir-
cumstances the use of human-derived protein C concentrate may rescue such
individuals until lifelong anticoagulation can be established. Heterozygous defi-
ciency has been estimated to occur in 1 in 300–500 of the population, but is also
found in <5% of those with venous thrombosis. As with protein S, there is no
evidence that protein C deficiency is linked with arterial thrombosis in adults. In
studies investigating blood donor populations, the majority of individuals identi-
fied (either biochemically or genetically) are symptom free. Protein C deficiency
has been classified into two types:
Type 1: a quantitative reduction in functionally normal protein C;
Type 2: a level of protein C activity that is less than the antigen level; a defi-
ciency that results from the production of a functionally abnormal protein C
molecule.
These deficiencies can result from a number of genetic mutations and, as a
result, the diagnosis of protein C deficiency is made from the detection of
reduced plasma levels, unless there is a known mutation in other family members
(see Table 7.2).
Antithrombin deficiency
Although originally termed antithrombin III, the use of III is now considered
redundant. This glycoprotein is capable of inactivating most of the activated clot-
ting factors (including FVIIa bound to tissue factor (TF)). The presence of
heparin increases its ability to inactivate FXa and FIIa (thrombin) around 1000-
fold. Functional plasma levels are reduced in acute thrombosis, DIC, and in
patients receiving heparin therapy. Otherwise, there is little variation in plasma
levels with age and sex, and levels often remain within the non-pregnant range
during pregnancy.
Antithrombin deficiency is inherited as an autosomal trait, which, in its severe
heterozygous form, increases the risk of VTE around 20-fold and in its less severe
form around 10-fold. A homozygous severe deficiency is probably incompatible
with survival, but milder homozygous deficiencies may be amenable to replace-
ment with human-derived or recombinant antithrombin concentrate until life-
long anticoagulation can be established. A number of genetic mutations, which
alter either the clotting or heparin binding sites, are described. More severe
defects reduce antigen and activity equally, and lesser defects result in less activity
than antigen. Akin to the situation with protein C and S, the number of potential
mutations means that deficiency is routinely diagnosed on the basis of reduced
plasma levels. Again, in some instances, it may be possible to detect, or exclude, a
mutation that is known to have occurred in other family members. The preva-
lence of defects in the general population leading to a severe deficiency is of the
order of 1 in 4000–5000, although milder defects are more common and may be
as prevalent as 1 in 400. As with protein C and S deficiency, antithrombin is
127
Obstetric CH07 18/10/05 10:23 am Page 128
found infrequently in those with VTE (<3%), but is more often associated with
more severe disease, or disease onset at an early age. Indeed, around 50% of
severe heterozygotes experience thrombosis before the age of 30 years. Akin to
the situation with protein S and protein C, there is no evidence that antithrom-
bin deficiency is associated with arterial thrombosis.
FV Leiden
FV Leiden occurs as a result of a G→A mutation at position 1691 in the gene
coding for coagulation FV. It is the commonest heritable cause of resistance to
aPC (see below) and results in a change in one of the parts of the FV molecule
that is the target for cleavage by aPC. It is inherited as an autosomal dominant
condition with both heterozygotes and homozygotes surviving into adulthood.
Heterozygotes are reasonably common in European populations, with a preva-
lence of between 2 and 15%, but are less common, or even absent, in other pop-
ulations. In those of European extraction, FV Leiden is found in its homozygous
form in around 1 in 1000. Heterozygotes are also found in around 50% of those
with a history of VTE. In heterozygotes it carries around a 5-8-fold risk of VTE,
with a higher risk (around 50–80-fold) in homozygotes, and an increasing risk
associated with increasing age (Table 7.3). Given its high prevalence, it is not
unusual to find it in combination with other thrombophilic disorders. Whether,
in combination with smoking, it has a causal role in myocardial infarction is a
matter of topical debate, but there is no evidence linking FV Leiden with arterial
thrombosis in any other circumstances. Other heritable causes of aPC resistance
include two other point mutations in the FV gene (FV Cambridge and FV Hong
Kong) and a group of FV polymorphisms, known as the HR2 haplotype. At
present, the clinical impact of these alterations is unknown.
Dysfibrinogenemia
See Chapter 4.
ence of diabetes mellitus, migraine, as well as some fetal factors, such as hydrops
fetalis, hydatidiform mole, triploidy and trisomy 13, are also linked with a higher
risk of PET. In recent years a link with various inherited and/or acquired throm-
bophilias has been made (see Table 7.4). The etiology of the condition remains
unknown, but is generally accepted to be related to abnormal placentation, and
probably represents a failure of the normal immune tolerance that the mother
should have to the presence of the fetus.
In addition to the similarities between the placental pathology of PET (acute
atheroma – a necrotizing arteriopathy with fibrinoid necrosis and lipid-laden
macrophage accumulation in the placental bed) and cardiovascular pathology,
PET is accompanied by acute changes in coagulation, including a supraphysio-
logical increase in von Willebrand’s Factor (vWF), FVIIIc and TF expression, as
well as platelet activation and a reduction in antithrombin activity. These changes
can progress to encompass the syndrome of disseminated intravascular coagula-
tion. PET also carries an increased risk of IUGR, and women with a past history
of PET appear more likely to have higher circulating levels of coagulation
factors, lipids and insulin resistance. This may be reflected in a higher risk of car-
diovascular disease in later life for the mother, as well as a higher risk of vascular
*The risks associated with individual thrombophilias relate predominantly to retrospective studies, and in many
cases these have not been confirmed in all studies and particularly in prospective series.
†
The relative risk relates to the risk in the highest quartile compared with the lowest.
‡
The relative risk of PET with a waist circumference (at pregnancy presentation) of 80cm compared with a waist
circumference of <80cm, or of a BMI >26kg/m2 (at pregnancy presentation) when compared with a BMI <26kg/m2.
APA, Antiphospholipid; AT, Antithrombin; HBP, High blood pressure; PMH, Past medical history
131
Obstetric CH07 18/10/05 10:23 am Page 132
disease and Type 2 diabetes mellitus in later life in those infants with a history of
IUGR.
A link between a history of previous VTE and the risk of PET has been
reported in one study, and several case-control studies have found at least one
inherited thrombophilia in anything up to 70% of women with PET (when com-
pared with <20% in control subjects). A number of studies have specifically
examined aPC resistance (both FV Leiden and acquired) and the risk of PET. In
those studies reporting a positive link, the magnitude of the risk attributed to
heterozygous FV Leiden is between 2- and 5-fold. In some of these studies it has
also been suggested that thrombophilia may be associated with more severe PET.
In the main, however, a positive link between thrombophilia has only been seen
in retrospective studies, and prospective studies have not confirmed any link.
Consequently, further work is required to determine whether this disparity in
evidence relates to differences in the prevalence of FV Leiden in the different
populations, differences in the severity of PET in different studies, or to a more
accurate diagnosis in prospective investigations.
