Anda di halaman 1dari 18

Blood Reviews (2000) 14, 4461

2000 Harcourt Publishers Ltd


DOI: 10.1054/ blre.1999.0123, available online at http://www.idealibrary.com on

State of the art

RhD haemolytic disease of the fetus and the


newborn

S. J. Urbaniak, M. A. Greiss

When an RhD negative mother is exposed to the RhD positive red cells (usually as transplacental
haemorrhage), she develops allo-anti-D which crosses the placenta and then results in the destruction
of fetal red cells. Clinical manifestations of RhD haemolytic disease (HDN) range from
asymptomatic mild anaemia to hydrops fetalis or stillbirth associated with severe anaemia and
jaundice. HDN was a significant cause of fetal mortality and morbidity until the introduction of
amniocentesis, intrauterine transfusion, controlled early delivery and exchange transfusion in the
management of severely alloimmunised women and their fetuses. The objective of monitoring
alloimmunised women is to identify fetal anaemia and prevent the development of life-threatening
hydrops. Evaluation involves assessing the history of previous pregnancies; serial estimation of
maternal anti-D levels; serial ultrasound measurements; serial amniocentesis; fetal blood sampling,
and intrauterine transfusion when indicated. Diagnostic genotyping by DNA-based methods can
identify at-risk RhD positive fetuses early in gestation. Identification of transplacental haemorrhage
(TPH) as the stimulus for anti-D antibody production led to the development of anti-D
immunoglobulin prophylaxis for at-risk RhD negative women who are not already alloimmunised.
Prevention includes administration of anti-D immunoglobulin for any event associated with TPH
during pregnancy, and at delivery of an RhD positive infant. Prophylactic routine administration of
anti-D immunoglobulin at 28 (and 34) weeks gestation, in addition to the above, has reduced
alloimmunisation to <1% of RhD negative women carrying an RhD positive fetus. 2000 Harcourt
Publishers Ltd

HISTORICAL BACKGROUND haemolytic anaemia (AIHA), haemolytic transfusion


reaction (HTR), since it involves the production of
In vivo haemolytic disease of the newborn (HDN) is antibody(ies) in one individual (maternal) and cell
the most complex of the three common forms of IgG- destruction in another (fetus).
mediated red cell destruction, i.e. HDN, autoimmune Allegedly, the first description of a clinical condi-
tion called hydrops fetalis was in 1609,1 when a
French midwife called Louise Bourgeois described
Stanislaw J. Urbaniak PhD, FRCP, FRCPE, FRCPath Professor
the birth of twins: the first was oedematous and died
of Transfusion Medicine, and Director of Academic Transfusion immediately after birth, the second became deeply
Medicine Unit, Department of Medicine and Therapeutics, jaundiced (icterus gravis) and died a few days later.
Aberdeen University, UK. Michel A Greiss MBBCh, PhD Clinical
Lecturer, Academic Transfusion Medicine Unit and Associate
Over the centuries, this clinical picture was recognised
Specialist, NE Scotland Regional Transfusion Centre, Foresterhill, and reported as two separate conditions (Table 1). It
Aberdeen, UK was not until 1932 that Diamond et al.2 realised that
Correspondence to: S.J. Urbaniak congenital anaemia, icterus gravis and hyrdrops

44
RhD haemolytic disease 45

Table 1 Summary of historical events

16091 Louise Bourgeois (midwife) first described hydrops fetalis


19322 Diamond et al. realised that congenital anaemia, icterus gravis and hydrops fetails were manifestations of the same disease,
which they named erythroblastosis
19343 Hawskley & Lightwood recognised that hydrops fetalis, haemolytic anaemia and icterus gravis are variants of the above
19444 Gilmour noted that erythroblastosis fetalis is equivalent to HDN
19385 Darrow noted haemolysis was due to transplacental passage of a maternal antibodies to the fetal circulation
19396 Levine & Stetson found an antibody that caused HDN
19407 Landsteiner & Weiner described the Rhesus blood group
19548 Chown noted that mothers were sensitised following an FMH
19669 A combined study on preventing HDN by anti-D prophylaxis from centres in England & Baltimore
196810 Pollack et al. Results of clinical trails of RhoGAM in women
197111 WHO Prevention of Rh sensitization
199812 Consensus conference on anti-D prophylaxis

fetalis were manifestations of the same disease, which that 25 g (125 iu) of anti-D Ig should be given i.m. to
they named erythroblastosis fetalis. It was also only cover a TPH of 1 ml RhD positive packed red cells or
comparatively recently confirmed that hydrops fetalis, 2 ml of whole blood.11 Most recently, this was reinforced
haemolytic anaemia of the newborn and icterus gravis by the joint working group of BBTS and RCOG.12
were all recognised to be variants of the same disease When RhD positive cells from the fetus pass into
(Hawksley & Lightwood3 and Gilmour4) now univer- the maternal circulation of an RhD negative mother,
sally referred to as haemolytic disease of the newborn this results in the production of antibodies, which
(HDN). The term erythroblastosis fetalis, which was cross the placenta to the fetus circulation and cause a
at one time widely used to describe the whole clinical variety of effects in the fetus ranging from very mild
syndrome, more correctly refers to the finding of to severe haemolytic anaemia and, on occasion,
extramedullary haemopoiesis and erythroblastaemia. intra-uterine death (IUD).
In 1938, Darrow5 postulated that the haemolysis was The most effective treatment is prevention of initial
due to transplacental passage of a maternal antibody RhD alloimmunization by prophylactic administra-
into the fetal circulation. One year later (1939), an tion of passive anti-D immunoglobulin. The mecha-
antibody was discovered by Levine & Stetson6 after an nism of the antibody-mediated suppression is
unusual transfusion reaction; there was an immediate incompletely understood (see later) but the most likely
reaction in a blood group O woman who had received mechanism is a negative modulation of the primary
her husbands group O blood, shortly after delivery of immune response. Since antigenantibody complexes
a stillborn fetus with erythroblastosis. The events sug- are bound to cells bearing Fc receptors in the lymph
gested that the infant had inherited a red cell agglu- nodes and spleen, these cells presumably stimulate
tinogen from the father which was foreign to the suppressor T cell responses, which prevent antigen-
mother. In 1940, Landsteiner & Weiner,7 using rhesus induced B cell proliferation and antibody formation.
monkeys, discovered the Rh antigen. They observed Alternatively, the splenic macrophages remove anti-D
that red blood cells from 85% of human subjects coated red cells prior to contact with the dendritic
agglutinated in the presence of rhesus monkey red cell antigen-presenting cell (APC), which have low FcR
antiserum, whereas the 15% of subjects who lacked expression, and the immune response is inhibited by
the antigen on their red cells did not. Nevertheless, it antigen deviation.1,1315
was not until 1954 that Chown8 reported that mothers Once sensitisation has occurred the following
were sensitised by a fetomaternal haemorrhage therapeutic options are open to the pregnant woman:
(FMH), and the association was made between fetal controlled early delivery, intrauterine transfusion
RBCs and the stimulation of maternal antibodies. (IUT)/direct intravascular transfusion (IVT), plasma
The FMH or transplacental haemorrhage (TPH) had exchange (PE), and/or intravenous immunoglobulin
been shown to occur mainly at delivery, and two (IV IgG). The IUT procedure is not without risk, and
groups in the UK and the USA were able to demon- with the reduced incidence of HDN is likely to
strate in clinical trials that immunisation in RhD become more hazardous as those skilled in the proce-
negative women could be suppressed, when concen- dure become fewer. With the recent advances in fetal
trated anti-D IgG was given intramuscularly (i.m.) medicine, introduction of direct intravascular transfu-
soon after delivery.9,10 Because of the difficulty in sion and improved neo-natology in special centres, the
measuring the TPH volumes with a degree of accu- survival of affected fetuses with HDN is now greater
racy, and to err on the safe side, it is recommended than 90%.13,1621
46 Blood Reviews

Table 2 Frequency of possible Rh alleleic combinations

Nomenclature Ethnic group

FisherRace Weiner Whites Blacks Asian

Rh Positive CDe R1 0.42 0.17 0.70


cDE R2 0.14 0.11 0.03
cDe Ro 0.04 0.44 0.03
CDE Rz 0.00 0.00 0.01
Rh Negative cde r 0.37 0.26 0.03
Cde r 0.02 0.02 0.02
cdE r 0.01 <0.01 <0.01
CdE ry <0.01 <0.01 <0.01

SCIENTIFIC BASIS A number of variants exist for each of the common


Rh antigens, e.g. Du, Cw, Cx, Ew. The most clinically
Rh antigens: structure and inheritance significant antigen D is a mosaic composed of at
least 30 determinants as identified by monoclonal
The Rh blood group antigens are associated with anti-Ds.27 The lack of expression of certain of these D
nonglycosylated human erythrocyte membrane pro- epitopes at the surface of variant red blood cells deter-
teins of molecular mass 30 KDa (the Rh poly- mines the so-called D category phenotypes which
peptides) and a glycoprotein of 40100 KDa (the arise from genomic rearrangements of the D and CE
Rh-associated glycoprotein). The precise function of genes.15,28,29 The Du or weak D phenotype is of par-
these Rh antigens are unknown. The Rh polypeptides ticular importance and is common in black popula-
are embedded in the lipid phase of the red blood cell tions. In most individuals weak D differs from normal
membrane and may interact with the membrane D only in having fewer antigenic sites per red cell, and
ATPase, functioning as part of a cation or proton these cells react weakly/variably with different anti-D
pump to control fluid and electrolyte fluxes across the sera.30 Molecular analysis of weak D phenotype
cell membrane.13,18,22 However, rare individuals with- shows that the majority have altered RhD proteins
out any Rh antigen (Rhnull) have defective red cell resulting in diminished expression of the D antigen.31
membranes associated with a degree of haemolytic Some weak D phenotypes are in fact D category vari-
anaemia.13,15,18,2325 ants and these individuals can be immunised to form
The human Rh locus is composed of two highly anti-D and should be identified as D variant at a
related genes RHD and RHCE (RhCcEe proteins), reference laboratory.
encoding the D, Cc and Ee blood group antigens If a structural D variant is excluded, it is not neces-
respectively. The expression of D indicates an RhD sary to treat such weak D individuals as RhD negative
positive person and the absence of D denotes an Rh for transfusion purposes. Similarly, weak D individu-
negative person. The genes which determine the anti- als are not considered candidates for either antenatal
gen are inherited as 2 haplotypes consisting of three or postnatal prophylaxis with passively administered
alleles, one haplotype being inherited from each par- anti-D immunoglobulin. However, if there are no
ent, CDe (phenotype designation R1) 4042%, cde facilities for excluding structural D variants then it is
(r) 3537%, and cDE (R2) 1416% being the most safer to treat all weak D individuals as being RhD
common in Caucasians (Table 2). Of the approxi- negative.15,18,29,32
mately 45 Rh antigens, D is the major cause of Rh
incompatibility. It is estimated that 1517% of whites,
57% of blacks and about 2% IndoEurasians do not Immunogenic factors
express the D antigen and so are called Rh-negative; The factors which influence the immune response to
RhD negative individuals (as determined serologi- the RhD positive cells are:
cally) are < 1% amongst indigenous Chinese.13,15,18
Recent molecular analysis shows that serologically 1. Concurrent ABO incompatibility can protect the
defined RhD-negative individuals of some ethnic mother against immunization, presumably because
groups, e.g. South African blacks (and their descen- leaked fetal red cells are promptly coated by
dants) carry an RHD pseudogene which does not circulating isohaemagglutinins (IgM) and
express RhD antigen.26 (probably) complement, and then removed from
RhD haemolytic disease 47

