Haemolytic Disease of The Fetus and Newborn: Review

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 15

Vox Sanguinis (2015) 109, 99–113

© 2015 International Society of Blood Transfusion


REVIEW DOI: 10.1111/vox.12265

Haemolytic disease of the fetus and newborn


M. de Haas,1,2 F. F. Thurik,2 J.M. Koelewijn2,3 & C.E. van der Schoot2
1
Department of Immunohaematology Diagnostics, Sanquin Diagnostic Services, Amsterdam, the Netherlands
2
Department of Experimental Immunohaematology, Sanquin Research Amsterdam and Landsteiner laboratory, Academic Medical Centre, University
of Amsterdam, Amsterdam, the Netherlands
3
Department of General Practice, University Medical Centre, Groningen, the Netherlands

Haemolytic Disease of the Fetus and Newborn (HDFN) is caused by maternal


alloimmunization against red blood cell antigens. In severe cases, HDFN may
lead to fetal anaemia with a risk for fetal death and to severe forms of neonatal
hyperbilirubinaemia with a risk for kernicterus. Most severe cases are caused by
anti-D, despite the introduction of antental and postnatal anti-D immunoglobulin
prophylaxis. In general, red blood cell antibody screening programmes are aimed
to detect maternal alloimmunization early in pregnancy to facilitate the identifi-
cation of high-risk cases to timely start antenatal and postnatal treatment. In
this review, an overview of the clinical relevance of red cell alloantibodies in
relation to occurrence of HDFN and recent views on prevention, screening and
Received: 24 April 2013,
treatment options of HDFN are provided.
revised 11 January 2015,
accepted 2 February 2015, Key words: haemolytic disease of the fetus and newborn, RBC antigens and anti-
published online 20 April 2015 bodies, alloimmunisation in pregnancy, anti-D prophylaxis, fetal genotyping.

induce HDFN is about 1 in 500 pregnancies, albeit there is


Introduction
a lower risk to develop severe disease [1–4, 7]. In the past
Haemolytic disease of the fetus and newborn (HDFN) is a decade, non-invasive monitoring of high-risk cases by
disease which – if untreated – can cause perinatal mortal- laboratory testing, including fetal antigen typing with
ity and morbidity with a substantial risk for long-term cell-free fetal DNA from maternal plasma, followed, if
sequelae [1–5]. HDFN is caused by maternal red cell al- necessary, by ultrasound-based techniques to judge the
loantibodies of the IgG class that are actively transported presence of fetal anaemia, replaced invasive procedures
across the placenta and destroy fetal erythroid cells carry- for monitoring fetal haemolysis and anaemia [8]. Since
ing the involved antigen. In most severe cases of HDFN, the potency of red cell antibodies to induce HDFN differs,
alloimmunization against the RhD antigen (in this review, it is questionable whether repeated antibody screening
we will further refer to the RhD antigen or phenotype as and laboratory monitoring is necessary in all pregnancies
D) is involved. Introduction of anti-D immunoglobulin (Ig) complicated by the presence of red cell alloantibodies.
prophylaxis has drastically decreased the prevalence of D Furthermore, introduction of antenatal anti-D prophylaxis
antibodies and of anti-D-mediated HDFN. Despite ade- in a screening programme, in combination with the cur-
quate antenatal and postnatal anti-D Ig prophylaxis 1 to 3 rent possibilities of non-invasive fetal RHD typing with
in 1000 D-negative women still develop anti-D [6]; overall cell-free fetal DNA from maternal plasma, has been the
in the whole pregnant population, the prevalence of anti- topic of several cost-benefit studies. In this review, we
D sensitized pregnancies is about 1 in 1000, as determined provide an overview of the clinical relevance of red cell
in the Netherlands (data not shown). The prevalence of red alloantibodies in relation to development of severe HDFN
cell antibodies other than anti-D with the potency to and summarize recent views on prevention, screening, fol-
low-up and treatment options of cases with HDFN.
Correspondence: Masja de Haas, Department of Immunohaematology
Diagnostics, Sanquin Diagnostic Services, Plesmanlaan 125, 1066 CX Literature search
Amsterdam, the Netherlands
E-mail: m.dehaas@sanquin.nl A literature search was conducted in Pubmed using the
De Haas and Thurik contributed equally to the paper. following search terms (free-text terms): hemolytic dis-

99
100 M. de Haas et al.