The prothrombin G20210A mutation has also been linked to an increased risk
of PET (between 2- and 7-fold) in a small number of studies, although the major-
ity of reports have not found any association. Protein S deficiency has also been
found in up to 25% of subjects with a history of PET. However, as there is a physi-
ological fall in protein S in normal pregnancy, the exact contribution of abnor-
mal levels to the development of PET requires further study. At present, due to
the relative rarity of the disorders, there is insufficient evidence to give an estim-
ate of risk for those with antithrombin (AT) or protein C deficiency. Although
MTHFR C677T has also been linked with around 2-fold risk of PET in some
reports, the majority of studies have not found an independent role for this
genetic variant. These studies suggest that levels of the hematinic cofactors of the
HCY–methionine pathway (such as folic acid) may be considerably more import-
ant than the C677T in the causation of disease.
Although this potential involvement of heritable thrombophilia in the causa-
tion of PET is intriguing, and may shed light on the pathogenesis of this disease,
there is no evidence that identification of thrombophilia carriers would reliably
identify those at risk of developing PET. Furthermore, there is no substantial
evidence that reduction of thrombin generation in such individuals, by the use of
anticoagulants, would result in a beneficial pregnancy outcome. At present, then,
there is no case that routine, or even selected, thrombophilia screening of indi-
viduals (based upon past medical or obstetric history) would be of benefit in the
management of this condition.
10th centile), with a relative 4–7-fold risk for carriage of either FV Leiden or pro-
thrombin G20210A. However, as with other pregnancy complications, a number
of studies have not shown any link between IUGR and heritable thrombophilia.
Further work, perhaps concentrating on births of <5th centile, are required
before a definite link with thrombophilia can be proven.
tal abruption and recurrent pregnancy loss, the vast majority of studies and met-
analyses have found that, despite a 3–4 increased risk of recurrent fetal loss asso-
ciated with hyperhomocysteinemia (see below), there appears to be little
independent contribution from the MTHFR C677T variant.
134
Obstetric CH07 18/10/05 10:23 am Page 135
Hyperhomocysteinemia
Mild–moderate hyperhomocysteinemia (hyperHCY) is found in around 5% of
North European populations, and may be twice as common in those with a
history of VTE and 2–5 times more common in those with atherosclerotic
disease. Although this is not a substantial risk, it is of interest, as hyperHCY may
be amenable to treatment with folic acid. However, the evidence that such inter-
vention reduces the risk of coronary artery disease is disputed and, at present,
there is no evidence at all that folic acid will substantially reduce the risk of recur-
rence of VTE. As noted in Chapter 3, the level of HCY is dependent on several
key enzymes in the HCY-methionine pathway, as well as the level of the vitamin
cofactors of these reactions (vitamins B12, vitamin B6 and folic acid).
135
Obstetric CH07 18/10/05 10:23 am Page 136
138
Obstetric CH07 18/10/05 10:23 am Page 139
References
Poort SR, Rosendaal FR, Reitsma PH, Bertina RM. A common genetic variation
in the 3´–untranslated region of the prothrombin gene is associated with ele-
vated plasma prothrombin levels and an increase in venous thrombosis. Blood
1996; 88(10): 3698–703.
139
Obstetric CH08 18/10/05 10:23 am Page 140
CHAPTER 8
Thrombocytopenia in
pregnancy
Maternal thrombocytopenia is commonly found in pregnancy. This is usually due
to physiological changes with increasing gestation (termed gestational thrombo-
cytopenia), or is caused by idiopathic thrombocytopenic purpura (ITP). Throm-
bocytopenia also occurs in association with a wide variety of both
pregnancy-associated and non-pregnancy-associated conditions (Table 8.1).
Gestational thrombocytopenia
Towards the end of pregnancy at least 5% of women have a platelet count below
the usual non-pregnant reference range (150–400 × 109/l). Although this gesta-
tional thrombocytopenia can be difficult to distinguish from ITP, the platelet
count is seldom <80 × 109/l (although counts as low as 50 × 109/l without
significant disease have occasionally been reported). Thus, the diagnosis of ges-
tational thrombocytopenia is most often made in the third trimester. It is con-
140
Obstetric CH08 18/10/05 10:23 am Page 141
Thrombocytopenia in pregnancy
Blood film to exclude platelet clumps, microangiopathic hemolytic anemia (MAHA) or other hematological
disorders
Coagulation screen (to include fibrinogen and D-dimer levels)
Renal and liver function tests
Antiphospholipid antibodies
Anti-DNA antibodies to exclude systemic lupus erythematosis (SLE) (ANA is sufficient as a screening test)
(Table 8.3). However, the main differential diagnosis is usually ITP or pre-
eclampsia, which account for around 4 and 21%, respectively, of cases of preg-
nancy thrombocytopenia. Clearly, it may be impossible to be certain that the
diagnosis is gestational thrombocytopenia rather than ITP, and in this situation it
is best to err on the side of caution and use the same precautions as in ITP to
minimize the risk of hemorrhagic complications in the baby (see Table 8.4).
Thrombocytopenia in pregnancy
Helicobacter pylori
Systemic lupus erythematosis (SLE)
Lymphoma chronic lymphatic leukemia (CLL)
Heparin-induced thrombocytopenia (HIT)
HIV
Post-transfusion purpura (PTP)
Drugs
143
Obstetric CH08 18/10/05 10:23 am Page 144
Helicobacter eradication
144
Obstetric CH08 18/10/05 10:23 am Page 145
Thrombocytopenia in pregnancy
Treatment of ITP
There have been few randomized clinical trials on the management of ITP in
pregnancy. As it is very likely that the platelet count will continue to fall as preg-
nancy progresses (reaching a nadir in the third trimester), and as spontaneous
bleeding problems are unlikely if the platelet count is >20 × 109/l, the manage-
ment of ITP in pregnancy is usually focused on achieving an adequate platelet
count for delivery, in particular to allow neuraxial anesthesia, and to minimize
the risk of fetal bleeding during labor and delivery.
The treatment options during pregnancy are similar to those used out-with
pregnancy, with due regard to the presence of the fetus. In general, if the
platelet count is >20 × 109/l and the patient is asymptomatic, then monitoring of
the platelet count and of the patient for bleeding is all that is required. If, at
delivery, the patient is asymptomatic then a spontaneous vaginal delivery or
Cesarean section could take place if the platelet count is >50 × 109/l (uncompli-
cated vaginal delivery is probably safe at levels >30–40 × 109/l, but as the risk of
Cesarean section is present in every labor it is best to aim for at least a level of
50 × 109/l). If the patient requires epidural anesthesia then a platelet count of
>80 × 109/l is recommended (Table 8.8). Following delivery, non-steroidal anti-
inflammatory drugs (NSAID) should be avoided for postoperative analgesia
unless the platelet count is >100 × 109 /l. It is also important to consider the
woman’s risk of venous thromboembolism (VTE), and for those considered at
increased risk, graduated elastic compression stockings and/or low-molecular-
weight heparin (LMWH) can be used. The latter is considered satisfactory if the
platelet count is >50 × 109/l, the patient has no hemorrhagic problems and has a
significant risk of VTE.
Firstline therapies
There are two firstline options currently used. These are oral corticosteroids and
intravenous human normal immunoglobulin (IVIgG). Both appear to have
145
Obstetric CH08 18/10/05 10:23 am Page 146
similar response rates – around 70% of patients will respond to IVIgG, with a
variable response rate for steroids, which is, at best, between 50 and 66%.