Fig. 1 Mechanism of Rh sensitisation and the effect of ABO incompatibility.

the circulation by the mononuclear phagocyte 2. The critical factor determining the magnitude of
system (MPS), mainly in the liver, which is less antibody response is the dose of immunizing
immunoresponsive than the spleen and therefore is antigen, i.e. the volume of RhD positive RBC.
less likely to stimulate antibody production.1,13,18 Haemolytic disease develops in neonates only
(Fig. 1) when an RhD negative mother has experienced a
48 Blood Reviews

significant transplacental haemorrhage (TPH). anti-D responses.13,18,22 However, recent evidence indi-
Although the average TPH occurring at delivery is cates that T-cell responses to RhD peptides is
less than 1 ml of whole blood, approximately 50% restricted by MHC class II.38
of all women with ABO-compatible pregnancies
have detectable circulating fetal red cells: only 19%
Sensitisation and development of allo anti-D
of those with ABO-incompatible pregnancies have
detectable circulating fetal red cells.1,9,14,18 Between 1940 and 1970 dramatic progress was made
3. The antigen expression on the RBC influences in reducing the mortality from HDN, from 50% to
immunogenicity, e.g. R2r infants are more effective 59% of all pre-natal deaths. Contributory events
in sensitizing their mothers to RhD than are were the introduction of Coombs antiglobulin tech-
infants of other phenotypes, since the R2 nique, exchange transfusions, amniocentesis, con-
phenotype expresses most D antigen.13,15,18 trolled premature delivery, and intrauterine
4. The immune responsiveness of the recipient, e.g. transfusion. By 1970 it was not possible to reduce
some women produce potent anti-D in a first further the number of babies which were affected by
pregnancy sufficient to cause severe hemolytic RhD hemolytic disease. Although since then the
disease. introduction of prophylactic anti-D has led to dra-
matic reductions in the incidence of cases, Rh HDN
The D antigen is a high-incidence and strongly does still occur mainly as a result of: (1) already
immunogenic antigen, 50 times more so than the immunised women subsequently becoming pregnant;
other Rh antigens. The incidence of primary RhD (2) failure to administer an adequate amount of anti-
immunization also depends on the dose of RhD posi- D immunoglobulin after a sensitizing episode (e.g.
tive red cells: 15% after 1 ml and 7090% after 250 ml abortion, amniocentesis, normal delivery); and/or (3)
given intravenously.33,34 On the other hand secondary failure of the anti-D immunoglobulin to protect even
immune response may occur after exposure to much when given correctly.1,13,16,18 In Rh, and other forms of
smaller amounts (as little as 0.03 ml of RhD positive HDN, the antibody is produced as a result of sensiti-
red cells).1,13,18,35 sation by pregnancy or transfusion. However, expo-
However, the immune system is not fully developed sure to an antigen does not necessarily cause
at birth. Infants less than four months of age appear production of an antibody, and a series of small
to be incapable of producing antibodies in response to immunizing doses (immunological challenges) is more
multiple transfusions,36,37 and this may be due to the likely to initiate antibody production than a single
immaturity of several lymphocyte and monocyte dose. Sensitisation through pregnancy occurs partly
functions. due to a small fetal bleed (fetal maternal haemor-
Nevertheless there is considerable variation among rhage [FMH]) into the mothers circulation via the
adults in immune responses to alloantigens presented placenta (TPH) and a larger volume at delivery when
during pregnancy, and the ability to produce antibod- the placenta separates from the uterus.1,13,39
ies, once developed, does not regress with age. These In the fetus the RhD antigen is well developed by
variations may be due to: (1) variable antigenicity of 3040 days gestation, and FMH can occur antenatally
epitopes; (2) the role of the placenta, i.e. the route of from as early as six weeks.40 Women immunised after a
exposure to antigen; and (3) variable maternal relatively small TPH or following an abortion (spon-
immune responsiveness. taneous or therapeutic) are referred to as good
Immune responsiveness is determined, at least in responders, and any RhD positive fetuses in subse-
part, by products of the major histocompatibility quent pregnancies may be severely affected. Evidently
complex (MHC) genes particularly HLA-DR genes the risk of TPH is increased as the pregnancy pro-
(class II). Several studies have sought to associate gresses (3% in the first trimester, 43% in the second
HLA phenotype and immune response to various trimester, 64% in the third trimester).1,16,39 For unex-
antigens, e.g. the HPA-1 platelet antigen has been plained reasons, a non-immunised unprotected RhD
strongly associated with the HLA-B8, DR3 haplo- negative mother has only a 16% chance of becoming
type.13,18 On the other hand, the production of anti-D immunised against her RhD positive fetus in any
has not been shown to correlate significantly with given pregnancy. Among these are RhD negative
HLA type. Perhaps this inability to demonstrate a mothers with RhD positive ABO compatible and
close relationship between HLA and blood group incompatible babies (approximately 8% and 2%
alloimmunisation is because the immune response is respectively) who will produce anti-D following
also influenced by genes independent of HLA, such delivery of their first baby; and another 8% will have
as T-cell receptor genes, and the T cell repertoire of an anamnestic (secondary) response in their second
the individual might contribute to the variability of RhD positive pregnancy.13,18,22 The primary response
RhD haemolytic disease 49

Table 3 Events following which anti-D Ig must be given to all RhD-negative women with no RhD
antibody and/or with antibodies other than anti-D

l delivery of an RhD positive infant* l antepartum haemorrhage (APH)

l abortion (see Table 4) l external version of the fetus

l invasive prenatal diagnosis l closed abdominal injury


amniocentesis
chorionic villus sampling (CVS) l ectopic pregnancy
fetal blood sampling (FBS)

l other intrauterine procedures l intrauterine death (IUD)


insertion of shunts
embryo reduction l stillbirth
*also if the RhD type of the infant has not been determined or is in doubt, and
the mother is to be discharged
Dose: before 20 weeks gestation 250 IU (50 g)
after 20 weeks gestation 500 IU (100 g)
In conjunction with a Kleihauer or equivalent test to assess the size of any TPH

Table 4 Prophylactic anti-D following abortions to all RhD negative women with no RhD antibody
and/or with antibodies other than anti-D

l therapeutic termination of pregnancy


l spontaneous abortion followed by instrumentation
l spontaneous complete or incomplete abortion after 12 weeks gestation
l threatened abortion before 12 weeks
when bleeding is heavy or repeated or is associated with abdominal pain, in particular, if these
events occur as gestation approaches 12 weeks
l threatened abortion after 12 weeks all women are eligible; in addition:
when bleeding continues intermittently after 12 weeks gestation, anti-D Ig should be given at
approx. 6 weekly intervals, and assess the size of TPH

is usually weak and often IgM antibodies, which do Anti-D IgG associated with HDN is produced
not cross the placenta, are produced; thereafter the only as subclasses IgG1 and/or IgG3. Cases of
majority will convert to IgG antibodies. There is HDN caused by IgG1 anti-D reputedly involve a
currently no practical way of preventing a pregnant or more serious anaemia than those caused by IgG3
transfused woman from becoming immunised to red anti-D presumably due to the longer exposure
cell antigens other than prophylaxis with administra- of fetal red cells in utero to IgG1 than to IgG3
tion of anti-D immunoglobulin. It is not always anti-D. Since the infants post-delivery bilirubin
remembered that potentially sensitizing episodes other level in HDN caused by IgG3 anti-D is often greater
than delivery are indications for administration of than that in IgG1-induced HDN it is conceivable
intra-partum anti-D immunoglobulin (e.g. abortion, that, once it reaches the infants circulation, IgG3
ante-partum haemorrhage, amniocentesis, external anti-D causes greater red cell destruction than does
version and adventitious trauma see Tables 34.). IgG1.13,18

Role of IgG subclasses Mechanisms of red cell destruction


The concentration of all four IgG subclasses has been The mechanisms of immune red cell lysis are basically
found to be slightly higher in cord than in maternal the same whether mediated by autoantibodies, i.e.
serum. IgG1 crosses the placenta early in pregnancy AIHA, allo-antibodies destroying transfused donors
and by 20 weeks gestation maternal IgG1 is detectable RBCs i.e. HTR, or transplacental transfer of allo-
in cord serum at levels which equal, or exceed, those in antibodies, i.e. HDN (Table 5). Binding of red cell
the maternal serum. In contrast, the level of IgG3 in antibodies to their antigenic determinants on the cell
cord serum does not reach the maternal concentration membrane does not directly damage red cells
until 2832 weeks gestation, and may never rise higher Although IgG Rh antibodies are almost exclusively
than this during the entire pregnancy.13,18,41 IgG1 and IgG3, they very seldom bind complement,
50 Blood Reviews