ease of the newborn OR haemolytic disease of the new- anti-GPMur [11]) induce destruction of fetal red cells as
born OR hemolytic disease of the newborn and fetus OR well as destruction of erythroid progenitor cells, resulting
haemolytic disease of the newborn and fetus OR fetal in early anaemia without erythroblastosis. Compensation
erythroblastosis OR maternal alloimmunisation OR preg- for the anaemia results in a hyperdynamic circulation in
nancy immunisation OR red-cell alloimmunisation OR the fetus causing cardiomegaly and eventually fetal hy-
irregular antibodies OR maternal alloimmunization OR drops, a condition consisting of oedema in the fetal skin
pregnancy immunization OR red-cell alloimmunization and serous cavities. Haemolysis of the fetal red cells
OR alloimmunised pregnant women OR alloimmunized results in raised bilirubin levels. Since bilirubin passes the
pregnant women. Limits were set for time (i.e. published placenta, excess of bilirubin is cleared via the maternal
in the last 10 years), species (i.e. humans), type of article circulation during pregnancy. After birth, the haemolytic
(i.e. exclusion of letters and editorials) and language (i.e. process continues, but the relatively immature liver of the
English and Dutch). Title and abstract were screened of neonate cannot sufficiently conjugate the excess of biliru-
1293 articles independently by two of the authors (FT bin. This may result in severe hyperbilirubinaemia and,
and MH). In addition, the snowball method was applied when untreated, even in irreversible damage to the cen-
and books written by experts on this specific topic were tral nervous system, a condition known as ‘kernicterus’.
consulted in order to avoid overlooking relevant litera- This condition is characterized by bilirubin deposition in
ture. The search query was last updated in June 2014. the basal ganglia and brain stem nuclei and is correlated
with long-term morbidity that can consist of a severe
form of athetoid cerebral palsy, hearing problems and
Pathophysiology of haemolytic disease of the
psychomotor handicaps. When the presence or develop-
fetus and newborn
ment of red cell alloantibodies is not detected during
Maternal alloimmunization can be triggered by prior pregnancy, the only, nonspecific and unpredictable clini-
incompatible blood transfusions or by fetomaternal haem- cal features of HDFN during pregnancy are a decrease of
orrhage (FMH) in a previous or the current pregnancy. fetal movements or sudden fetal death, while after birth
Only antibodies of the IgG class are actively transported (early), neonatal jaundice can occur. When the presence
across the placenta. Red cell alloantibody-mediated of maternal alloantibodies is detected by screening, the
destruction of fetal red cells in the fetal spleen may result pregnancy can be monitored by repeated laboratory tests
in anaemia (see Fig. 1). If the child is positive for the and – if necessary – by clinical investigations. In those
involved red blood cell antigen, the maternal alloantibod- cases, prenatal anaemia can be corrected by intrauterine
ies will bind. Fetal anaemia will induce compensatory blood transfusions to prevent development of hydrops
erythropoiesis, nonetheless this may be insufficient. Red and asphyxia at birth and development of kernicterus can
cell alloantibodies directed against antigens of the Kell be prevented by timely starting treatment with photother-
system [9, 10] and also against the MNSs system (e.g. apy or, if necessary, with exchange transfusions to lower

Maternal red cell antibodies


Mother IgM IgG

placenta
Antigen-positive
Fetus fetal blood cells
Kell and GP antibodies
Extravascular Destruction
haemolysis red cell progenitors

Anaemia with erythroblastosis

Bilirubin Extramedullary Hydrops Child movements


erythropoiesis fetalis
(ultrasound) Blood flow rate
Enlargement (Doppler
liver and spleen Heart failure ultrasound)
(ultrasound)
Fetal death
Newborn Icterus
Fig. 1 Illustrates the pathogenesis of
haemolytic disease of the fetus and newborn in
Kernicterus
the mother, fetus and newborn.

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Haemolytic disease of fetus and newborn 101

Maternal
alloimmunisation Fetus antigen- No HDFN Normal
negative*
Antibody with development
risk to cause Fetus antigen- Postnatal moderate
severe HDFN positive* hyperbilirubinaemia
and/or anaemia
No kernicterus
Postnatal severe
hyperbilirubinaemia Neonatal
Fetal death Antenatal anaemia +/– Kernicterus
severe death
anaemia
Postnatal severe
(hydrops)
anaemia
hyperbilirubinaemia +/–
Long-term
Non-invasive Laboratory monitoring: sequelae
fetal antigen typing Repeated titer/bio assay
possible for: If above critical titers:
RhD, C, c, E, K* IUT Phototherapy Exchange RBC
Clinical monitoring with: transfusion transfusion
Doppler ultrasonography

Fig. 2 Disease model maternal alloimmunization leading to haemolytic disease of the fetus and newborn and laboratory testing and eventual clinical
testing to discriminate high-risk cases. *If non-invasive fetal antigen typing is not available, pregnancies should be monitored as if the fetus is antigen
positive when the biological father is homozygous or heterozygous positive for the respective antigen. Pregnancies can be monitored as if the fetus is
antigen negative upon a negative non-invasive fetal antigen typing result, or when the biological father is typed with certainty as homozygous negative
for the respective antigen.

bilirubin levels postnatally (Fig. 2). Thus, a screening pro- anti-K (Table 1). In the Netherlands, we performed a pro-
gramme aims to identify those pregnancies in which spective index-cohort study based on 298 000 screened
intrauterine treatment of the fetus is needed and/or deliv- pregnant women to study the development of HDFN,
ery should be induced to lower the risk of development caused by non-D antibodies. K antibodies caused severe
of severe haemolytic disease (Fig. 2). Without a screening HDFN in 26% of the pregnancies at risk (antigen-positive
programme, treatment may be delayed. In this review, fetus), c antibodies in 10%, E antibodies in 2% and anti-
‘very severe HDFN’ is used for cases of intrauterine death bodies directed against another Rh antigen in 5% of the
and cases treated by intrauterine transfusions (IUTs) and/ pregnancies [7]. Phototherapy was necessary in 42% of the
or exchange transfusion (ET) after birth, ‘severe HDFN’ pregnancies complicated by K antibodies (antigen-positive
also includes cases treated with top-up transfusions and fetus), in 33% of the pregnancies complicated by c anti-
cases in which preterm induction of labour was needed. bodies, in 19% when E antibodies were present and in 20%
A ‘mild course’ is defined as cases without a need of pre- of the pregnancies complicated by antibodies directed
term induction of labour and in whom treatment with against other antigens of the Rh blood group system [7].
phototherapy suffices. This is comparable to other published single-centre studies
[14, 15]. Antibodies against Duffy antigens (e.g. Fya) have
been reported to induce severe HDFN that requires intra-
Clinical relevance of different red cell
uterine blood transfusion. This, however, rarely occurs [7,
alloantibody specificities
12, 16]. In our study, we observed no cases of severe HDFN
The risk of developing severe HDFN depends on several in 42 antigen-positive fetuses but phototherapy was
factors, including Ig class, specificity of the red cell alloan- needed in 14% of the cases, compared to 4% in controls
tibodies and level of expression of the involved blood (no antibodies present or antigen-negative fetus) [7]. In the
group antigen on the fetal red cells and other tissues. In literature, a wide variety of red cell alloantibodies is
Table 1, an overview is given of red cell antibody specifici- reported as case reports to cause severe HDFN. In popula-
ties in regard to their risk to induce HDFN (Table 1) [1–4, 7, tion studies, however, these antibodies are seldom reported
12]. Anti-D is correlated with the highest risk of fetal mor- as cause of severe HDFN (Table 2). However, this may
tality and morbidity [4, 13]. The risk to develop severe depend on the ethnic background of the population stud-
HDFN is much lower in pregnancies complicated by other ied. For example, in Asian population anti-GPMur may
maternal red cell alloantibodies, with the exception of cause severe HDFN [11] and recently some case reports