Clinicians vary in their choice, with some preferring IVIgG. This is most often
due to the perceived side effects of oral corticosteroids, although such side
effects are not usually critical with a short duration of high-dose steroids used in
anticipation of delivery. The cost of IVIgG and, more importantly, the fact that it
is a human blood product must also be considered. (Table 8.9). In general, if
treatment is to be required for a short time, then corticosteroids are a useful
option at a dose of prednisolone of 1mg/kg/day in the first instance, continued
for 2–4 weeks then tailing off the dose. If the duration of therapy is likely to be
longer (treatment starting in the first or second trimester), then IVIgG, at a dose
of 0.4g/kg/day for 5 days or 1g/kg/day for 2 days, has been used with an equiva-
lent response rate evident in at least one randomized trial. However, depending
on the product (see Chapter 2), there may be a higher risk of renal impairment
with the more intense schedule, and the first schedule is to be preferred if there
are pre-existing fluid balance problems or renal impairment. IVIgG most often
results in a response of 2–3 weeks duration and repeat dosing may be required if
a continuing hemostatic challenge is anticipated.
The mechanism of action of IVIg in ITP is largely unknown, but it may involve
the blockade of Fc receptors on macrophages and other effectors of antibody-
dependent cytotoxicity, or be due to the presence of anti-idiotype antibodies in
IVIgG which block autoantibody binding to circulating platelets. IVIgG may also
block Fc receptors on the placenta and so may prevent transfer of maternal
antiplatelet antibody but this is unproven in practice and remains a theoretical
benefit of IVIgG.
Secondline therapies
This includes high-dose methylprednisolone (1000mg) or azathioprine, which is
satisfactory for use in pregnancy, or a combination of these therapies with IVIgG.
Obviously, danazol, vinca alkaloids and cyclophosphamide are not suitable for
the pregnant patient. For patients who do not respond to conventional second-
line therapies, anti-RhD immunoglobulin (see Table 8.6), and even interferon
and mycophenolate mofetil or retuximab could be considered, but experience,
even in the non-pregnant, is very limited.
Splenectomy is best avoided in pregnancy, but, if considered essential, is best
carried out between 13 and 20 weeks gestation. At present, the role of lapro-
scopic splenectomy in pregnancy is not known. In 66% of non-pregnant sub-
jects, splenectomy will result in a normal platelet count. A number of studies
have suggested that the patient’s response to IVIgG may predict the response to
splenectomy. However, it is not known if this has any predictive value in preg-
nancy. The use of indium-labeled autologous platelet scanning to identify
whether the platelet destruction is splenic, hepatic or diffuse would not be a
favored option in pregnant subjects. This test is, however, the most sensitive pre-
146
Obstetric CH08 19/10/05 11:23 am Page 147
Table 8.9 – Treatment and outcome of thrombocytopenia in pregnancy
Thrombocytopenia in pregnancy
infection Mortality 0.6%
Pre-eclampsia (PET) Platelet/endothelial Supportive Allergy, fever, FBC None Yes
hemolysis, elevated activation (±platelets/fresh transmissible agents Day 1–5 Platelets
liver enzymes, and frozen plasma Intrauterine
low platelets (HELLP) (FFP)) growth restriction
147
(IUGR)
Deliver Abortion
Thrombotic thrombo- ADAMTS–13 Plasma exchange Fluid imbalance, BP Mortality – Yes
cytopenic purpura ±FFP Allergy, fever, 10–30%
(TTP)/hemolytic transmissible agents
uremic syndrome (HUS)
Acute fatty liver ? Long chain fatty Glucose Mortality Mortality <15% ?Yes
acid deficiency Fluid balance 5–20%
(LCHAD) General support
Liver transplant
Following splenectomy
Thrombocytopenia in pregnancy
avoided due to risk of intracranial bleeding if the baby is affected, but a straight-
forward mid- or low-cavity forceps delivery would be considered appropriate, if
required. At delivery, a cord platelet count should be determined in all babies
whose mother has been diagnosed with ITP. Close monitoring is required over
the next 2–5 days. In children with evidence of bleeding, or a platelet count
<20 × 109/l, IVIgG (at 1g/kg) produces a rapid response. Neonatal life-threaten-
ing hemorrhage may also require platelet transfusion.
Thrombocytopenia in pregnancy
FBC/Film
Reticulocyte count
Coagulation screen
Urea and electrolytes
LFT
LDH
Coomb’s test
Urinalysis
FBC, Full blood count; LDH, Lactate dehydrogenase; LFT, Liver function tests
present in around 50% of cases. The CNS signs, thrombocytopenia and hemoly-
sis are, however, more marked than with PET or HUS, but the hypertension may
not be severe (Table 8.12). TTP, particularly recurrent TTP, usually presents
before 24 weeks of pregnancy and intrauterine growth restriction (IUGR) and
fetal death is usual. As noted above, isolated episodes of TTP can occur and
relapse is common in pregnancy.
Although routine clotting tests are often normal in the early stages of
TTP/HUS, as the disease progresses there may be activation of coagulation and
disseminated intravascular coagulation (DIC). However, the accompanying eleva-
tion of D-dimer levels may often be difficult to distinguish from the elevated D-
dimer levels of normal pregnancy (see Chapter 1).
A comparison of the symptoms and investigations in thrombocytopenic dis-
orders is shown (Table 8.3). It has been reported that plasma antithrombin levels
may be of assistance in the distinction of PET/HELLP (reduced levels) from
TTP/HUS (normal levels) (Table 8.3). However, reduced and normal levels may
be seen in either condition, and establishing a reduced level requires a local
pregnancy-related reference range.
151
Obstetric CH08 18/10/05 10:23 am Page 152
Treatment of TTP/HUS
With perhaps the exception of bacterial endotoxin-related HUS (where support-
ive care is the main aspect of therapy) and recurrent or congenital TTP (see
below), it is unlikely that a clear distinction between the two syndromes of TTP
and HUS will be possible in the majority of pregnancy-related cases. As a con-
sequence, both will be considered predominantly as a single syndrome when con-
sidering therapy, particularly as there may be benefit in plasma exchange in
non-toxin-related HUS (Table 8.13).
Firstline therapies
The mainstay of treatment of TTP/HUS is plasma exchange, which removes the
large vWF multimers and autoantibodies, and supplements the vWF cleaving
enzymes. This has produced a remarkable reduction in the mortality of this con-
dition. The treatment schedules used in pregnancy are derived from those used
in non-pregnant subjects. Plasma exchange should be instituted within 24 hours
of presentation, as this has a bearing on prognosis, and subjects should be
managed in a facility that can provide an uninterrupted service. The optimal
number of plasma exchange procedures required is not known, but most author-
ities recommend that there should be a further two procedures after complete
clinical remission. Indeed, many facilities would taper off the exchanges rather
than stopping them abruptly, even after these two further procedures. Similarly,
the optimal exchange regime has not been determined, and either daily
exchanges of 1 or 1.5 plasma volumes for the first 3 days followed by 1 plasma
volume thereafter have both been used to good effect.