Table 5 Antibody-mediated RBC destruction

RBC + (IgM) antibody + C1C9 complement activation intravascular lysis


RBC + (IgG or IgM) antibody + complement activation to C3 removal by mononuclear phagocytic system (mainly liver)
RBC + (IgG) antibody directly removed by mononuclear phagocytic system (mainly spleen)
RBC + (IgG) antibody antibody-dependent cell-mediated lysis

possibly because of steric hindrance due to the loca- receptors, thus providing optimal interactions
tion of the Rh antigens within the RBC membrane. It between opsonised erythrocytes and macrophages.
has been suggested that Rh antigens are mobile within The sensitised red cell is either wholly phagocy-
the membrane and may cluster during antigen- tosed, or it loses part of its membrane (scission) as a
antibody reactions.14,15 It is possible that this ability of result of attack by macrophages and returns to the
the antigens to cluster may be important in immune circulation as a microspherocyte. Spherocytes are less
red cell destruction as it may alter the presentation of easily deformed than normal discoid red cells and
membrane-bound IgG to the macrophage (e.g. there tend to be trapped in the spleen, thus shortening their
is a greater likelihood of IgG forming doublets). life span.13,15,18 In addition to Fc receptor-mediated
phagocytosis it is now thought that antibody-depen-
dent cell-mediated cytotoxicity (ADCC) may also
Mechanism of extravascular haemolysis
contribute to cell damage during the phase of close
The interaction of macrophages with RBCs coated contact with splenic macrophages.13,15,18,4144
with IgG occurs through receptors specific for the Fc
portion of the IgG (especially IgG1 and IgG3) dou-
blet.1315,42 The subclass of the IgG antibodies is also CLINICAL FEATURES
important for binding to macrophage Fc receptors
(IgG3 > IgG1). The pathogenicity of red cell anti- The clinical manifestations of HDN range from pal-
bodies depends on additional factors: lor in the mildly affected infant to intense jaundice,
widespread oedema, and signs of neurological
1. The amount of the antibody involvement in the severely affected fetus a
2. Avidity for the erythrocyte antigen syndrome called hydrops fetalis.16,45
3. Antigen distribution and density The anatomical findings in erythroblastosis fetalis
3. The optimum temperature for binding, i.e. 37C vary with the severity of the haemolytic process.
4. The degree of complement binding (not in the Infants may be stillborn, die within the first few days,
case of Rh) or recover completely. In its mildest form the anaemia
may be only slight and the child may survive without
In the case of HDN, additional factors are the further complication. Plasma protein levels sometimes
efficiency of placental transfer of antibody, the func- drop as low as 2025 g/l because of reduced hepatic
tional maturity of the fetal spleen, and the presence of synthesis. Hydrothorax can compromise neo-natal
HLA-related blocking antibodies. Each of these fac- respiratory status after birth.14,16,45
tors probably contributes independently and cumula-
tively to erythrocyte destruction.
Pathogenesis of HDN
Immune haemolysis in vivo begins with opsonisa-
tion of red cells by antibodies. Opsonised red blood The process of HDN is initiated in utero where fetal
cells are recognised and cleared from the circulation RBCs are coated with maternal antibody and are
by macrophages primarily in the spleen, and to a removed from the fetal circulation by its MPS system
lesser extent, the liver. The splenic environment is and destroyed. The anaemia resulting from the RBC
especially conducive to immune clearance. A red destruction is associated with reduction in the oxygen
blood cell lightly coated with IgG (and therefore inca- carrying capacity of the blood which causes the fetal
pable of activating the complement cascade) is prefer- haemopoietic tissue to produce more RBCs and to
entially removed in the sluggish circulation of the release them prematurely. This results in an increased
spleen. IgG-coated red cells are efficiently trapped number of reticulocytes and nucleated red blood cells.
from the haemo-concentrated circulation within the In the gravely ill infant with hydrops fetalis there is
spleen. The relatively low plasma concentration in intense jaundice, widespread oedema, signs of neuro-
splenic sinusoids tends to alleviate competition logical involvement, fatty degeneration, hemosiderin
between plasma IgG and IgG-coated RBCs for Fc deposition and engorgement of hepatic canaliculi.
RhD haemolytic disease 51

Cardiac enlargement, pulmonary haemorrhage and who did not receive an IUT.48 However, neonates who
pleural and pericardial effusions may occur in addi- receive a direct IVT in particular shortly before
tion to massive ascites and subcutaneous oedema delivery, may present with absent reticulocytes.
which results in severe dystocia.45 Historically, before Furthermore, neonates who receive exchange transfu-
the disease was well understood it was identified by its sion can develop reticulocytopenia,49 which is charac-
more obvious symptoms and due to the increase in the teristic of hyporegenerative anaemia observed in
number of nucleated RBCs it was frequently called infants with the later anaemia of haemolytic dis-
erythroblastosis fetalis and cases exhibiting severe ease.5052 In the presence of a high concentration of
jaundice (as a result of increased bilirubin) icterus nucleated RBCs, the automated leukocyte count may
gravis neonatorum. be falsely elevated and require correction by a factor
Compensatory mechanisms exist within affected proportionate to the fraction of nucleated red blood
fetuses, e.g. anaemia and hyperbilirubinaemia may cells.53 Spherocytes are more commonly seen in cases
not occur because the marrow hyperactivity compen- of ABO incompatibility and in certain RBC
sates for the red cell destruction and the hepatic membrane disorders.54
excretor system (including feto-maternal transfer of Accelerated red cell destruction leads to various
bilirubin) may be capable of handling the increased degrees of haemolysis and fetal anaemia. Prolonged
load of bilirubin. When the compensatory capacity of haemolysis results in erythroid hyperplasia of the
the marrow is exceeded, extramedullary haemopoiesis bone marrow, extramedullary haemopoiesis in the
in the liver and spleen may suffice to maintain normal fetal spleen and liver, and hydrops which is charac-
levels of red cells in the blood. Reduced synthesis of terised by various degrees of hepatomegaly,
albumin by the liver leads to hypoalbuminaemia, a fall splenomegaly and placental oedema.55,56 This
in plasma oncotic pressure, and the formation of haemopoietic activity is sufficiently striking to
hydrops.16 When the haemolytic reaction is severe and account for increased numbers of reticulocytes,
the serum bilirubin level exceeds 20 mg/dl (approxi- normoblasts and erythroblasts in the circulating
mately 350 mol/L) the anaemia is associated with blood.48 Evidence of subcutaneous and visceral
jaundice in the newborn. This unconjugated bilirubin oedema is present in the hydrops syndrome, along
is water insoluble and has an affinity for lipids and in with fluid in the peritoneal, pleural and pericardial
the infant with a poorly developed bloodbrain cavities.13,45,57
barrier the bilirubin may bind to the lipids in the brain
and cause severe damage. The severity of all these
Jaundice
changes varies quite considerably in each affected
infant.13,18,45 The most serious threat in erythroblastosis is central
nervous system damage known as kernicterus. In
Haemolytic findings in Rh haemolytic disease of the jaundiced infants unconjugated bilirubin appears to
newborn be particularly toxic to the brain tissue. The brain is
enlarged and oedematous, and when sectioned is
Haemoglobin (Hb) found to have a bright yellow pigmentation (ker-
Cord blood Hb is often abnormally low, but does not nicterus), particularly in the basal ganglia, thalamus,
always correlate with severity. Mollison46 reported cerebellum, cerebral grey matter, and spinal cord.58
that approximately half of the infants affected with This pigmentation is transient and fades within 24
HDN had a Hb exceeding 145 gm/L. In 16 of 141 hours despite prompt fixation. It is therefore neces-
cases the Hb was > 175 g/L; on the other hand, in six sary to section the brain immediately in suspected
cases it was < g/L. cases in order to establish the diagnosis. It is worth
noting that such brain pigmentation is not limited
exclusively to erythroblastosis fetalis, as it has also
Erythrocytes
been described in milder form in infants with severe
The erythrocyte count of cord blood reflects the Hb physiological jaundice of the newborn.45
content, i.e. in infants without anaemia the counts are Bile pigmentation of the brain is of considerable
as high as 5.5 1012/L and at the end of the scale, theoretical interest, because in the adult a
counts < 2.0 1012/L are seen in infants born mori- bloodbrain barrier blocks the passage of bilirubin
bund. There is marked macrocytosis and the MCV are into the spinal fluid and substance of the brain, even
greater in mature normal newborns.47 when there is severe jaundice. The precise bilirubin
Stained blood films showed anisocytosis with many level that will induce kernicterus is not predictable, but
macrocytes and a small degree of poikilocytosis. The neural damage rarely occurs if the serum bilirubin
reticulocyte count can be as high as 3040% in infants concentration is below 20 mg/dl.45,58 However,
52 Blood Reviews