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
102 M. de Haas et al.

Table 1 Correlation of red cell alloantibody specificities with occurrence Given the high rate of ABO incompatibility between the
of haemolytic disease of the fetus and newborn [1–4, 7, 12] mother and her fetus, the prevalence of clinically signifi-
cant haemolysis in neonates because of anti-A or anti-B is
Risk to develop HDFN in antigen-positive children
relatively low. And, if occurring, it shows a mild clinical
and clinical course of disease
course. A and B antigens are expressed by placental tissue
ABO Lowb risk for disease, in general milda, and may bind maternal alloantibodies to some extent.
incidentally severea Furthermore, the expression of A and B blood group anti-
Rh gens on fetal red cells is not fully developed. Anti-A and
D Highb risk for disease, often (very) severe, anti-B are predominantly of the IgM class, whereas anti-
otherwise mild A,B in group O women is often of IgG class; during preg-
c High risk for disease, (very) severe or mild
nancy, the formation of anti-A, anti-B and anti-A,B titres
E Mediumb risk for disease, sometimes severe,
of the IgG class can be strongly increased. There is a strik-
but mostly mild
Other Rh Medium risk for disease, incidentally severe,
ing, and yet not understood, difference in the incidence of
antigens but mostly mild ABO-mediated HDFN between populations. The incidence
Kell is around 03–08% in the Caucasian population, versus 3–
K High risk for disease, (very) severe or mild 5% in Black or Asian populations, also with a more severe
Other Kell Medium risk mild to severe disease clinical course [18–21].
antigens Expression of some blood group antigens is very low
Duffy on fetal red cells (e.g. Lub, Yta), and therefore, significant
Fya/Fyb Medium risk for disease, mostly mild haemolysis is not induced (Table 1). Finally, in some
Kidd cases, for example with Lutheran antibodies, another rea-
Jka/Jkb Low riskb for disease, only mild
son for absence of HDFN could be the presence of antigen
MNS
on placental cells, preventing transfer of antibody to the
M, N, S, s Low riskb for disease, mostly mild disease,
very rarely severe
fetus.
Other antigens Low riskb for disease, mostly mild disease,
very rarely severe Laboratory monitoring of alloimmunized
I, Le, P1, Lu, Yt No risk, because of very low expression of these
cases
antigens by fetal cells
Other antigen Very low risk, very rarely severe disease can develop Since the last decade, non-invasive fetal blood group typ-
systems ing with cell-free fetal DNA from maternal plasma is a
a
clinical reality and is offered by many laboratories for
Very severe disease: Need for intrauterine treatment and/or exchange
pregnancies complicated by antibodies that can cause
transfusion after birth. Severe disease: Need for intrauterine treatment
and/or preterm induction of labour and/or blood transfusions after birth.
severe HDFN (Fig. 2) [22–28]. Because cell-free fetal DNA
Mild disease: Only treatment with phototherapy is needed. represents only a fraction of the total cell-free DNA in
b
High risk: >50%, medium risk >10–50%, low risk 1–10%, very low/inci- the maternal plasma and is present in extremely low con-
dentally <1%. centrations, highly efficient DNA extraction and sensitive
detection technology, currently with quantitative PCR, is
required. In the literature, different assays have been
report on the occurrence of anti-Ge3 in Hispanic families described for reliable genotyping of D, C, c, E and K in
responsible for HDFN with severe late-onset haemolysis the first trimester of pregnancy and thus in time before
and hyperbilirubinaemia [17]. treatment should start [22–25, 27, 28]. In these assays,
Anti-M is very often found in pregnant women as a the fetal antigen-negative phenotype is not detected
so-called naturally occurring antibody (formed without a directly, but concluded based on a negative result of the
previous immunization event) of the IgM class. Anti-M of antigen-specific PCR. Therefore, confirming the presence
the IgG class can, however, cause severe HDFN, like other of fetal DNA with other paternally inherited DNA
antibodies directed against antigens of the MNS blood sequences (e.g. SRY) or a universal fetal DNA marker (e.g.
group system [11]. We observed in 69 women with methylated RASSF1a) is used by some laboratories to
anti-M of the IgM class early in pregnancy that a class report conclusive results in one single assay [22, 29]. Or
switch to IgG had not occurred as analysed in week 24, the assay is repeated later in pregnancy to assure the
30 and 36 of pregnancy [7]. Therefore, if anti-M of the presence of sufficient amounts of fetal DNA. One can
IgM class is detected early in pregnancy, it is not likely conclude that non-invasive fetal genotyping can reliably
that this antibody will switch to IgG and induce severe be used to target laboratory and clinical monitoring to
HDFN; therefore, repeated testing is not necessary. cases at risk for the development of severe HDFN.