In addition, the optimum fluid replacement has not yet been ascertained.
Fresh frozen plasma (FFP) is the standard therapy in most countries. However,
cryosupernatant may offer some advantages over FFP. Cryosupernatant is derived
from the manufacture of cryoprecipitate and contains lesser amounts of the
larger vWF multimers, but may not be widely available. In view of the potential
risk of viral contamination of FFP, solvent detergent-treated FFP (SD-FFP) can
152
Obstetric CH08 18/10/05 10:23 am Page 153
Thrombocytopenia in pregnancy
also be used as the fluid replacement in plasma exchange. SD-FFP contains lesser
amounts of the larger vWF multimers, and may also carry a lesser risk of adverse
reactions, than standard FFP.
When plasma exchange is not immediately available, FFP infusion (at
30ml/kg), with due attention to the risks of fluid overload, may be beneficial
whilst exchange is being organized. In the rare congenital TTP with deficiency of
the cleaving metalloproteinases, exchange may not be required and FFP infusion
alone may be sufficient to induce remission.
Additional therapies
Any agent that could have precipitated TTP, such as clopidogrel, should be
stopped. As an immunoglobulin-mediated reduction in ADAMTS-13 activity
occurs in association with TTP, intravenous methylprednisolone has been used as
an adjunct to plasma exchange, although the benefit:risk ratio of this approach
in pregnancy has not been formally assessed. However, there is no specific con-
traindication to corticosteroid use in this situation.
Similarly, as many of the effects of TTP may be mediated through platelet activa-
tion, the use of antiplatelet agents is an attractive option. In this regard, there may
be some benefit in the empirical addition of aspirin to the plasma exchange regime
when the platelet count is restored to >50 × 109/l. Use of aspirin when the patient is
severely thrombocytopenic can, however, precipitate hemorrhagic complications.
All patients should receive folic acid and be vaccinated (when the platelet
count allows) against hepatitis B. Red cell transfusion should be given according
to clinical need, but platelet transfusions should be avoided in TTP. As with
other conditions associated with thrombosis, there may be a role for thrombo-
prophylaxis with LMWH, although the use of this requires an individual assess-
ment of the relative bleeding/thrombosis risk.
Secondline therapies
The patient is regarded as being in remission if there is a resolution of neurologi-
cal symptoms, the platelet count has normalized, the lactate dehydrogenase
(LDH) has reduced to within reference limits and the hemoglobin is rising.
Refractory disease would be defined as a failure of this after seven exchanges. If
the patient deteriorates, or does not respond to the initial plasma exchange
regime, a change to a higher plasma volume exchange (1.5 plasma volumes), a
higher frequency (12 hourly) or a different replacement fluid (cryosupernatant
or SD-FFP rather than standard FFP) has been recommended. The use of other
immunosupressive therapies such as vincristine, cyclophosphamide and
cyclosporine would be problematic whilst there is still a viable fetus and their use
in pregnancy would be on a case-by-case basis. Generally, if the patient has
reached this stage, she will usually have been delivered in either the maternal or
fetal interest, or have suffered an intrauterine death.
153
Obstetric CH08 18/10/05 10:23 am Page 154
Table 8.14 – Hemolysis, elevated liver enzymes, and low platelets (HELLP) summary
IU/l, International units/liter; LDH, Lactate dehydrogenase; RUQ, right upper quadrant; U/l, Units/liter
154
Obstetric CH08 18/10/05 10:23 am Page 155
Thrombocytopenia in pregnancy
Diagnosis of AFLP
The diagnosis of AFLP is suspected by a combination of clinical features (see
Table 8.3). In particular, the detection in the third trimester, of a cholestatic
pattern of liver function tests (LFTs), hypoglycemia and features of failure of syn-
thetic hepatic function (e.g. hypofibrinogenemia and a reduction in plasma
antithrombin levels) are suggestive of the disorder. The disease may be difficult
to distinguish from PET/HELLP and will require the exclusion of other causes of
hepatitis and confirmation of fatty infiltration in the liver by ultrasonography or
magnetic resonance imaging (MRI). In general, when compared to HELLP syn-
drome, AFLP is more likely to be associated with higher serum levels of aspartate
transaminase (AST) and hypoglycemia, and is more often accompanied by
marked hyperuricemia, DIC and leukocytosis. The combination of these features
will lead to a diagnosis in the majority of cases without the need for liver biopsy.
A potential causative mechanism is suggested by the link between AFLP and the
carriage of a fetus with an inborn error of metabolism (resulting in a failure to
oxidize long-chain fatty acids). Oxidation of fatty acids requires adequate function of
mictochondrial trifunctional protein (MTP). MTP is coded by chromosome 2 and
consists of three enzymes that carry out consecutive oxidation of free fatty acids. Free
fatty acid metabolism is used to generate glucose and eventually adenosine triphos-
phate (ATP). In the absence of free fatty acid metabolism and inadequate glucose, a
hypoglycemic lactic acidosis occurs, which is accompanied by infiltration (by abnor-
mal lipids) of the liver, skeletal muscle, myocardium, kidney, pancreas and lungs.
A number of cases of acute fatty liver of pregnancy have been associated with
the presence of fetal homozygous deficiency of one, or all three, of the com-
ponents of the MTP. However, how alterations in placental free fatty acid
metabolites lead to maternal disease is unknown, and further work is required to
determine the exact contribution of inborn errors of fatty acid oxidation to the
development of AFLP as well as PET/HELLP syndrome.
Treatment of AFLP
Treatment is aimed towards biochemical correction of hypoglycemia and fluid
balance, correction of any associated renal insufficiency, coagulopathy, hyperten-
sion, concomitant sepsis and preparation for delivery. Severe hepatic failure may
require the use of liver transplantation. It is known for AFLP to recur in sub-
sequent pregnancies, but the exact incidence is hard to estimate. This may be
due to the substantial mortality associated with the condition and the natural
reluctance of some survivors to undertake a further pregnancy.
Thrombocytopenia in pregnancy
Diagnosis of HIT
A variety of tests to determine the presence of antibodies against the combined
heparin-platelet Factor 4 antigen are employed. These are often based on
enzyme-linked iummunosorbent assay (ELISA) technology or platelet function
testing. The principal difficulty with these assays is the lack of standardization,
and often the lack of information about their sensitivity and specificity for HIT.
Because of this the diagnosis of HIT should remain principally a clinical one.
Prevention of PTP
Effective prevention in pregnancy would require universal screening to deter-
mine the HPA phenotype of pregnant women, as well as the presence of existing
maternal anti-HPA antibodies. It would also be necessary to supply appropriate
antigen-negative blood throughout pregnancy. At present, it would seem unlikely
that universal screening would be either cost or clinically effective. However, as
noted in Chapter 2, subjects already known to have such antibodies should be
transfused with appropriate HPA antigen-negative blood.
157
Obstetric CH08 18/10/05 10:23 am Page 158
Treatment of PTP
For treatment options see Chapter 2.