premature infants are at risk from lower serum albu- to 2.5 standard deviations above the expected Hb for
min levels. The mechanism by which free (unconju- the gestational age (using nomograms) and to
gated) bilirubin enters the brain in the newborn is not lengthen the time between transfusions to 23 weeks.
clearly understood, but the level of serum albumin The effect of the first transfusion is shortest because
(which binds bilirubin), pH of the blood and hypoxia of the continued presence of antibody coated fetal red
are all important factors in allowing passage of cells; thereafter fetal haemopoiesis is suppressed and
bilirubin into the brain.16,53,5860 transfused red cells fall by 1% per day. The last fetal
transfusion is given at about 35 weeks with delivery at
37/38 weeks gestation, in preference to early delivery
TREATMENT and exchange transfusion because the fetus tolerates
larger transfusion volumes than the neonate due to
The availability of tests in vitro to monitor the sever- the large capacity of the placenta. With advances in
ity of the haemolytic reaction has opened a variety of fetal medicine, introduction of direct intravascular
avenues for the treatment of haemolytic disease of the transfusion (IVT) and improved neonatalogy in
newborn. When analysis of amniotic fluid discloses specialised centres, survival of affected fetuses with
critical levels of bilirubin, premature delivery may be HDN is now greater than 90%.13
induced if the fetus is judged to be viable with regard
to size and maturity. Earlier, fetal transfusion may be
Plasma exchange (PE)
attempted. Post-natally, a variety of supportive mea-
sures may be employed including phototherapy Removal of as much as 45 litres anti-D containing
(visual light oxidises toxic unconjugated bilirubin to plasma, and replacement with albumin/crystalloid
harmless, readily excreted water-soluble dipyrroles) solutions, is possible using automated apheresis
and, in severe cases, total exchange transfusion of the devices. However, the efficacy of this procedure in the
infant.19,20 absence of immunosuppression to prevent rebound
synthesis of IgG is debatable, and needs to be
repeated two or three times a week to maintain
Intrauterine fetal transfusion
reduced levels of anti-D. Results of PE have been vari-
The first intravascular transfusions (IVT) were per- able,1618 and this procedure is not to be undertaken
formed fetoscopically in the early 1980s. The method lightly as the mother is subjected to an arduous proce-
used most widely is ultrasound-guided blood sam- dure. It may be indicated if there is a previous history
pling from the umbilical vein at the placental cord of fetal loss or a severely affected infant, a strong pos-
insertion.13,1720 Intravascular transfusion has become sibility that the father is homozygous for RhD, and to
the mainstay of management of very severe alloim- maintain pregnancy until cordocentesis or IVT sam-
munization in pregnancy, although intraperitoneal pling and transfusion can be carried out safely. If
transfusion still has a place in early gestation between decided upon, PE should be started early in preg-
14 and 18 weeks or, for example, when the cord root is nancy, and potentially immunising factors (e.g.
inaccessible or in order to top-up an incomplete IVT. amniocentesis) should be avoided until 2628
Despite considerable experience with IVT, it has been weeks.1921
shown that there is still a need to continue familiarity
with the intraperitoneal approach.13,1921 Intravascular
Intravenous immunoglobulin (IV IgG)
transfusions carry significantly increased risks for
both mother and fetus when compared to cordocente- There have been recent reports of the benefits of high
sis alone, and this technique should be offered only dose intravenous immunoglobulin (HDIV IgG)
within a fetal medicine unit. It has been recommended administration in severely alloimmunized pregnant
that about 2030 cordocenteses and 1015 transfu- women.13,18,61 Doses of 2 g/kg maternal body weight
sions need to be performed annually to maintain com- every two weeks can reduce circulating allo-
petence.13,19 antibody levels by up to 50%, and this is thought to be
With accurate determination of fetal genotype by mainly due to the negative feedback produced by total
PCR on amniocytes (see below) there is a tendency to circulating maternal IgG levels of 2530 g/l.13,61,62
delay the first IUT until after 20 weeks, or signs of Further benefits of IV IgG therapy may be
hydrops are developing. If severe hydrops is suspected interference with trans-placental transfer of maternal
very early, the preference is for intraperitoneal trans- antibodies by trophoblastic Fc receptor saturation,61,62
fusion at 1415 weeks followed by FBS/Intravascular and (similarly) reduction of IgG-coated fetal red cell
transfusion. With greater understanding of fetal haemolysis by fetal mononuclear phagocytic Fc recep-
responses to transfusion, there is a trend to transfuse tor saturation. This is in agreement with the outcome
RhD haemolytic disease 53

of a controlled study63 which showed that the


frequency of transfusional therapy was reduced
when HDIV IgG was combined with conventional
phototherapy.
Combined PE/IV IgG therapy may be considered
in the same circumstances as when intensive plasma
exchange would be considered, and only from 1012
weeks gestation. 13,18,53

BLOOD GROUP SEROLOGY MONITORING Sample required for antibody screen


Sample required when the antibody(ies) are associated with HDN other than D, c, Kell, e.g.
Duffy (Fy), Kidd (Jk).
The mothers first prenatal visit booking, includes a For ABO, Rhd group; weak D test if apparently negative;29 antibody screen
Confirmation of blood group/antibody; estimation of FMH
history of pregnancies and blood transfusions, ABO For ABO, Rhd group; weak D test if apparently negative; direct antiglobulin test, antibody
elution and identification if positive; and red cell phenotyping as required
and RhD grouping, and antibody screening. If the * All patients except those with antiD, antic or Kellrelated antibodies.
** Antibody associated with severe HDN e.g. D, c, and Kell related antibodies.
RhD group is negative, a test for weak D should be
Delivery in Specialized Hospital is advised when there is a possibility or risk of transfusion
performed. Conventional serology is directed towards problem and/or HDN.

the identification of IgG antibodies which react at Fig. 2 Suggested frequency of ante-natal testing
37C by using indirect antiglobulin test (IAT) with
untreated red cells. Further testing of all women with-
out antibodies is advised at least once between 28 and fortnightly from 3036 weeks and weekly from 36
32 weeks gestation18,64,65 (prior to administration of weeks onwards (Fig. 2).65,69,70 Where quantification of
any antepartum anti-D, immunoglobulin), and at the anti-D is not routine, laboratories establish critical
time of delivery, which should be accompanied by a titres below which hydrops fetalis is not anticipated,
cord blood sample for ABO/RhD grouping and direct usually below 1/161/32, with close monitoring if
antiglobulin test, and an uncoagulated maternal > 1/8.71
sample for estimation of FMH6668 to decide on Serological assays measure only the capacity of
the administration/dose of prophylactic anti-D antibodies for agglutination whereas in vivo destruc-
immunoglobulin (Fig. 2). Some authorities recom- tion involves interaction of antibody coated red cells
mend screening twice during pregnancy, at 2832 with Fc receptors of the mononuclear phagocyte
and 3436 weeks gestation.18,65 Although enzyme- system. Considerable interest has therefore been
enhanced techniques may identify antibody at a lower shown in developing bioassays as predictors of HDN,
level compared to the IAT, the vast majority of these including monocyte monolayer assays, which measure
additional antibodies are of no clinical significance as adherence/phagocytosis, monocyte chemilumines-
regards HDN.69 cence (CL) which measures the oxidative respiratory
Where alloantibodies are detected which are clini- burst associated with adherence/phagocytosis and
cally significant, i.e. known to be associated with antibody dependent cytotoxicity (ADCC) assays
HDN, IAT titre is performed, and followed up by which measure extracellular haemolysis.14,15,18,42,72,73
assessment of the level of antibody and the monitor-
ing of any change during the course of pregnancy. In
the case of anti-D, Auto Analyser quantification is MONITORING AND EVALUATION OF
also done and reported in iu/ml (5 iu = 1 g) in com- ALLOIMMUNIZED WOMEN
parison with the national reference preparation.18
Anti-D levels
Samples should be stored frozen and tested in parallel
with the current sample using the same techniques.70 For mild disease (maternal anti-D levels <2.5 iu/ml,
The partner is also phenotyped for the relevant anti- with good obstetric history or IAT <1/8), monthly
gen(s). The absolute level of the antibody is not as anti-D quantification and Ultrasonography should be
important as the trend, with a rising level requiring adequate. When the anti-D concentration remains
more frequent monitoring especially with a homozy- <5 iu/ml (<1 g/ml), fewer than 5% of affected fetuses
gous partner, and if there is a history of previous will require neonatal exchange transfusion.
HDN.18,19,65,70 The likelihood of significant HDN due Amniocentesis may be the preferred method of inves-
to anti-D below 4 iu/ml is very remote, whereas above tigation when anti-D levels are from 4 to 15 iu/ml
15 iu/ml all cases should be treated as at risk of severe (IAT > 1/32),71 as moderate fetal anaemia may occur.
HDN until proven otherwise by the clinical investiga- Fetal blood sampling (FBS) may be performed early
tions i.e. testing monthly from booking to 30 weeks, (at 18 weeks) when anti-D levels are >20 iu/ml,13,18,19
54 Blood Reviews

or at a titre established locally as being critical. These


levels are only guidelines, and do not precisely predict 1.00

the degree of fetal anaemia. Immediate Delivery

Intrauterine
Transfusion Upper Zone
0.50
(severe)

Ultrasonography
0.20
Ultrasonography is useful to monitor Rh-alloimmu-
nized pregnancies, and in experienced hands early

OD450(nm)
0.10
hydrops can be reliably detected.13,18 Fetal ascites or
hydrops can be observed with placental thickening,
loss of architecture and increased homogeneity and 0.05

hydramnios. The most common progression is from Mid Zone


(moderate)
hydramnios to placental thickening, to enlargement of
0.02
the fetal liver, to hydrops. However, placental thick-
ness, extra and intrahepatic umbilical vein diameters, Lower Zone
(mild)
0.01
abdominal circumference and head/abdominal cir- 24 28 32 36 40
cumference ratios in one study were not found to be Weeks of Gestation
reliable markers of severe anaemia.13,19 Further work
76
needs to be performed to assess the value of ultra- Fig. 3 Liley Graph.
sonic markers of early fetal ascites, such as double
outlining of small bowel loops and abdominal fluid
rim. However, a recent prospective study suggests that When readings are approaching the upper zone, the
pregnancies with a mild or no history of fetal anaemia fetus requires immediate attention as severe disease is
may be successfully monitored without invasive tech- predicted. Accurate prediction of the severity of
niques by a combination of serial anti-D quantifica- HDN by using amniotic fluid requires serial OD 450
tion and Doppler monitoring of umbilical vein measurements, and amniocentesis is also more accu-
maximal flow velocity (UVVmax); liver length and rate in the third trimester than in the second trimester.
spleen perimeter measurements were not as helpful.74 Final values falling in the upper and lower zones have
A similar multicentre prospective study showed that an accuracy of prediction of 95%. On the other hand,
Doppler estimation of peak systemic blood velocity final readings remaining in the mid zone have accu-
in the middle cerebral artery accurately predicted racy of prediction of approximately 90%.13,16,18 It
moderate or severe anaemia in non-hydropic fetuses.75 should be borne in mind that amniocentesis is not
without risk, with up to 1% fetal mortality directly
associated with the procedure, and boosting of anti-D
Amniocentesis
levels following leak of fetal cells into the maternal
Amniocentesis allows spectrophotometric measure- circulation.
ment of the deviation in optical density at 450 nm
( OD 450) due to bilirubin in amniotic fluid, which
Fetal blood sampling (FBS)
reflects fetal red blood cell haemolysis. It does not,
however, provide information on fetal erythropoiesis, The current approach of using ultrasound needle
and is unreliable prior to 27 weeks gestation.9,13 guidance was first introduced for prenatal diagnosis in
Different degrees of change are measured/interpreted the early 1980s.13,16,20 The most accurate method of
differently, because the normal level of bilirubin assessing fetal anaemia is to measure the haematocrit
varies with gestational age.16,18,22 Liley (1961)76 was the of fetal blood directly by cordocentesis, and is ideally
first to develop a method to measure this deviation performed when the fetus is estimated to be
(Liley Graph- Fig. 3); various modifications have been significantly anaemic, but before decompensation and
made in an attempt to extrapolate to the first hydrops has developed. Also, the Rh group of the
trimester, e.g. Queenans modification,77 or Whitfield fetus may be determined with accuracy, particularly in
action line78 but none are entirely satisfactory in the cases where there is a poor obstetric history, the father
first trimester. If all the OD 450 values remain in the is heterozygous for RhD, and high levels of anti-D
lower zone this should indicate either no disease or may be sustained from a previous pregnancy. It is not
only a mildly affected fetus, but 10% may require technically safe to undertake FBS earlier than 1820
exchange transfusion. Values in the mid zone indicate weeks gestation and procedural losses have been
a moderately affected fetus which is a candidate for an estimated at 12% in non-hydropic cases. Procedural
early delivery and possible exchange transfusion. mortality in hydropic cases is approximately 15%
RhD haemolytic disease 55