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Table 2 Studies on the prevalence of maternal alloimmunization for other specificities than anti-D and subsequent occurrence of haemolytic disease of the fetus and newborn

Prevalence of non-D alloimmunization and subsequent


very severe HDFNa

Detected upon 1st Detected after 1st


screening screeningb
Screening Antibodies causing
Study Period Study Population/setting moment(s) Immunization HDFN Immunization HDFN severe HDFN Remarks

Hardy, Wales, 1981 [75] 1948–1978 Retrospective D+ women, regional Unclear 019% 0003% NRc Denominator

Vox Sanguinis (2015) 109, 99–113


733/380 790 11/380 790 women instead
of pregnancies
Bowell, UK, 1986 [76] 1983–1984 Retrospective All pregnant women, Intake 045% NR 029% NR Denominator:
regional week 28 315/70 000 201/70 000 estimation
after week 28

© 2015 International Society of Blood Transfusion


Belfrage, Sweden, 1992 [77] 1983–1989 Retrospective Referral university ‘Initially’ ND 001% Unclear
hospital 17/147 068 (E, c, K, etc.
Cw+Fya)
Heddle, Canada, 1993 [78] 1980–1990 Retrospective Community hospital + 1st trimester 033% NR 024% NR
1987–1989 secondary care hospital 2nd screening 58/17 568 42/17 510
moment
unclear
Gottvall, Sweden, 1993 [79] 1983–1989 Prospective All pregnant women, Week 25 024% 001% Moment of NR Only pregnancies
regional Week 35 188/78 300 8/78 300 detection unclear with an antigen-
Filbey, Sweden, 1995 [80] 1980–1991 Retrospective All pregnant women, Week 10 015% 0005% Moment of c, E, cE, K, Fya positive partner
regional Week 35 171/111 939 6/111 939 detection unclear were included
Wong, Hong Kong, 1997 [81] 1995–1996 Prospective Pregnant women, Intake 025% 0 ND
regional 5/1997 0/1997
Howard, UK, 1998 [82] 1993–1994 Retrospective All women 7 maternity Intake 065% 0018% c, ce, E
units 144/22 264 4/22 264
Rothenberg, USA, 1999 [83] 1988–1997 Retrospective D+ women, tertiary 1st trimester 037% 0011% 006% 0 Fya
care referral 3rd trimester 35/9348 1/9348 6/9313 0/9313
centre + public
hospital
Andersen, Denmark, 1996 Prospective D+ women, regional 1st trimester 11% NR 043% 0 1st trimester:
2002 [84] 3rd trimester 34/3046 13/3012 0/3012 including Lewis
antibodies
Lee, Hong Kong, 2003 [85] 1997–2001 Retrospective National screening Intake 027% 0005% NR NR E
laboratory, ethnic 57/21 327 1/21 327
Chinese women
Haemolytic disease of fetus and newborn 103
Table 2 (Continued)

Prevalence of non-D alloimmunization and subsequent


very severe HDFNa

Detected upon 1st Detected after 1st


screening screeningb
Screening Antibodies causing
104 M. de Haas et al.

Study Period Study Population/setting moment(s) Immunization HDFN Immunization HDFN severe HDFN Remarks

Jovanovic, Yugoslavia, 1995–2000 Retrospective National screening Unclear 058% ND Unclear whether
2003 [86] laboratory 124/21 370 number of ab’s
or number of
pregnancies is
reported; unclear
whether non-D
antibodies are
combined with
D antibodies
Lurie, Israel, 2003 [88] 1999–2002 Retrospective Women’s Health 1st trimester 016% 0
Centre 1/631 0/631
Ameen, Kuwait, 2005 [87] 1992–2001 Retrospective National Blood Bank Unclear 62% ND
182/2932
Adeniji, UK, 2007 [89] 2002–2004 Retrospective D+ women, regional 1st trimester ND 024% 0
>week 28 34/14 143 0/14 143
Gottvall, Sweden, 2008 [90] 1992–2005 Retrospective All pregnant women, Week 10 025% 001% Moment of c, E, c+E, Fya Only cases with
regional Week 35 196/78 145 8/78 145 detection antigen-positive
unclear father included
Koelewijn, Netherlands, 2002–2004 Prospective All pregnant women, Intake 033% 0007% 0002% c, E, K, C+Jka, Mur Denominators
2008 [7] national 1002/305 700 21/305 700 8/393 500 estimated
Dajak, Crotia, 2011 [12] 1993–2008 Retrospective All pregnant women, Intake 016% 004% 001% c, C, E, Rh17, Hospital-based
regional 143/84 000 30/84 000 11/84 000 Cw, K, Fya, S study. Inclusion
from population
with putative
higher risk for
alloimmunization

a
Very severe HDFN: need for intrauterine transfusion and/or exchange transfusion, perinatal death or fetal hydrops because of maternal RBC antibodies.
b
Incidence of antibodies, newly detected later in pregnancy, after a negative first trimester screening.
c
NR Not-reported.