Sepsis
Major trauma
Placental abruption
Amniotic fluid emboli
Mismatched transfusion
Hepatic failure
Malignancy
Pancreatitis
158
Obstetric CH08 18/10/05 10:23 am Page 159
Thrombocytopenia in pregnancy
Treatment of DIC
The treatment, as in non-pregnant subjects, is directed to the cause of the
DIC. Supportive therapy with coagulation factor products and platelets may be
required if the patient is bleeding, or bleeding is likely. In amniotic fluid
embolism, and in disorders such as acute promyelocytic leukemia, the pre-
dominant manifestation may be ischemia and thrombosis. In these circum-
stances, anticoagulation may be required. Occasionally, there is both bleeding
and thrombosis, and a careful balancing act of therapeutic clotting factors
(avoiding concentrated products where possible) and anticoagulation is
required.
The coagulation and platelet product doses should be determined by fre-
quent laboratory monitoring, with the aim of restoring platelets to at least
50 × 109/l (perhaps even 80 × 109/l) and the coagulation screen to within refer-
ence limits, if the patient is bleeding. Traditionally, the fibrinogen is restored to
≥1g/l. Whether this is sufficient, given the higher reference range at the end of
pregnancy, is unknown. Other therapies, such as heparin, antithrombin concen-
trate, or tissue factor pathway inhibitor do not yet have a proven role in the man-
agement of DIC. Recent evidence suggests that recombinant activated protein C
therapy may have a beneficial effect on mortality from sepsis. However, a role for
such therapy in DIC in pregnancy is untested.
159
Obstetric CH08 7/11/05 2:13 pm Page 160
Diagnosis of NAIT
The prevalence of the HPA-1a-negative platelet phenotype is about 2%. In most
countries, there is no routine screening procedure for anti-HPA-1a, or anti-HPA-
5b, so the first affected baby is usually diagnosed after birth. Ten per cent of ICH
occur antepartum (often between 30 and 35 weeks gestation), and occasionally
NAIT will be diagnosed in utero, when investigations have been carried out for
other reasons. Sixty per cent of identified cases occur in first pregnancies that are
otherwise uneventful, making it difficult to predict who may be at risk. The clini-
cal presentation of the affected infant may vary from incidental thrombocytope-
nia and/or a purpura rash to the presence of ICH. NAIT can lead to
developmental changes in 25% of affected subjects, with more severe disease
resulting in motor dysfunction and intellectual impairment. The diagnosis may
also come to light after investigations to exclude other causes of fetal thrombocy-
topenia, such as infection, birth defects, chromosomal abnormalities, intrauter-
ine growth restriction (IUGR), hemangioma or a maternal thrombocytopenia. It
may also be suspected where there has been a history of fetal ICH, recurrent mis-
carriage or fetal hydrocephalus. A diagnosis of NAIT should only be made after
exclusion of maternal history of an autoimmune disorder, thrombocytopenia or
drug abuse.
Predicting NAIT may be difficult, as antibody testing can produce conflicting
results. A mother with a positive antibody test may have an unaffected infant and
a mother with a negative result may have an affected infant. However, any woman
with a history of severe disease, such as resulting in neonatal death, should be
considered at risk and, consequently, tested for platelet alloimmunization. If
160
Obstetric CH08 18/10/05 10:23 am Page 161
Thrombocytopenia in pregnancy
Treatment of NAIT
First pregnancy
As the platelet count may continue to fall in the first days after birth, infants
should be monitored daily. The presence of significant thrombocytopenia
requires ultrasound scanning for possible ICH. The passive thrombocytopenia
from NAIT persists for 1–3 weeks postpartum, depending on the rate of removal
of maternal platelet antibodies from the baby’s circulation. Compatible HPA-1a
negative or HPA-5b negative (as appropriate) allogenic platelets are most often
used for the neonatal treatment. In some instances, washed, irradiated (to
prevent graft versus host disease), maternal platelets may be used. If platelets are
unavailable, high-dose Ig therapy, at a dose of 1g/day for 1–3 days, has been
used. This requires 24–48 hours for an effect to be seen, but may be beneficial in
up to 75% of cases.
Second pregnancy
Two potential strategies for the management of a second pregnancy are:
the use of IVIgG ± steroids in the mother from around 24 weeks gestation to
inhibit antibody production and reduce placental transfer;
serial platelet transfusions for the fetus from around the same gestation.
At present, which management strategy is optimal is not yet clear. Indeed, a
combination of these strategies may also be useful.
IVIgG has been reported to improve the fetal platelet count in only 27% of
cases in some studies and in 66% in others, with an associated risk of ICH varying
161
Obstetric CH08 18/10/05 10:23 am Page 162
162
Obstetric CH09 18/10/05 10:23 am Page 163
CHAPTER 9
163
Obstetric CH09 18/10/05 10:23 am Page 164
The thalassemias
The thalassemias are a heterogeneous group of genetic disorders of hemoglobin
synthesis: they are named after the hemoglobin that is deficient and can be
described as shown in Table 9.2. The mutation may result in a reduced rate of
production from the affected gene (for the β gene the notation used for this is
‘β+’ and for the α gene the notation is ‘α+’), or result in no globin chain synthesis
at all (again for the β gene the notation for this is ‘β0’ and for the α gene the
notation is ‘α0’). The majority of thalassemias are inherited in a Mendelian reces-
sive manner and there are known mutations that affect hemoglobin transcrip-
tion, mRNA processing, translation and post-translational modification.
α Deletional
Non-deletional
β Deletional
Non-deletional
Normal HbA2 variant
Dominant variant
δβ Deletional
Non-deletional
γ –
δ Deletional
Non-deletional
164
Obstetric CH09 18/10/05 10:23 am Page 165
Given the diversity of genetic defects, and the possibility of genetic combina-
tions, thalassemias (irrespective of their molecular basis) are often classified by
their clinical effects into thalassemia minor, thalassemia intermedia and tha-
lassemia major. In general, thalassemia carriers are often symptomless and fall
into the minor category. Intermediate levels are more severely affected and may
often have anemia, although this does not require regular transfusion (Table 9.3).
In its major form, thalassemia presents with a lifelong transfusion dependency.
Alpha thalassemia
The α globin chain is common to both HbF and HbA, and deficiencies in the
gene (the alpha thalassemias), are found in subjects of Middle Eastern, Mediter-
ranean or African descent. As noted above, the human α globin gene cluster is
located on chromosome 16 and, normally, each individual has a total of four α
globin genes, with two on each chromosome. Although these genes are identical,
the α-2 globin gene location contributes significantly more α-globin chain pro-
duction to hemoglobin than the α-1 gene location. There are gene mutations
that can affect either one, or both, of the α globin genes on each chromosome.
Alpha thalassemia most often results from a deletion of these genes, although
non-deletional forms also occur (Table 9.4). The severity of the thalassemia phe-
165
Obstetric CH09 18/10/05 10:23 am Page 166
hemoglobins fail to deliver adequate oxygen to the tissues and these tetramers
also tend to precipitate within red cells, which shortens red cell survival by
hemolysis in the spleen (see Table 9.4).