before 20 weeks, and 5% thereafter. Complicat- of detecting all common variants have been
ions include fetal bradycardia, haematoma formation, reported.8890 It is prudent practice to test maternal
fetal haemorrhage and boosting of anti-D levels, pre- samples in parallel, and to include paternal samples
term rupture of membranes and premature labour, where D variants, or non-caucasians are being
and infection. The technique of fetal blood sampling genotyped.
requires a lengthy learning curve and should be
performed only in specialist referral centres with
Summary of evaluation
facilities to perform immediate intravascular fetal
transfusion if necessary. There is no agreement yet on Prediction of the outcome of Rh HDN is currently
a critical haematocrit level which indicates the need based on a number of factors (Box 1).
for transfusion. Transfusion has been advocated when
the fetal haematocrit is below 25% before 26 weeks
gestation, and below 30% after that.13,20 Box 1

1. The outcome of previous pregnancies gives some


Polymerase Chain Reaction (PCR) RhD genotyping indication of expected severity, especially when the father
is homozygous for RhD.
The single most important advance in recent years in
2. Serial estimation of maternal anti-D during pregnancy
the antenatal management of alloimmunised women (single antibody titres are not always helpful). RhD
has been the introduction of molecular methods of positive babies will be affected by HDN if the maternal
blood grouping from genomic DNA. Techniques such anti-D level is greater than 4 iu/ml, (as follows) although
as PCR28,7984 and sequence-specific oligonucleotide exceptions can occur:
primers (SSP) to detect fetal RHD and RHCE, KEL, anti-D < 4 iu/ml HDN unlikely
FY and JK genes82,83 are in routine use and it is now
possible to determine the fetal blood group genotype anti-D 415 iu/ml moderate risk of HDN

using DNA extracted from amniotic fluid cells. Such anti-D > 15 iu/ml high risk of hydrops fetalis
tests are of value in cases with history of severe HDN 3. Serial ultrasound measurements allows detection of early
where the partner is heterozygous for the correspond- ascites, and the planning of invasive techniques, such as
ing antigen (or unknown) and allow more accu- FBS and IUT.
rate, early prediction of the relevant blood group 4. Diagnostic amniocentesis may be undertaken in at-risk
genotype of the fetus. Diagnostic amniocentesis to cases for early PCR RhD genotyping.
obtain cells of fetal origin (as for detecting fetal 5. Serial amniocentesis followed by optical density
genetic disorders) can be done as early as 1214 measurement ( OD 450 mm) of amniotic fluid for
weeks. This allows early and intensive monitoring of bilirubin content gives an indication of the degree of
fetuses at risk of death in utero, with early interven- haemolysis, and impending hydrops.
tion when necessary. Selective termination of an 5. Fetal blood sampling allows direct assessment of fetal
RhD-positive pregnancy may be considered. anaemia and the Rh group of the fetus.
Conversely, when the fetus is RhD negative, further 6. Functional assays, such as monocyte binding, CL and
invasive techniques may be avoided. ADCC have been used recently in the prediction of the
There are certain caveats to be considered. In outcome of RhD HDN by measuring the potential lytic
principle, any test which distinguishes between the activity of the maternal anti-D.
presence or absence of the RHD gene is suitable, but
primers for the PCR reactions need to be chosen with
care to minimise false positives and false negatives, Combined estimates based on previous history, anti-
due to gene conversions between RHD and RHCE, D levels and amniocentesis can enable accurate
point mutations and dysfunctional RhD genes.30,85 prediction of the outcome in approximately 95% of
Recent studies have shown that some ethnic groups, cases.1,13 Nevertheless, unexpectedly mild (or severe)
such as South African blacks [and their descen- cases can still occur.1,17,18 When a high level of anti-D
dants],26,30 Japanese,86 and Chinese have a high justified amniocentesis but the fetus is shown to be
incidence of partially intact but functionally inert only mildly affected, there is a risk of further anti-D
RHD genes, and will type as RHD positive with the production as a result of accidental fetal-maternal
usual primers devised for Caucasians. Currently, no haemorrhage. Conversely, a low level of anti-D might
single method is sufficient for diagnostic use, and two allay suspicion until it becomes apparent that the fetus
or more PCR reactions for different exons of the is already severely affected. It is in these situations that
RHD gene are required, with due regard for the functional assays such as ADCC or CL may provide
ethnicity of the samples.8587 Multiplex assays capable additional useful information.14,42,72,73
56 Blood Reviews

PREVENTION seen by the dendritic antigen-presenting cells


(APC).13,18,22
Prophylactic anti-D immunoglobulin and mechanisms Antigen blocking cannot explain AMIS because:
of action (1) less than 20% of available antigen is bound by
RhD-immune globulin; and (2) in murine models
Development of RhD alloimmunization prophylaxis of immunization by sheep erythrocytes, F(ab)2
in the 1960s was based on the well-recognised phe- fragments, which bind avidly to sheep red blood cells,
nomenon of antibody-mediated immune suppression do not suppress the murine immune response, whereas
(AMIS), in which passively administered specific anti- whole antibody with an intact Fc region does.14,18,22
body was known to prevent active immunization by However, recent evidence has challenged this
the corresponding antigen. Even today the mecha- hypothesis.92
nism of the AMIS response is not completely under- Central inhibition may represent a plausible expla-
stood.1,13,22 Three general theories have been nation for AMIS.1,22 Fetal red cells coated with anti-D
proposed: (1) antigen deviation or diversion; (2) com- are filtered out of the maternal circulation by the
petitive inhibition by antigen blocking; and (3) central spleen and lymph nodes, where the increase in local
inhibition, i.e. negative modulation of the primary concentration of complexes of anti-D bound to RhD
immune response. antigen may suppress the primary immune response
There is some evidence for the antigen by interrupting helper T cell-mediated clonal expan-
deviation/diversion theory in the observation that sion of RhD-specific B cells. Furthermore, the Fc
RhD immunization is less common in offspring when region appears to be required for AMIS; neither F(ab)
the father is ABO incompatible with the mother.1,13,22 nor F(ab)2 fragments are effective. This model is also
It has been estimated that (in Whites) groups A and B consistent with the observation that neither AMIS
incompatibility between infant and mother gave 90% nor prophylactic anti-D inhibits secondary immune
and 55% protection respectively against RhD sensiti- responses.
zation. If the fetal red cells which carry the RhD anti- In animal models,13,22 the antibody and the effector
gen are ABO compatible with the mother they T cells remaining in an individual which has already
circulate freely (intact) until removed by the mononu- been immunised also prevent activation of naive B
clear phagocytes (MPS) of the spleen, which is also a and T cells by the same antigen. This can be shown by
major site of antibody production. Concurrent ABO passively transferring antibody or effector T cells to
incompatibility can protect the mother against immu- naive recipients; when the recipient is then immu-
nization, presumably because leaked fetal red cells are nised, naive lymphocytes do not respond to the origi-
promptly coated by circulating isohaemagglutinins nal antigen while responses to other antigens are
(IgM) and (probably) complement, and then removed unaffected. While the precise mechanism of suppres-
from the circulation by the MPS, mainly in the liver sion is unclear, it is known that cross-linking of the
(Fig. 1). The liver is less immunoresponsive than the antigen receptor on B cells to Fc receptor II (FcRII)
spleen and therefore is less likely to stimulate antibody on the B-cell surface inhibits the activation of naive B
production. Experimental evidence of a non-specific cells, which may explain this kind of suppression.13,22,29
suppressive effect of passive IgG antibody to human For some reason, memory B-cell responses are not
RBC was obtained when red cells possessing two anti- inhibited by antibody to the antigen, so for this treat-
genic determinants (D+ve and K+ve) were injected ment to be useful the RhD negative mothers at risk
into D-ve, K-ve persons together with anti-K18,91 This must be identified before a response has occurred.
antibody suppressed the response to both K and D. This also shows that memory B cells can be activated
The mechanism of destruction of red cells due to anti- to produce antibody even when they are exposed to
K is similar to anti-D, i.e. initiates red cell destruction pre-existing antibody, allowing secondary antibody
predominately in the spleen. On the other hand, responses to occur in immunised individuals.
studies of Cr-labelled RhD positive red cells infused Could RhD haemolytic disease of the newborn be
into RhD negative volunteers18 have shown that RhD eliminated by administration of prophylactic anti-D
immunoglobulin increases clearance of RhD positive immunoglobulin during pregnancy? It has been
cells; the antigen itself is not destroyed. Rather, the shown in clinical trials that antenatal administration
intact antibody-coated-cells (deformed/tagged) are of anti-D in the third trimester can suppress immuni-
removed to the spleen or lymph nodes, where an sation to approximately 0.2%.1,13 It is possible that as
immune response would normally be initiated. It is the D antigen is processed, free anti-D molecules
possible that the splenic macrophages, which have (from the Rh immune globulin) compete successfully
high FcR expression, efficiently remove the anti body for the processed D antigen and prevent it from bind-
coated cells, and destroy the RhD antigen before it is ing to the D antigen receptor of the IgM monomers of
RhD haemolytic disease 57