Vox Sanguinis (2015) 109, 99–113


© 2015 International Society of Blood Transfusion
Haemolytic disease of fetus and newborn 105

In many countries, cases with an increased risk for amniotic fluid upon amniocentesis. Amniocentesis is an
severe HDFN (further: ‘high-risk cases’) are discriminated invasive procedure with complications including
based on red cell alloantibodies titres, as determined by spontaneous miscarriage and amniotic fluid leakage
the indirect antiglobulin test performed in saline (Fig. 2) [40]. Moreover, each invasive procedure carries an
[3, 30–32]. The blood sample is retested at regular inter- increased risk for FMH, which can trigger an antibody
vals until a threshold or cut-off value (‘critical titre’) is response [41].
exceeded, which implies an increased risk for severe Advances in Doppler ultrasonography enabled non-
HDFN and referral to specialized care is indicated (see invasive detection of early fetal anaemia. Mari et al. [42]
‘clinical assessment of high-risk cases’). Efficient prese- reported that peak systolic velocity measurement of the
lection of high-risk cases reduces the number of admis- middle cerebral artery (MCA-PSV) can be used for
sions for specialized clinical diagnostics. In some detection of fetal anaemia. A velocity of more than 15
countries, only pregnant women with anti-D, anti-c or multiples of the median (MoM) for gestational age pre-
anti-K are regularly retested and pregnant women with dicted all 70 cases of moderate to severe anaemia correct
other types of red cell antibodies are only tested again with a sensitivity of 100% (95% CI, 86–100). The false-
in week 28 to detect an infrequent high-risk case with positive rate was 12%.
high antibody titres in this group. Moise concludes that In a multi-centre international study, the diagnostic
in most laboratories for anti-D, a critical titre between 8 accuracy of MCA-PSV-Doppler ultrasonography for pre-
and 32 is used [3]. In the UK, a threshold value of 15 diction of severe fetal anaemia in alloimmunized cases
international units anti-D/ml has been recommended as showed a sensitivity of 88% (74 cases), a specificity of
critical titre [30]. In general, for non-D alloantibodies, 82% and an accuracy of 85% (95% CI, 78–93%, 73–89%
except for anti-K, a cut-off of 32 is used [3, 32]. Hack- and 79–90%, respectively). In comparison with amniocen-
ney et al. [14] reported for anti-c induced HDFN, tesis, Doppler ultrasonography was more accurate by 9
defined as need for intrauterine transfusion or haemo- percentage points (95% CI, 11–159) [43]. Therefore,
globin levels at birth of <11 g/dl, that a titre >32 pre- Doppler ultrasonography is nowadays used predominantly
dicted all severe cases. Similarly, Joy et al. concluded to identify cases with fetal anaemia among high-risk
for anti-E and McKenna et al. for anti-K, a cut-off of pregnancies [44].
32 [15, 33]. However, Leggat et al. and Van Wamelen
et al. reported that severe HDFN can already occur at
Antenatal treatment
very low anti-K antibody titres [34, 35]. Therefore, both
the British and American guidelines state that anti-K ti- In severe fetal anaemia, it is essential to treat HDFN ante-
tres <32 may already be relevant; a titre of 8 has been natally to prevent hydrops fetalis and fetal death. IUT
proposed by Moise [3, 30–32]. should be considered when clinical assessment indicates
In the past, several laboratories performed biological fetal anaemia, for example MCA-PSV >15 MoM [5]. Most
assays for the prediction of the antibody activity [36, 37]. studies reporting on series of IUT-treated cases report
The antibody-dependent cell-mediated cytotoxicity perinatal survival rates around 90% [45, 46]. Furthermore,
(ADCC), a monocyte-driven biological assay, is used in a long-term follow-up study on neurodevelopmental out-
the Netherlands to assess the risk of developing severe come shows a normal development in more than 95% of
disease. Oepkes et al. [38] showed that the use of this 291 included children [47].
assay improves the diagnostic accuracy of selection of In a cohort of 537 IUT-treated fetuses from the refer-
high-risk cases, because of the higher specificity of the ence centre for fetal treatment in the Netherlands, the
test than antibody titres alone. The higher positive predic- vast majority of IUTs were given because of alloimmuni-
tive value of the ADCC was also confirmed for non-D zation against D (81%), compared to K in 13%, c in 5%
antibodies [39]. From these studies, it was concluded that and antibodies against other antigens in 2% (D. Oepkes,
a non-D alloantibody titre of 16 or higher or an ADCC Leiden University Medical Centre, personal communica-
test result of 30% or higher indicate high-risk cases for tion).
which clinical monitoring is recommended [7]. In case of Intrauterine transfusion may lead to further alloimmu-
anti-D antibodies, the advice to the clinician is mainly nization in 25% of the mothers, despite preventive
based on the obtained ADCC test value [38]. matching for Rh and K antigens [41].
Additional treatment of alloimmunized women with
intravenous immunoglobulin (IVIg) with or without plas-
Clinical assessment of high-risk cases
mapheresis has only been described in three small case
The standard test to assess the severity of fetal anaemia series, but the efficacy of these experimental treatments
used to be the quantification of bilirubin levels in has not yet been proven [48–50].

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
106 M. de Haas et al.