Fetal
Third trimester stillbirth/neonatal death
Large placental volume
Pallor
Generalized edema
Marked hepatosplenomegaly
Other congenital anomalies
Hb 6–8g/dl
80% Hb Barts/20% Hb Portland
Maternal
Pre-eclampsia (PET)
APH and PPH due to placentomegaly
Survival to adulthood
Inheritance of ‘(--,-α)’ or homozygous non-deletion ‘α+α+’
Variable anemia
Variable splenomegaly
Periodic hemolysis with oxidant drugs or infection
Prominent red cell HbH inclusions
Hb 7–10g/dl
<40% HbH, variable HbA and HbA2
168
Obstetric CH09 18/10/05 10:23 am Page 169
the change from ζ-chains to α-chains occurs by the 7th week of gestation, there is
a lack of fetal oxygenation from early in development. This results in fetal
hypoxia, abnormal organogenesis and placental enlargement. A number of con-
genital anomalies have also been reported in association with Hb Barts, which
include abnormal brain, cardiac, skeletal or urinary tract development.
The clinical signs on Hb Barts-delivered fetus may vary from signs of cardiac
failure to gross edema, hepatosplenomegaly and pallor (see Table 9.5). Although
the vast majority of fetuses with Hb Barts hydrops fetalis die, there are a number
of cases of severe alpha thalassemia which survive, to be maintained on transfu-
sion and chelation regimes, with or without, the use of subsequent bone marrow
transplantation.
FBC +
Hb ELECTROPHORESIS
MCH >27 MCH <27 MCH <27 MCH <27 MCH <27 MCH <27
Folic acid
Avoid antioxidant drugs
If Hb <6g/dl consider transfusion
Treatment of pre-eclampsia (PET) and gallstones
Screening for infant carriage of thalassemia
Beta thalassemia
The beta thalassemias are found in those of Baltic, Mediterranean, Middle
Eastern, African, Indian, Southern Chinese and South-East Asian extraction.
Akin to the situation in alpha thalassemia, the defect of β-chains in beta tha-
lassemia leads to the formation of α-chain tetramers, which interfere with the
maturation of the red cell and result in red cell destruction within the marrow
and the spleen (Table 9.8). In beta thalassemia, this reduction in β-chains is
accompanied by an increase in δ-chain hemoglobin production, which is identi-
fied as an increase in the level of HbA2 (α2δ2).
In the homozygous, or compound heterozygous, beta thalassemic states, there
is a lifelong chronic dyserythropoietic anemia, which results in splenomegaly and
171
Obstetric CH09 18/10/05 10:23 am Page 172
172
Obstetric CH09 18/10/05 10:23 am Page 173
Fetal
At birth, absence of HbA is a feature of beta thalassemia major. Although there is
little evidence that early detection is cost or clinically effective, early detection
will allow the commencement of surveillance and the early onset of transfusion
therapy. When heel stab cards are used for diagnosis, storage changes may occur
if there is a long delay in the analysis of the neonatal sample, making identifica-
tion of hemoglobins difficult. If a cord sample is used for neonatal diagnosis,
cross-contamination with maternal blood may be a problem. This can be sus-
pected if there is a high level of HbA, but careful cleaning of the cord may
markedly reduce the risk of such contamination. Hemoglobin identification can
be achieved by cellulose acetate electrophoresis, although immuno-electro-
phoresis and high-performance liquid chromatography (HPLC) are more sensi-
tive methods. The nature of the genetic mutation can be determined by a
number of different PCR-based techniques, often using a combination of probes
that relate to mutations that are common in the ethnic population of the patient
under study. The exact screening technique used depends upon the ethnic mix
and knowledge of predominant mutations.
Thalassemia intermedia
This term covers a variety of thalassemic disorders, in which a combination of
defects results in an anemia that is more severe than that found in carrier states
but does not result in lifelong transfusion dependence (see Table 9.3). This clini-
cal definition includes HbC or HbE thalassemia, Hb Lepore and thalassemic syn-
dromes that arise as a result of a variety of combinations of α, β and δβ
deficiencies. As noted above, the inheritance of an α-chain mutation, along with
a homozygous β-chain disorder, will ameliorate the clinical picture by reducing
the β/α-chain imbalance and the consequent ineffective erythropoiesis.
175
Obstetric CH09 18/10/05 10:23 am Page 176
These combinations result in clinical syndromes which run from mild anemia
to the more obvious signs of a chronic hemolytic anemia, such as folate defi-
ciency, gallstones, leg ulceration, infection and extramedullary hemopoiesis
(with skeletal abnormality and splenomegaly).
graft rejection and graft failure. On account of this, when a transplant is being
considered for a young, otherwise fit, individual with a likelihood of a good
outcome, then full conditioning is probably to be recommended. Lesser induc-
tion regimens may be considered for the older, less fit, individual.
Sickle-cell disease
Sickle-cell disease is the most common hemoglobinopathy in the USA, with around
8% of African Americans carrying the sickle-cell gene. Disorders which result in
clinically significant sickling of the red cell result from the combination of a sickle-
cell gene on one β-chain combined with (on the other chromosome) either:
Another sickle-cell mutation (HbSS); or
An HbC mutation (HbSC); or
A beta thalassemia gene (HbSβ0).
Sickle-cell disease can result in a lifelong crippling hemolytic disorder (char-
acterized by crises caused by infection, aplasia, infarction and hemolysis) to a
diagnosis only detected on a routine blood film examination (Table 9.11). This
variation in symptoms can be due to the co-inheritance of other hemoglobin
changes, such as the HPFH or α thalassemia. The overall clinical expression of
the disease is also highly dependent on the availability of health care.
177
Obstetric CH09 18/10/05 10:23 am Page 178
Sickle-cell disease most often presents with infarction of the fingers in child-
hood, called dactylitis. There may be also recurrent infection or sequestration of
blood into the spleen causing profound anemia. However, due to repeated
infarction, it is unusual for there to be a palpable spleen by the teenage years,
and indeed there is, more often, hematological evidence of hyposplenism. These
infarction crises are often precipitated by dehydration or infection, and present
with pain which can be localized or diffuse. Childhood infection with any organ-
ism, but particularly parvovirus, may trigger an aplastic crisis with profound
anemia. In childhood, infarction crises may also present with a cerebral ischemia
of arterial origin (CIAO). There is also the possibility of sequestration of sickle
cells into the liver, spleen and lungs. Such lung sequestration will result in a
rapid deterioration in oxygenation and is a common cause of death.
With repeated crises there may be the development of bone deformity,
osteomyelitis, renal failure, myocardial infarction, leg ulceration, gallstones and
cardiac failure. Indeed, almost 30% of young adults with sickle-cell disease
develop a widespread vasculopathy with glomerulosclerosis, retinopathy, restric-
tive lung disease and stroke. With repeated transfusions there may also be an
increased risk of blood-borne infections and iron overload.
In SS disease, but more severely in the case of SC disease, there is an increased
risk of a proliferation of blood vessels in the retina (due to hypoxia), ultimately
causing vitreous hemorrhage, retinal detachment and blindness. This lifelong
series of painful crises and hospital admission also results in a disturbance of
schooling and development.
pregnancies resulting in a baby below the 50th centile for the population and, in
up to 27%, the baby is below the 10th centile. This may relate to the known
association of sickling disorders and placental infarction, although the risk of a
small baby has also been linked with the occurrence of anemic events during
pregnancy and inversely related to maternal HbF levels.