appropriate B cells. While it is appreciated that the in Ante-partum anti-D immunoprophylaxis


vivo life of the passively administered anti-D
The administration of anti-D immunoglobulin to
molecules will be considerably shorter than that
RhD negative (previously non-RhD-sensitised)
expected of ABO compatible RhD positive fetal cells,
women within 72 hours of each delivery of an RhD
the presence of anti-D may result in the faster (but not
positive fetus prevents Rh immunization in the vast
immediate) sequestration and removal of the fetal
majority of cases.1,32,9397 Although immunoprophy-
cells. It is also possible that in the presence of free
laxis is highly successful in preventing RhD sensitiza-
anti-D molecules, B cells with receptors for D in their
tion, it is not yet perfect.9597 Approximately 2% of
IgM monomers, do not bind processed D antigen
women at risk develop anti-D in the last trimester or
because they interpret the presence of anti-D as indi-
within 72 hours after labour, and thus fail to benefit
cating lack of necessity to make anti-D. Perhaps most
from Rh immunoprophylaxis.19 Some of these cases
likely, the presence of free anti-D may prevent or
may result from unrecognised previous RhD positive
block the signal from the appropriate T-helper cells
abortions, but as many as 40% of all new cases are
that would normally initiate production of anti-D by
considered to be due to significant TPH during the
B cells.1,13,18
early part of the third trimester.1,18,32,65,96,97 In principle
it should be possible to prevent sensitization in such
Dose and administration of anti-D Ig28 women, and thereby increase the success of Rh
immunoprophylaxis if anti-D immunoglobulin is
The clinical situations in which anti-D immuno-
administered ante-partum in the third trimester as
globulin is recommended are detailed in Tables 3 & 4.
well as post-partum.1,12,18,32,37,94107 In North America,
The general principle is that 100 iu (20 g) of anti-D
RhD negative women receive 1500 iu (300 g) of anti-
will provide protection against 1 ml RhD positive
D between 28 and 32 weeks gestation in all pregnan-
fetal red cells (i.e. not whole blood)33,65 when given
cies.1,13,29 If the infant is RhD positive a similar dose is
intramuscularly, and this may be increased to 125 iu
given after delivery. In most European countries, a
(25 g) per ml fetal red cells to give a margin of
smaller dose (10001250 iu) is given in divided doses
safety.11 Different vial sizes are conventionally used in
at 28 and 34 weeks. In the UK,101 (Table 6) a con-
different countries for historical reasons. A dose of
trolled trial of 500 iu at 28 and 34 weeks reduced
300 g (1500 iu) is commonly used in North America,
alloimmunization to 0.2%, as efficacious as the larger
100 g (500 iu) in the UK, and 200250 g
doses used elsewhere. Theoretically, the divided doses
(10001250 iu) in Europe and elsewhere. Prescribing
should be more effective in practice than a single large
details are given in the manufacturers product leaflets,
dose since a higher level of maternal anti-D will be
but the principles are as above. In the UK the recom-
maintained at a time when larger FMH are more
mended dose of anti-D in the event of any potential
common.
TPH depends on the period of gestation (i.e. 250 iu
Antenatal prophylaxis is not universally applied, due
for an event which may precipitate a FMH at less than
to a variety of factors including the incidence of RhD
20 weeks, and 500 iu if the gestation is more than 20
negative women, the birth rate, the availability and
weeks) the larger standard dose will suffice provided
cost of anti-D, and the failure rate with standard post-
that the FMH is less than 4 ml. A Kleihauer acid elu-
natal prophylaxis;108 a cost-benefit analysis would be
tion test, or equivalent,6668 to demonstrate fetal cells
required for each country to determine the utility of
should be performed on the maternal blood (Table 3),
prophylaxis.109,110
and if the FMH is more than 4 ml a larger dose of
anti-D immunoglobulin given (i.e. 125 iu/1 ml of fetal
red cells).32 The 1500 iu dose will protect against 15 ml SAFETY OF ANTI-D IMMUNOGLOBULIN
of fetal RhD positive red cells, and this is the thresh-
old TPH for further anti-D to be given with the larger Because anti-D immunoglobulin is manufactured
dose.1,16,29 Once instituted it is important to repeat from large numbers of plasma donations from indi-
therapy at 6-week intervals in the event of intermit- viduals exposed to human red cells, considerable
tent bleeding as very low levels of passively-adminis- effort is expended in validating and accrediting both
tered anti-D may enhance antibody formation in the the red cell and the plasma donors.34 The potential
presence of an antigenic stimulus.18 If an RhD-posi- implications of transfusion transmitted infection are
tive fetus is delivered, a Kleihauer, or alternative test considerable, since two individuals are at risk the
should be performed to assess any large FMH, and mother, and her unborn child. Fortunately, the safety
then anti-D should be given in the usual way, even if record of intramuscular injections of immune globu-
residual activity from antenatal prophylactic anti-D lins produced by cold-ethanol Cohn fractionation is
can be demonstrated in the maternal serum.28,85,86 excellent since the process itself reduces viral load
58 Blood Reviews

Table 6 Chronology of antepartum prophylactic anti-D trials

1978a99 Bowman JM, Pollock JM. Rh isoimmunisation during pregnancy: antenatal prophylaxis
1978b100 Bowman JM, Pollock JM. Antenatal prophylaxis of Rh isoimmunisation: 28 week gestation service program
1983101 Tovey LAD et al. The Yorkshire antenatal anti-D immunoglobulin trial in primigravidae
1984102 Herman M, Kjellman H, Ljunggren C. Antenatal prophylaxis of Rh immunisation with 250 mcg of anti-D immunoglobulin
1987103 Bowman JM, Pollock JM. Failures of intravenous Rh immune globulin prophylaxis: an analysis of reasons for such failures
1987104 Huchet J et al. The antepartum use of anti-D immunoglobulin in rhesus negative women
1989105 Trolle B. Prenatal Rh-immune globulin prophylaxis with 300 mcg immune globulin anti-D in the 28 week of pregnancy
1989106 Thornton JG et al. Efficacy and long term effects of antenatal prophylaxis with anti-D immunoglobulin
1995107 Lee D, Rawlinson V. Multicentre trial of antepartum low dose anti-D immunoglobulin

(even prior to screening) by several logs and this prod- Anti-D produced by biotechnology may be made
uct is safe from viral transmission (reviewed in ref. available in the future by means of monoclonal anti-
111). The few recorded incidents of transmission of body production.116 Material derived from
HCV by anti-D immunoglobulin relate to products EpsteinBarr virus-transformed lymphocytes is being
manufactured by anion exchange chromatography evaluated in volunteer studies, and if successful, new
when not subject to virus-inactivation.112 preparations of anti-D will become available to be
The position with respect to vCJD is different, tested in large clinical trials.
since there are no screening methods, the extent of the Finally, prospects are emerging for the manufac-
infection in the donor population is unknown, and the ture of synthetic peptide vaccines to regulate the
transmissibility by blood products is not known immune response to the RhD antigen with the identi-
(reviewed in ref. 113). For these reasons, the manufac- fication of T-cell epitopes on the Rh polypeptide38
ture of immune globulins from UK plasma has In principle, active suppression of both primary
stopped, and material is imported from countries not and secondary responses to RhD could be achieved in
known to have clusters of vCJD infection. Similarly, all RhD negative females prior to puberty leading
some countries (including the USA and Canada) to eventual eradication of severe RhD haemolytic
do not accept blood from donors who have been disease.
resident in the UK for a cumulative period of 6
months or more between 1980 and 1996, to minimise REFERENCES
the theoretical risk of transmission of vCJD.
1. Bowman JM. The prevention of Rh immunization. Trans
Med Rev 1988; 2: 129150.
FUTURE PROSPECTS AND RESEARCH 2. Diamond LK, Blackfan KD, Baty JM. Erythroblastosis
fetalis and its association with universal edema of the fetus,
Adequate and timely administration of anti-D icterus gravis neonatorum and anemia of the newborn.
immunoprophylaxis must remain a priority, since J Pediatr 1932; 1: 269309.
3. Hawksley JC, Lightwood R. A contribution to the study of
administrative failures are still a significant cause of erythroblastosis: icterus gravis neonatorum. Quart J Med
Rh immunisation. More data from continued research 1934; 3: 155209.
may provide firmer criteria for the timing of invasive 4. Gilmour JR. Erythroblastosis fetalis. Arch Dis Child 1944;
19: 125.
transplacental procedures, and to provide better 5. Darrow RR. Icterus gravis (erythroblastosis neonatorum,
non-invasive alternatives such as Doppler ultrasound, examination of etiologic considerations). Arch Pathol 1938;
and other biophysical measurements of the fetal 25: 378417.
6. Levine P, Stetson RE. An unusual case of intra-group
condition.74,75 agglutination. J Amer Med Ass 1939; 113: 126127.
Recently, the introduction of molecular DNA tech- 7. Landsteiner K, Wiener AS. An agglutinable factor in human
nology has permitted determination of fetal blood blood recognized by immune sera for rhesus blood. Proc Soc
Exp Biol Med 1940; 43: 223.
group status from first trimester chorionic villus sam- 8. Chown B. Anemia from bleeding of the fetus into the
pling, or from amniotic fluid cells.28,80,81 These tests are mothers circulation. Lancet 1954; 1: 12131215.
invasive, with risks of fetal loss and boosting of anti- 9. Combined Study. Prevention of Rh-haemolytic disease:
results of the clinical trial. A combined study from centres in
body levels, and current research is focused on identi- England and Baltimore. BMJ 1966; 2: 904907.
fying fetal genotype from maternal blood samples, 10. Pollack W, Gorman JG, Freda VJ et al. Results of clinical
isolating fetal nucleated red cells, or plasma as a trials of RhoGAM in women. Transfusion 1968; 8: 151.
11. WHO. Prevention of Rh sensitization. Technical Report
source of fetal DNA/RNA.114 Accurate fetal genotyp- Series 1971; 468.
ing on maternal plasma samples115 would allow 12. Proceedings of the Joint Royal College of Physicians of
automation, and the possibility of restricting antena- Edinburgh and the Royal College of Obstetricians and
Gynaecologists Consensus Conference on Anti-D
tal anti-D immunoglobulin to mothers carrying a Prophylaxis. Urbaniak SJ (ed), Br J Obstet Gynaecol 1998;
RhD positive fetus. 105 suppl 18.
RhD haemolytic disease 59