similar number of top-up transfusions. In particular, neo-


Postnatal treatment
nates suffering from severe HDFN due to anti-c also
require top-up transfusions in the first months of life, with
Phototherapy
a positive correlation between the titre of anti-c measured
Phototherapy has been proven to be effective by drasti- in cord blood and the need for postnatal transfusion [60].
cally decreasing the necessity of exchange transfusion in
the treatment of hyperbilirubinemia [51]. Guidelines for
Intravenous immunoglobulin
treatment of neonatal icterus have been published by the
American Academy of Pediatrics (AAP) in 2004 and state In the literature, neonatal treatment with intravenous
that factors other than bilirubin level should be taken into immunoglobulin (IVIg) has been discussed as an alterna-
consideration in clinical assessment, such as gestational tive therapy for exchange transfusion. Initially, studies
age, birth weight and cause of hyperbilirubinemia [52]. were published that favoured this approach [61–63], but
In children treated with IUT, a shortened period of two later placebo-controlled randomized control trials did
phototherapy was needed compared to neonates suffering not show a beneficial effect of IVIg treatment [64, 65].
from HDFN who did not receive IUT (38 and 51 days, And recently Santos et al. [66] showed that IVIg does not
respectively, P-value 001) [53]. Rath et al. [54] found prevent the need for exchange transfusion in neonates
that less phototherapy was also required in neonates with anti-D-mediated HDFN. Hence, efficacy of IVIg
affected by anti-K-mediated haemolytic disease. Since treatment has not been proven.
anti-K also destroys erythroid progenitor cells, in these
cases anaemia manifests earlier and is more pronounced
Erythropoietin
relative to bilirubin levels.
Anaemia presented in the first weeks of life (‘late anae-
mia’) occurs in 71–83% of neonates with HDFN, and it is
Exchange and top-up transfusions
characterized by depressed erythropoiesis [57]. Although
Exchange transfusions (ET) have the advantage that both administration of EPO may prevent late anaemia and
bilirubin and maternal alloantibodies are removed from reduce the need for top-up transfusion of red blood cells,
the circulation, and anaemia is corrected [55]. Up to 24% there is no sufficient evidence to support this [57, 59].
of neonates treated with ET suffer from a complication,
such as electrolyte imbalance, cardiac arrhythmias, embo-
Screening programmes
lism, necrotizing enterocolitis, infection and sepsis. There
is a risk of almost 03% of ET-associated mortality in In most countries, pregnant women are typed for ABO and
term neonates [56, 57]. The guidelines published by the D early in pregnancy and screened for the presence of red
AAP do not recommend early ET, such as within the first cell antibodies [1, 4, 31, 44, 67–69]. Nowadays, screening
12 h of birth, and bilirubin thresholds at which time ET tests are very sensitive early in pregnancy. The level of
should be started differ depending on the gestational age antibodies developed earlier upon incompatible pregnan-
at which the baby is born [52]. Rath et al. [58] showed cies or transfusions, however, may be too low to detect.
that more restrictive ET criteria reduced the number of During pregnancy, small FMHs may initiate a secondary
exchange transfusions but resulted in an increase of the antibody response leading to sufficient high antibody levels
number of top-up transfusions. in the fetus to induce disease. To ascertain that all clinically
In contrast to the effect of IUT on phototherapy, De Boer relevant alloimmunizations are detected, screening pro-
et al. [53] found no difference in the number of exchange grammes in many western countries comprise two or even
transfusions required in neonates treated with or without three screening moments: in the first trimester, the second
IUT. Since maternal alloantibodies can be active for and/or third trimester. A repeated screen is meant to detect
months in the neonate [59] and recovery of erythropoiesis primary responses which may occur during pregnancy and
may be delayed as an effect of transfusions, additional secondary responses in women already previously immu-
transfusions may be necessary in the first months of life nized. For example, Dajak and co-workers showed that
[53]. De Boer et al. [53] found that neonates treated with severe anti-D-mediated HDFN was not detected in 12% (8/
IUT and suffering from anti-D-mediated HDFN needed 68) of the cases upon first trimester screening, but only
more top-up transfusions compared to neonates who did upon screening at week 28 or 34 of pregnancy (in women
not receive an IUT (77% and 265%, respectively, with a P- who did not receive anti-D Ig prophylaxis) [12]. We
value < 001). Compared to neonates suffering from anti- observed in a study in D-negative women who delivered
D-mediated HDFN, neonates suffering from anti-K-medi- previously a D-positive child that administration of both
ated HDFN require less ET and less phototherapy, but a antenatal and postnatal anti-D prophylaxis did not reduce

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Haemolytic disease of fetus and newborn 107