Anemic events do not appear to be limited to any specific gestation, but they
are more common in sickle/thalassemia combinations than they are in pure
sickle-cell disease (Table 9.12). In African Americans, the incidence of acute
chest syndrome appears not to be increased in pregnancy. There is, however,
likely to be an increased risk of premature rupture of membranes, pre-term deliv-
ery and postpartum maternal infection. Multiple pregnancies, as a result of a
reduction in available nutrition and oxygen transfer to the fetus, appear to be at
more risk of poor fetal growth. Whether the use of narcotic analgesia affects fetal
development is not clear, although some may have a vasoconstrictive effect on
the placental vasculature.
An increased risk of a growth-restricted baby and postpartum infection has
also been reported in subjects with SC disease, although there is a lesser need for
transfusion and fewer painful crises in SC than in SS disease. In developing coun-
tries the outcome of pregnancy in subjects with a major sickling disorder may be
substantially worse, with maternal mortality approaching 25% and a fetal mortal-
ity approaching 40%.
Blood loss
Infection
Vitamin deficiency
Aplastic crisis
Inflammation
179
Obstetric CH09 18/10/05 10:23 am Page 180
that the obstetrician and the mother are unaware of the problem. These women
are, however, at risk of severe sickling crises during pregnancy and the peurperium.
Pregnancy-specific management
At the first antenatal visit the presence of any factors which could affect preg-
nancy outcome should be assessed. This includes the past obstetric history, evid-
ence of chronic organ damage and the use of, and need for, narcotic analgesia.
At all stages a multidisciplinary approach involving obstetrician, hematologist,
anesthetist and general/infectious disease physician is required for appropriate
management and planning of pregnancy cases.
181
Obstetric CH09 18/10/05 10:23 am Page 182
Regional anesthesia, both spinal and epidural, appear safe for operative and non-
operative delivery, although epidural is often preferred. There must be, however,
adequate hydration to avoid any hypotension occurring secondary to sympathetic
blockade, but such blockade could, in theory, increase blood flow and reduce
the risk of vaso-occlusion. As noted above, there is a need to consider thrombo-
prophylaxis against VTE in such patients.
After delivery, there is a need to carefully attend to pain relief and hydration
to counteract the effects of increased metabolism and diuresis, and so avoid sick-
ling. In such circumstances, an acute chest crisis may be misdiagnosed as infec-
tion, fluid overload or a reaction to anesthesia.
Despite the improvements in feto-maternal outcomes in recent decades,
mothers with a major sickling disorder are still at risk of renal, cardiovascular and
cerebrovascular complications during pregnancy. In poorly developed countries
a number of factors are important in the success of pregnancies. These include:
adequate maternal nutrition; education on the nature of sickle-cell disorders; the
early recognition and treatment of urinary and respiratory infections; the mainte-
nance of adequate hydration and urinary output; the regular use of antimalarial
prophylaxis; systematic supplementation with multivitamins, including folate and
iron (depending on iron status); and the use of transfusion when the symptoms
of anemia are intolerable or appear likely to lead to cardiac failure. In these pop-
ulations, methods such as these may reduce the maternal and fetal mortality to a
level comparable with local non-sickle-cell patients and with sickle patients in
developed countries.
Other hemoglobinopathies
The combination of sickle-cell with HbC (HbSC) results in a lesser sickling dis-
order, although it carries a higher risk of retinopathy. The diagnosis can be sus-
pected by the presence of a large number of target cells and sickle cells on the
blood film and the presence of HbC on electrophoresis. However, both HbC and
HbE disease (without S) are associated with a relatively mild anemia and
splenomegaly, and often no specific treatment is required. Indeed, aside from
mild anemia, which may be exacerbated by iron or folic acid deficiency, both
HbCC and C/β0 syndromes are likely to be associated with a normal pregnancy
outcome.
Another Hb variant is HbD, which migrates to the same position as HbS on gel
electrophoresis. In homozygous DD there is a mild hemolytic anemia, and when it
occurs as a compound heterozygote with HbS it can result in a sickling disorder.
gene (see Chapter 4). Patients are most often well between the episodes of severe
hemolysis. G6PDH deficiency is also associated with neonatal jaundice in an
affected male. The mechanism for this is unclear, but the management is similar
to other causes of neonatal jaundice, and includes phototherapy and, where
necessary, exchange transfusion. There are a variety of methods for detection of
G6PDH levels in the red cell that can be used to distinguish this disorder from
other forms of Heinz-body-forming hemolysis.
The porphyrias
The porphyrias result from a number of inherited defects in the enzymes which
are required for the synthesis of heme. Although there are a wide variety of mole-
cular defects, seven clinical types of porphyria can be distinguished. These can be
considered by whether the defect results in the accumulation of porphyrins
(whose fluorescent properties result in the generation of free radicals and a
marked, often scarring, photosensitive skin rash), porphyrin precursors (which
result in neurological manifestations), or both. In some instances, the enzyme
defects are inducible, which can lead to an acute disorder. The exact diagnosis is
determined by the combination of detectable porphyrins and precursors in
plasma, erythrocytes, urine and stool. The effect on hemoglobin production may
be slight, as many of implicated enzymes are not rate limiting in heme biosynthe-
sis, although there may be a mild hemolytic anemia.
Porphyria cutanea tarda (PCT) First trimester has highest risk of symptoms
There may be glucose intolerance
There may be associated hepatitis C or B
Avoid alcohol
Avoid sun
No routine iron
If symptoms, venesect
If refractory symptoms, consider chloroquine
If symptoms:
Remove precipitant
Hydrate patient
Carbohydrate supplements
Hematin
Pain control
Seizure treatment
185
Obstetric CH09 18/10/05 10:23 am Page 186
CEP
In CEP there is often severe, cutaneous, scarring, photosensitivity that is evident
from childhood. This is accompanied by a normochromic, normocytic anemia,
with basophilic stippling evident in the red cell and nucleated erythrocytes a
common feature.
Standard suncreams are often ineffective, as they do not screen out the wave-
lengths of around 400nm that lead to the photosensitivity. Reflective creams,
with zinc or titanium, may be effective. Other strategies such as activated char-
coal (binding bile porphyrin) and hypertransfusion (to suppress endogenous
erythropoiesis) have been used successfully in some cases. If anemia is a problem,
it may respond to splenectomy.
One of the CEP mutations is associated with severe disease, which can result
in non-immune hydrops fetalis, or transfusion-dependant anemia from birth. In
many cases, prenatal diagnosis from amniotic cells is possible.
PCT
PCT results from a reduction in activity of one of the heme synthesis enzymes in
the liver. This leads to an accumulation of uropophyrins in the plasma, which are
excreted in the urine. PCT results in marked skin fragility in light-exposed areas,
facial hypertrichosis and hyperpigmentation. It is not, however, associated with
neuropsychiatric disturbance. It presents late, often in association with alcohol
excess, iron overload and/or hepatitis B or C carriage.