13. Anderson KC, Ness PM. Scientific Basis of Transfusion 38. Urbaniak SJ, Stott LM, Wilson DW et al. The identification
Medicine. Implication for Clinical Practice (Chapters 5,6,19). of peptides which stimulate helper T-cells in vitro.
WB Saunders Company, 2000. Transfusion 1999; 39: supplement 1S.
14. Garratty G. Immunobiology of Transfusion Medicine. 39. Bowman JM, Pollock JM, Penston LE. Fetomaternal
Marcel Dekker 1994. transplacental hemorrhage during pregnancy and after
15. Issitt PD, Anstee DJ. Applied Blood Group Serology. delivery. Vox Sang 1986; 51: 117121.
Montgomery Scientific Publications 1998. 4th edition. 40. Hann IM, Gibson BES, Letsky EA. Fetal and Neonatal
16. Bowman JM. The management of Rh-isoimmunisation. Haematology (Chapters 5 & 7). Bailliere Tindall 1991.
J Obstet Gynaecol 1978; 52: 116. 41. Quinley ED. Immunohaematology: Principles and Practice.
17. Bowman JM. Treatment options for the fetus with Chapter 14. J B Lippincott Company 1993.
alloimmune hemolytic disease. Trans Med Rev 1990; 4: 42. Engelfriet CP, Ouwehand WH. ADCC and Other Cellular
191207. Bioassays for Predicting the Clinical Significance of Red Cell
18. Mollison PL, Engelfriet CP, Contreras M. Blood Transfusion Alloantibodies. In: Contreras M, ed. Blood Transfusion: The
in Clinical Medicine, 10th ed., Chapters 3, 5, 12. Oxford: Impact of New Technologies, Baillieres Clinical
Blackwell Scientific Publications, 1997. Haematology Series, Vol. 3. London: Bailliere, 1990; 32137.
19. Murphy KW, Whitfield CR. Rhesus disease in this decade. 43. Urbaniak SJ. ADCC (K-cell) lysis of human erythrocytes
obstetrics. Contemp. Rev. Obstet. Gynaecol 1994; 6: 617. sensitized with rhesus antibodies II. Investigation into
20. Tannirandorn Y, Rodeck CH. New Approach in the mechanism of lysis. Br J Haematol 1979; 42: 315328.
Treatment of Haemolytic Disease of the Fetus. In: Contreras 44. Urbaniak SJ, Greiss MA. ADCC (K-cell) lysis of human
Marcela, ed. Blood Transfusion: The Impact of New erythrocytes sensitised with rhesus antibodies III.
Technologies. Baillieres Clinical Haematology Series, Vol. 3. Comparison of IgG anti-D agglutinating and lytic (ADCC)
London: Bailliere, 1990; 289320. activity and the role of IgG subclasses. Br J Haematol 1980;
21. White CA, Goplerud CP, Kisker CT et al. Intrauterine fetal 46: 447453.
transfusion, 19651976, with an assessment of the surviving 45. Schofield D, Cotran RS. Diseases of Infancy and Childhood.
children. Am J Obstet Gynecol 1978; 130: 93342. In: Cotran RS Kumar V, Collins T (eds.). Robbins Pathologic
22. Stites DP, Tern AI, Parslow TG. Basic & Clinical Basis of Disease. Chapter 11. W B Saunders Company 1999;
Immunology 8th ed. Chapters 3, 15, 42. A LANGE Medical 6th edition.
Book. 46. Mollison PL. Blood transfusion in clinical medicine, 3rd Ed,
23. Sturgeon P. Hematological observation on anaemia 1961: p614
associated with blood type Rhnull. Blood 1970; 36: 310. 47. Dacie JV. Haemolytic disease of the newborn. In Haemolytic
24. Lauf PK, Joiner CH. Increased potassium transport and Anaemia, 3rd. vol 5, Churchill Livingstone 1999.
ollabain binding in human Rhnull red blood cells. Blood 1976; 48. Naiman JL. Erythroblastosis fetalis. In Oski FA, Naiman JL
48: 457468. (eds): Hematologic problems in the newborn. Philadelphia.
25. Kuypers F, van Linde-Sibenius-Trip M, Roelofsen B et al. WB Saunders, 1982; pp 283346.
Rhnull human erythrocytes have an abnormal member 49. Koenig JM, Christensen RD. Neutropenia and
phospholipid organisation. Biochem J 1984; 221: 931934. thrombocytopenia in infants with Rh hemolytic disease.
26. Singleton BK, Green CA, Avent ND et al. The presence of J Pediatr 1989; 114: 625631.
an RHD pseudogene containing a 37 base pair duplication 50. Koenig JM, Ashton RD, De VG, Christensen RD. Late
and a nonsense muatation in Africans with the RhD-negative hyporegenerative anemia in Rh hemolytic disease. J Pediatr
blood group phenotype. Blood 2000; 95: 1218. 1989; 115: 315318.
27. Scott ML, Voak D, Jones JW et al. A model for RhD the 51. Thorp JA, OConnor T, Callenbach J et al. Hyporegenerative
relationship of 30 serologically defined epitopes to predicted anemia associated with intrauterine transfusion in rhesus
structure. Biotest Bulletin 1997; 5: 459466. hemolytic disease. Am J Obstet Gynecol 1991; 165: 7981.
28. Daniels G, Lomas-Francis C, Wallace M et al. Epitopes of 52. Ohls RK, Wirkus PE, Christensen RD. Recombinant
RhD: Serology and Molecular Genetics. Molecular and erythropoietin as treatment for the late hyporegenerative
Functional Aspects of Blood Groups Antigens. Ed. anemia of Rh hemolytic disease. Pediatrics 1992; 90: 678680.
Silberstein LE. American Association of Blood Banks, 53. Koenig JM. Evaluation and treatment of erythroblastosis
Bethesda, Maryland 1995; 193228. fetalis in the neonate. In Christensen RD (ed.): Hematologic
29. Vengelen-Tyler V, ed. Technical Manual, 13th ed., Chapter 23 problems of the neonate. Philadelphia, WB Saunders, 2000;
American Association of Blood Banks, 1999. pp 185207.
30. Avent ND, Reid ME. The Rh group system: a review. Blood 54. Zipursky A, Chintu C, Brown E et al. The quantification of
2000: 95; 375387. spherocytes in ABO hemolytic disease. J Pediatr 1979; 94:
31. Wagner FF, Gassner C, Muller DS et al. Molecular basis of 965967.
weak D phenotypes. Blood 1999; 93: 385393. 55. Bowman JM. Immune hemolytic disease. In Nathan DG
32. Lee D, Contreras M, Robson SC et al. Recommendations for (ed.): Hematology of infancy and childhood. Philadelphia,
the use of anti-D immunoglobulin for Rh prophylaxis. WB Saunders, 1997; pp 5378.
Transfusion Medicine 1999; 9: 9397. 56. Dunn PM. Obstructive jaundice and haemolytic disease of
33. Pollack W, Ascari WQ, Kochesky RJ et al. Studies on Rh the newborn. Arch Dis Child, 1963; 38: 54.
prophylaxis. 1. Relationship between doses of anti-Rh and 57. Phibbs RH, Johnson P, Tooley WH. Cardiorespiratory status
size of antigenic stimulus. Transfusion 1971, 11: 333339. of erythroblastotic newborn infants. II. Blood volume,
34. Urbaniak SJ, Robertson AG. A successful programme of hematocrit, and serum albumin concentration in relation to
immunising Rh-negative male volunteers for anti-D hydrops fetalis. Pediatrics 1974; 53: 13.
production using thawed blood. Transfusion 1981; 21: 58. Volpe JJ. Bilirubin and brain injury. In Volpe JJ (ed.):
6469. Neurology of the newborn. Philadelphia: WB Saunders,
35. Jakobowicz R, Williams L, Silberman F. Immunisation of 1987, pp 386408.
Rh-negative volunteers by repeated injection of very small 59. Harris LE, Farrell FJ, Shorter RG et al. Conjugated serum
amounts of Rh-positive blood. Vox Sang 1972; 23: 37681. bilirubin in erythroblastosis fetalis: an analysis of 38 cases.
36. Voak D, Cann R, Finney RD et al. Guideline for Mayo Clin Proc 1962; 37: 574.
administration of Blood products: transfusion of infants and 60. Shenker S, Dawber NH, Schmid R. Bilirubin metabolism in
neonates. Transfusion Medicine 1994; 4: 6369. the fetus. J Clin Invest 1964; 43: 32.
37. Napier JAF, Chapman JF, Forman K et al. Guideline for pre- 61. Margulies M, Voto LS, Mathet E et al. High does intravenous
transfusion compatibility procedures in blood transfusion IgG for the treatment of severe rhesus alloimmunisation.
laboratories. Transfusion Medicine 1996; 6: 273283. Vox Sang 1991; 61: 181189.
60 Blood Reviews