the incidence of ‘late’ development of anti-D as detected at the observed incidence of early and that of late detected
week 30 of pregnancy [6]. But in the group of women who non-D antibodies are low (ranging from 006% to 04%)
received both antenatal and postnatal prophylaxis and in and because the presence of these red cell antibodies is cor-
whom immunization was detected in week 30 of preg- related with a lower risk for development of severe HDFN,
nancy, the incidence of severe HDFN was reduced to 3% (1 most studies lack sufficient power to determine the clinical
out of 29) versus 28% (6 out of 215) if only postnatal anti- relevance of (repeated) red cell alloantibody screening. In a
D Ig prophylaxis was given in the first pregnancy [6]. The retrospective two-year nationwide study, representing
working mechanism of anti-D Ig prophylaxis is not yet elu- about 393 500 screened pregnant women, we identified
cidated, and an immunomodulatory effect of anti-D Ig pro- seven cases of non-D-mediated severe HDFN with a nega-
phylaxis has been proposed [70–72]. tive screening result early in pregnancy, anti-c: n = 3,
Although antenatal screening for the presence of red anti-c and anti-E: n = 2 and anti-E: n = 2. It was calcu-
cell antibodies is established practice in several countries, lated that a repeated screening test in c-negative (majority
there is discussion about necessity and cost-benefit ratio CCDee) pregnant women late in pregnancy would increase
of the screening itself and of repetition of red cell anti- the sensitivity of the screening programme with 22%. Simi-
body screening in pregnancies from D-positive women. A larly, Dajak et al. [12] showed that 275% (11/40) of cases
positive red cell antibody screening test does not only with severe HDFN were undetected by a first trimester
identify red cell antibodies correlated with an increased screening and detected at week 28 of pregnancy. It con-
risk of development of HDFN, but also a broad range of cerned cases with, respectively, anti-c (n = 7, 6 received an
irrelevant antibodies (e.g. cold agglutinins) and antibodies ET, one only a top-up transfusion), anti-E (n = 1, ET) anti-
which only rarely cause HDFN (Table 1). Antibody identi- C (n = 2, only top-up transfusion) and one case of anti-
fication and subsequent laboratory monitoring and possi- Rh17 (perinatal death). The involved pregnant woman in
bly also clinical monitoring result in substantial costs for the latter case and in one of the cases with anti-C received
the healthcare system. In order to evaluate (novel) screen- an incompatible blood transfusion during pregnancy. Both
ing programmes, the WHO criteria designed by Wilson Koelewijn and Dajak observed that parity is a strong risk
and J€ungner can be used [73]. One of the criteria is that factor for the presence of clinically relevant antibodies dur-
the test should be acceptable to the population. In the ing a subsequent pregnancy [12, 91, 92].
Netherlands, a survey among 233 screened women Thus to increase the cost-benefit ratio of repeated
showed a positive attitude towards the red cell antibody screening in pregnancy, one might consider to limit this
screening programme, with a positive balance between to all D-negative women and to multiparous, or earlier
perceived utility and burden of the screening programme, transfused, D-positive women (see Fig. 3).
independent of the screening test results [74]. Table 2 lists
a number of population studies looking into the benefits
Prevention of alloimmunization in pregnant
of a first trimester red cell antibody screening test in D-
women due to blood transfusion
positive women [7, 12, 75–90]. From this overview, it is
apparent that the prevalence of maternal alloimmuniza- We demonstrated that RBC transfusion is the most impor-
tion shows a wide variation from 015% to as high as tant risk factor for non-D immunization, even after
62% and that of severe HDFN from 0 to 18/100000 adjustment for parity [92]. Over 50% of women with clin-
births (Table 2). This variation can be explained by the ically relevant red blood antibodies had a history of prior
heterogeneity of the screening protocols, by the absence blood transfusion. This percentage was even higher in
of an unequivocal definition of ‘the presence of clinically women with anti-K antibodies (>80%).
relevant alloantibodies’ and, most importantly, by the use In blood transfusion guidelines, it is already included
of preselected high-risk study populations with an for a long time that premenopausal D-negative women
unknown relation to the general population of pregnant should receive D-negative blood. Since anti-c, anti-K and
women. Population data are available from three regional albeit to a lesser extent anti-E, can cause severe HDFN,
studies in Sweden, one in Croatia and our studies in the one should consider to transfuse girls and premenopausal
Netherlands [7, 12, 80, 90]. These latter studies show a women with RhcDE and K-compatible blood.
prevalence of 015–025% of alloimmunized pregnancies
with a risk for HDFN (partner antigen positive) caused by
Anti-D prophylaxis
a red cell antibody other than anti-D (non-D mediated).
To assess the validity of repeating the screening in preg- Routine postnatal administration of anti-D – introduced in
nancies for D-positive women, a study should be of a suffi- the 1960s – has been shown to substantially decrease
cient sample size to investigate the effectiveness of a maternal alloimmunization detected in a subsequent preg-
screening programme. In the studies listed in Table 2, both nancy from 15% to 16% [93]. In the more recent years,

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
108 M. de Haas et al.

Actions Actions
OBSTRETIC CARE GIVER/ LABORATORY
PAEDIATRICIAN

D typing and red cell antibody screening


Blood sampling Consider c and E typing (1)
Booking visit If D-negative mother: consider fetal D typing early in pregnancy to guide antenatal anti-D prophylaxis(5)

Anti-D Ig if indicated,
to women without Red cell antibody Red cell antibody screen positive
anti-D (consider fetal screen negative antibody specificity
RHD typing (5))

No clinical relevant Anti-D, anti-K or other Other


antibodies anti-Rh antibodies specificities

Blood sampling Antigen typing father


father Consider non-invasive fetal antigen typing D, c, C, E, K
(2)

Fetus or father Fetus or father


antigen negative antigen positive

Laboratory monitoring
Consider differential policy (3)
Consider selected second screen in D-positive -Anti-Rh or anti-K repeated testing
D- women for example c-negative women or risk until critical thresholds are met
Blood sampling -Other specificities: one test in last
Week 27–30 negative factor based (4)
trimester to judge risk for HDFN

Consider (5): Antibody.screening


Above critical thresholds
Fetal RHD typing Antibody.specifity
Clinical monitoring by
Doppler ultrasonography

Fetus D-positive Fetus D-negative Anti-D, -K, - other


–Rh antibodies

Between
week 28–34,
consider Anti-D Ig
week 30 Consider single dose (6)

If mother is Consider: no cord D typing, but use RHD typing child


D-negative: fetal RHD typing result (7)
Cord blood
at birth
Anti-D Ig D-positive D-negative

Check HDFN
- Clinical
- Laboratory work-up,
PAEDIATRICIAN only if necessary

- Follow-up of children
with severe HDFN

Fig. 3 Red cell antibody screening and red cell antigen typing for timely detection of haemolytic disease of the fetus and newborn and to prevent D
alloimmunization. White background: actions taken by health care professionals (gynaecologist, midwife, paediatrician). Gray background: tests
performed by laboratories. Box: action or test; box with rounded angles: test result followed by action or test.