Exogenous estrogen is associated with expression of PCT and it has also been
diagnosed for the first time in pregnancy, although there is debate as to whether
pregnancy exacerbates the disease or not. If exacerbation does occur, this is
likely to be in the first trimester, as there is a reduction in urine porphyrin and
placenta-derived estrogen and progestogen production in the second trimester.
In pregnancy, it would be sensible to monitor known cases (as there may be a
greater risk of glucose intolerance), to minimize exposure to alcohol and sun-
light, and to refrain from iron supplementation. In non-pregnant subjects, avoid-
ance of alcohol, removal of iron by venesection, recombinant erythopoietin and
treatment of hepatitis C, if present, with α-interferon have all been reported as
186
Obstetric CH09 18/10/05 10:23 am Page 187
EPP
EPP results from a defect in ferrochelatase, which is the last enzyme in the heme
pathway. The level of the enzyme does not, however, correlate well with symp-
toms. EPP most often becomes evident in childhood, with exposure to sunlight
leading to itching and burning, and local erythema. It can also present in the dif-
ferential diagnosis of a sideroblastic anemia and, in later life, may lead to gall-
stones due to precipitated protoporphyrin. Rarely, it may manifest with a
neurological crisis resembling AIP.
EPP is diagnosed by the detection of elevated levels of erythrocyte-derived free
protophorphyrin. Although elevated levels also occur in iron deficiency and lead
poisoning, the levels in EPP are usually considerably higher than those in iron
deficiency (normal <50µg/dl, iron deficiency anemia (IDA) <300µg/dl, EPP and
lead poisoning 300–4500µg/dl), but unlike lead poisoning and iron deficiency,
the protophorphyrin of EPP is non-chelated.
Treatments include avoidance of sun exposure, zinc- or titanium-based sun-
screens, induction of hypercarotenemia (which can reduce photosensitivity),
hematin therapy (which reduces the production of porphyrins and their precur-
sors), cholestyramine (which binds bile protoporphyrins) and activated charcoal.
It is possible that there may be an improvement in symptoms in pregnancy,
although this requires confirmation and the mechanism of such an improvement
is unknown.
AIP
AIP results from a decreased conversion of porphobilinogen to prophyrins. It is
transmitted as an autosomal dominant condition, with the majority of cases due
to single base mutations. The mechanism of neurotoxicity is unknown, but could
be due to a rise in the porphyrin precursor 5-aminolevulinic acid (which may
inhibit neurotransmitters), or a depletion of cellular heme (leading to reduction
in energy production in neural tissues).
The condition is characterized by the acute onset of autonomic neuropathy,
which commonly presents with abdominal pain, constipation, tachycardia, hyper-
tension and vomiting. This may be accompanied by acute anxiety, confusion, psy-
chosis and seizures. In 50% of subjects, persisting hypertension and renal
impairment may also occur. The blood count is usually normal aside from a
raised white cell count in the acute phase. Both 5-aminolevulinic acid and por-
phobilinogen can be detected in the urine, especially during an acute attack.
A number of circumstances can lead to an acute attack. These include
gonadal hormones (particularly progestogens), decreased caloric intake, alcohol
187
Obstetric CH09 18/10/05 10:23 am Page 188
188
Obstetric Index 19/10/05 10:19 am Page 189
Index
ABO antigens 23 diagnosis in pregnancy to FVIII 66
ABO blood system 15 169–70 in hemolysis 23, 24
activated clotting factor fetal carriers 168 HIT 95
concentrates 78 diagnosis 170 warm 55, 56, 57
activated partial fetal HbH disease 168 see also autoantibodies
thromboplastin time genetics 166–7 anticoagulation, therapeutic
(APTT) 14 maternal carriers 167 61
and activated protein C maternal HbH disease long-term oral 107
resistance 134 167–8, 170–1 new anticoagulants 100–1
in afibrinogenemia 84 mechanism 165 and prosthetic heart valves
in dysfibrinogenemia 86 pregnancy, problems in 166–7 fetal outcomes 106
in fluid replacement therapy severity 166–7 maternal outcomes 105
42 treatment 170–1 see also specific agents
in hemophilia 77, 78 amniocentesis antiphospholipid syndrome
in pregnancy 10 bilirubin level determination 99, 137–9
prolongation in hemophilia A 34 definition 137
67 in hemophilia diagnosis 71 management options 138
in von Willebrand’s disease anagrelide 59 and pregnancy outcome
81 anamnestic response 16, 23 138–9
activated protein C (aPC) 6 anemia 43–8 antiplatelet agents 60, 153
therapy 159 autoimmune hemolytic 20–1 antithrombin (AT) 6, 8, 10
activated protein C resistance dilutional 1 and fetal loss 133
(aPCR) 13 fetal loss 34 in pregnancy 10
in pregnancy 10, 11, 134–5 fetal assessment 34 and thrombophilia
outcome 135 folate deficiency 51 assessments 126
and thrombophilia Heinz body 183–4 antithrombin deficiency 124,
assessments 126 iron deficiency 43–8 127–8
acute chest syndrome 179 diagnosis 45–7 antithrombotic therapy
acute fatty liver of pregnancy in pregnancy 44–8 88–101
(AFLP) 155–6 prevention 47 antitrypsin 6, 8, 10
diagnosis, differential 142 treatment 47–8 aplasia, acute 51
mortality 155 macrocytic 50–1 argatroban 98–9
treatment/outcome 147 megaloblastic 51, 54 arterial thrombosis 102–4
acute intermittent porphyria pernicious 54 in pregnancy 102–4
(AIP) 187–8 physiological 1 risk in young women 102
in pregnancy 185, 188 in sickle-cell disease 178, 179 Ashkenazi Jews 76, 132–3
acute myocardial infarction anesthesia in sickle-cell disease aspirin 99–100
108–11 182–3 in acute myocardial infarction
diagnosis/management angiotensin-converting enzyme 110
110–11 (ACE) inhibitors 110 in CIAO 108
afibrinogenemia 84 anti human leukocyte antigen and heparins 99
age and VTE risk 128 23, 24 in TTP/HUS 153
AIHA see autoimmune anti-RhD immunoglobulin autoadsorption 21
hemolytic anemia (AIHA) immunoprophylaxis 36–7, autoantibodies 20, 55
allergic reaction 144 removal 21
to heparins 97 antibodies autoimmune hemolytic anemia
to transfusion 22, 25 antiplatelet 143 (AIHA) 20–1, 55–8
alloantibodies 20–1 cold 56, 57 causes 57
alpha thalassemia formation 16 in pregnancy 56–7
Obstetric Index 19/10/05 10:19 am Page 190
Index
190
Obstetric Index 19/10/05 10:19 am Page 191
Index
191
Obstetric Index 19/10/05 10:19 am Page 192
Index
192
Obstetric Index 19/10/05 10:19 am Page 193
Index
193
Obstetric Index 19/10/05 10:19 am Page 194
Index
194
Obstetric Index 19/10/05 10:19 am Page 195
Index
195
Obstetric Index 19/10/05 10:19 am Page 196
Index
196