62. Urbaniak SJ, Duncan JI, Armstrong-Fisher SS et al. Transfer 83. Hessner MJ, McFarland JC, Eindeen DJ. Genotyping of
of anti-D antibodies across the isolated perfused human KEL1 and KEL2 of the human Kell blood group system by
lobule and inhibition by high dose intravenous PCR-SSP. Transfusion 1996; 36: 4959.
immunoglobulin: a possible mechanism of action. Br J 84. Lighten AD, Overton, TG, Sepulveda W, et al. Accuracy of
Haematol, 1997; 96: 186193. prenatal determination of RhD type status by polymerase
63. Voto LS, Sexer H, Ferreiro G, et al. Neonatal administration chain reaction with amniotic cells. Am J Obstet Gynecol
of high-dose intravenous immunoglobulin in rhesus 1995; 173: 11825.
hemolytic diease. J Perinat Med 1995; 23: 443451. 85. Avent ND, Martin PG, Armstrong-Fisher SS et al. Evidence
64. Moise KJ. Changing trends in the management of red blood of genetic diversity underlying RhD-, weak D (Du), and
cell alloimmunization in pregnancy. Arch Pathol Lab Med partial D phenotypes as determined by multiplex polymerase
1994; 118: 4218. chain analysis of the RHD gene. Blood 1997; 89: 25682577.
65. Voak D, Mitchell R, Bowell P et al. Guidelines for blood 86. Okuda H, Kawano M, Iwamoto S et al. The RHD gene is
grouping and red cell antibody testing during pregnancy. highly detectable in RHD-negative Japanese donors. J Clin
Transfusion Medicine 1996; 6: 7174. Invest 1997; 100: 373379.
66. Sebring ES. Fetomaternal Hemorrhage Incidence and 87. Crombach G, Niederacher D, Larbig D et al. Reliability and
Method of Detection and Quantitation. Hemolytic Disease clinical application of fetal genotyping with two different
of the Newborn. ed. G Garratty. American Association of fluorescent duplex polymerase chain reaction assays: three
Blood Banks 1984; 87117. years experience. Am J Obstet Gynecol 1999; 180: 435440.
67. Van den Veyver I, Saade G, Lockett L, et al. The use of 88. Avent ND, The rhesus blood group system: Insights from
Kleihauer-Betke stain in pregnant patients with abdominal recent advances in molecular biology. Trans Med Revs 1999;
trauma. Am J Obstet Gynecol. 1993; 168: 432. 13: 245266.
68. Kelsey P, Reilly JT, Chapman JF et al. The estimation of 89. Maaskant-van Wijk PA, Faas BH, de Ruiter JA, et al.
fetomaternal haemorrhage. Transfusion Medicine 1999; 9: Genotyping of RHD by multiplex polymerase chain reaction
8792. analysis of six RHD specific exons. Transfusion 1998; 38:
69. Clark D, Greiss MA, Urbaniak SJ. A prospective study of 10151021.
routine antenatal enzyme antibody screening demonstrates 90. Flegel WA, Wagner FF, Muuler R, Gassner C. Rh
lack of clinical value in predicting haemolytic disease of the phenotyping prediction by DNA typing and its application to
newborn. Br J Haematol 1999; 106: 824826. practice. Transfus Med 1998; 8: 281302.
70. Bruce M, Chapman JF, Duguid J et al. Addendum for 91. Woodrow JC, Clark CA, Donohoe WTA et al. Mechanisms
guidelines for blood grouping and red cell antibody testing of Rh prophylaxis: An experimental study on specificity of
during pregnancy. Transfusion Medicine 1999; 9: 99. immunosuppression. BMJ 1975; 2: 5759.
71. Clark AC. Red-cell antibodies in pregnancy: evidence 92. Karlsson MC, Wernersson S, Diaz de Stahl T et al. Efficient
overturned. Lancet 1996; 347: 4856. IgG-mediated suppression of primary antibody responses in
72. Urbaniak SJ, Greiss MA, Crawford RJ et al. Prediction of Fc-gamma receptor-deficient mice. Proc Natl Acad Sci USA
the outcome of rhesus disease of the newborn: additional 1999; 96, 5: 22449.
information using an ADCC assay. Vox Sang 1984; 46: 93. National Blood Transfusion Service Immunoglobulin
323329. Working Party. Recommendations for the use of anti-D
73. Hadley, AG, Wilkes A, Goodrick J et al. The ability of the immunoglobulin. Prescribers Journal 1991; 31: 137145.
chemiluminescence test to predict clinical outcome and the 94. Standing Medical Advisory Committee. Memorandum on
necessity for amniocenteses in pregnancies at risk of haemolytic disease of the newborn, 1976 addendum 1981.
haemolytic disease of the newborn. Br J Obstet Gynaecol London: Department of Health and Social Security, 1982.
1998; 105: 231234. 95. Contreras M, The prevention of Rh haemolytic disease of
74. Iskaros J, Kingdom J, Morrison JJ et al. Prospective non- the fetus and newborn General Background. Br J Obstet
invasive monitoring of pregnancies complicated by red cell Gynaecol, 1998; 105: Supplement 18, 710.
alloimmunization. Ultrasound Obstet Gynaecol 1998; 11: 96. Robson SC, Lee D, Urbaniak SJ. Anti-D immunoglobulin in
432437. RhD prophylaxis. Commentaries. Br J Obstet Gynaecol 1998;
75. Mari G. Noninvasive diagnosis by Doppler ultrasonography 105; 129134.
of fetal anemia dut to maternal red-cell alloimmunization. N 97. Tovey LAD. Haemolytic Disease of the Newborn and its
Engl J Med 2000; 342: 914. Prevention. Ed Marcela Contreras. ABC of Transfusion.
76. Liley AW. Liquor amnii analysis in management of BMJ Publishing Group 1992, 2225.
pregnancy complicated by rhesus immunisation. Am J Obstet 98. Standing Medical Advisory Committee. Haemolytic Disease
Gynecol 1961; 82: 1359. of the Newborn. London: Department of Health and Social
77. Queenan JT, Tomai TP, Ural SH et al. Deviation in amniotic Security, 1976.
fluid optical density at a wavelength of 450 nm in Rh- 99. Bowman JM, Pollock JM. Rh isoimmunisation during
immunized pregnancies from 14 to 40 weeks gestation: a pregnancy: antenatal prophylaxis. Can Med Assoc J 1978;
proposal for clinical management. Am J Obstet Gynecol 118: 623627.
1993; 168: 13701376. 100. Bowman JM, Pollock JM. Antenatal prophylaxis of Rh
78. Whitfield CR, A three year assessment of an Action Line isoimmunisation: 28 week-gestation service program. Can
method of timing intervention in rhesus isoimmunisation. Med Assoc J. 1978; 118: 627630.
Am J Obstet Gynecol 1970; 108: 12391244. 101. Tovey LAD Townley A, Stevenson NJ et al. The Yorkshire
79. Bennett PR, Le Van Kim C, Colin Y, et al. Prenatal antenatal anti-D immunoglobulin trial in primigravidae.
determination of fetal RhD type by DNA amplification. Lancet 1983; ii: 244246.
N Engl J Med 1993; 329: 60710 [comment in N Engl J Med 102. Herman M, Kjellman H, Ljunggren C. Antenatal prophylaxis
1993; 329; 65860 and 1994; 330: 7956]. of Rh immunisation with 250 mcg of anti-D
80. Simsek S, Blecker PMM, Von dem Borne AEGKr. Prenatal immunoglobulin. Acta Obstet Gynecol Scand (suppl), 1984;
determination of fetal RhD type. N Engl J Med, 1994; 330: 124: 415.
795. 103. Bowman JM, Pollock JM. Failures of intravenous Rh
81. Simsek S, Faas BHW, Blecker PMM, et al. Rapid RhD immune globulin prophylaxis: an analysis of the reasons for
genotyping by polymerase chain reaction-based amplification such failures. Transfus Med Rev 1987; 1: 101112.
of DNA. Blood 1995; 85: 297580. 104. Huchet J, Dallemagne S, Huchet C, et al. The antepartum use
82. Gussner C, Schmarda A, Kilger-Nogler B, et al. RhD/CE of anti-D immunoglobulin in rhesus negative women. Parallel
typing by polymerase chain reaction using sequence-specific evaluation of fetal blood cells passing through the placenta.
primers. Transfusion 1997; 37: 10206. The results of a multicentre study carried out in the region of
RhD haemolytic disease 61

Paris. J Gynecol Obstet Biol Reprod (Paris) 1987; 16: 112. Power JP, Lawlor E, Davidson et al. Molecular epidemiology
101111. of an outbreak of infection with hepatitis C virus in
105. Trolle B. Prenatal Rh-immune globulin prophylaxis with recipients of anti-D immunoglobulin. Lancet 1995; 346:
300 mcg immune globulin anti-D in the 28th week of 372373.
pregnancy. Acta Obstet Gynecol Scand. 1989; 68: 4547. 113. Turner ML, Ironside JW, New-variant Creutzfeldt-Jacob
106. Thornton JG, Page C, Foote G, et al. Efficacy and long term disease: the risk of transmission by blood transfusion. Blood
effects of antenatal prophylaxis with anti-D immunoglobulin. Revs 1998; 12: 255268.
BMJ 1989; 298: 16711673. 114. Al-Mufti R, Howard C, Overton T et al. Detection of fetal
107. Lee D, Rawlinson V. Multicentre trial of antepartum low messenger ribonuceic acid in maternal blood to determine
dose anti-D immunoglobulin. Transfus Med 1995; 5: 1519. fetal RhD status as a strategy for noninvasive prenatal
108. Urbaniak SJ. The Scientific Basis of Antenatal Prophylaxis. diagnosis. Am J Obstet Gynecol 1998; 179: 2104.
Br J Obstet Gynaecol, 1998; 105: Supplement 18, 1118. 115. Lo YMD, Hjelm NM, Fidler, et al. Prenatal diagnosis of
109. Vick S, Cairns J, Urbaniak S et al. An economic evaluation of fetal RhD status by molecular analysis of maternal plasma.
antenatal anti-D prophylaxis in Scotland. Health Economics N Engl J Med 1998; 339: 17341738.
1996; 5: 319328. 116. Thompson KH, Hughes-Jones NC. Production and
110. Cairns JA. Economics of Antenatal Prophylaxis. Br J Obstet characterisation of monoclonal anti-Rh. In: Contreras
Gynaecol. 1998; 105: Supplement 18, 1922. Marcela (eds.), Blood Transfusion: The Impact of new
111. Tabor E, The epidemiology of virus transmission by plasma Technologies. Bailliere Clinical Haematology Series, Vol 3
derivatives: clinical studies verifying lack of transmission of London: Bailliere, 1990; 24353.
hepatitis B and C viruses and HIV type 1. Transfusion 1999;
39: 11601168.

Anda mungkin juga menyukai