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Haemolytic disease of fetus and newborn 109

routine antenatal and postnatal anti-D prophylaxis, in in 2/2312 (009%) of false-negative test results were
which antenatal anti-D is administered between week 28 obtained upon fetal RHD typing at 26 weeks of gestation
and 34 of gestation, has become the standard care for and that in 17% of cases, anti-D was unnecessarily admin-
D-negative women in many developed countries [94]. Koe- istered. The latter was also calculated by Finning et al.
lewijn et al. [6] showed in 2008 in a nationwide observa- [98], who calculated that in about 21% of cases, anti-D
tional study, involving over 41 000 women, that antenatal prophylaxis would be given unnecessarily because of either
administration of anti-D Ig prophylaxis halves the risk of false-positive (007%; 14/1869) or inconclusive (014%;
anti-D immunization, detected early in the next pregnancy 25/1869) test results.
(RR 046; 95% CI 009–084), and of subsequent HDFN (RR Calculations on cost efficiency of the combined intro-
045; 95% CI 0–108). Crowther et al. [95] reported in a duction of fetal RHD genotyping and antenatal anti-D
systematic review, including two randomized controlled prophylaxis have been published [102–104]. In Denmark
trials (RCT), involving over 4500 women, that the risk of and the Netherlands, fetal RHD genotyping is now rou-
immunization during the current pregnancy (RR 042; 95% tinely performed to target anti-D Ig prophylaxis. Cost-
CI 015–117) until 12 months after birth (RR 030; 95% CI benefit ratios are influenced by costs of tests, which are
010–162) was not significantly reduced by antenatal anti- largely influenced by economy of scale, and costs of
D Ig prophylaxis in the current pregnancy, but that the risk anti-D Ig prophylaxis, the latter differs between countries.
to have a positive Kleihauer test both during pregnancy The earlier in pregnancy fetal RHD genotyping can be
(RR 060; 95% CI 041–088) and within 12 h after birth performed reliably, the more one will benefit, since
(RR 060; 95% CI 046–079) was significantly lower after administration of anti-D given upon possible sensitizing
antenatal administration of anti-D Ig prophylaxis. Also events can also be targeted [105–107]. In the Netherlands,
Turner et al. [96] demonstrated the effect of antenatal anti- the reliability of the fetal RHD genotyping assay has been
D Ig prophylaxis, based on a meta-analysis of these two regarded to be sufficient to discontinue D cord blood typ-
aforementioned RCTs and eight observational studies, ing. Since January 2013, both antenatal and postnatal
adjusted for differences in study design and quality. anti-D Ig administration policy is based on the result of
Despite appropriate prophylaxis D alloimmunization still the fetal RHD genotyping assay. Only in case of twin
occurs [91]. The dosages and timing of antenatal anti-D pregnancies and in case of an inconclusive fetal typing
prophylaxis differ between countries. In theory, a higher result, cord blood D typing will be performed.
dosage or two dosages given at different time points in
pregnancy will lead to higher anti-D levels in pregnant
Summary
woman. However, compliance to a prevention programme
with one single injection of anti-D prophylaxis may be Figure 3 summarizes the considerations one nowadays
higher. In the Netherlands, one injection of 1000 IU can make in the design of a screening programme. It has
(250 lg) of anti-D is administered in week 30 of preg- been shown that an antenatal screening test for red blood
nancy. We analysed in D-alloimmunized women, who were cell antibodies results in timely detection of fetuses at risk
pregnant with their second child, potential risk factors for for a disease that can be fatal or can result in life-long
immunization despite correctly administration of antenatal sequelae. Anti-D, anti-c, anti-K and anti-E, the latter to a
and postnatal anti-D prophylaxis in the first pregnancy. lesser extent, are responsible for the majority of cases
Post-term pregnancy (≥42 weeks), non-spontaneous deliv- with severe HDFN that need treatment with intrauterine
ery, a blood transfusion around delivery, which may be an transfusions or exchange transfusions after birth. Other
indicator of a prolonged third stage of labour, and maternal antibody specificities may be related to an increased risk
age (OR 089/year) at first delivery were all found to be of neonatal icterus demanding phototherapy (e.g. anti-
independent risk factors of D immunization. In post-term Fya) but cause very rarely life threatening or disabling
pregnancy, levels of anti-D prophylaxis may drop below disease. Therefore, intensive laboratory monitoring should
protective levels, other risk factors point to (unexpected) be performed for red cell antibodies directed against Rh
large FMH or (higher) activity of the immune system antigens or antigens of the Kell blood group system and
(maternal age) [91]. can be less frequent in case of other antibody specifici-
Several large-scale feasibility studies showed that non- ties. Laboratory monitoring should be aimed to detect
invasive fetal RHD typing can be used to restrict antenatal high-risk cases, that is cases at high risk for severe HDFN,
anti-D administration to D-negative women carrying a D- for example those with high antibody titres or relevant
positive child (60%) [97–100]. In Denmark and the Nether- biological activity as measured in a functional assay such
lands, fetal RHD typing to target antenatal anti-D prophy- as the ADCC (Fig. 3, consideration 2 and 3). Since red cell
laxis has already been introduced in 2010 and 2011, antibodies causing severe HDFN may not be detected
respectively [97, 101]. Clausen et al. [97] showed that only early in pregnancy, a repeated screening test, early in the

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
110 M. de Haas et al.

third trimester of pregnancy, for example around week sound to identify fetuses in need of intrauterine
30, should be aimed to detect anti-D, anti-c and anti-E. transfusions. Finally, intensive phototherapy is a safe
We produced some evidence that a repeated screening test intervention to sufficiently lower bilirubin levels in order
would suffice solely in those women with an increased to prevent kernicterus. Affected children may need one or
risk to develop maternal alloantibodies, such as D- and c- more transfusions in the first months of life, since the
negative pregnant women (Fig. 3, consideration 1 and 4). maternal antibodies stay active during this period. In this
The last decades, invasive procedures to monitor decade, cost-benefit calculations regarding scenario’s to
fetuses at risk have been abandoned. Fetal blood group introduce non-invasive fetal RHD typing to target admin-
typing can be performed with cell-free fetal DNA present istration of anti-D Ig in pregnancy and to make cord
in maternal plasma during pregnancy (Fig. 3, consider- blood D typing presumably obsolete will be made in
ation 2). Fetal anaemia can be assessed by Doppler ultra- many centres (Fig. 3, consideration 5, 6 and 7).

References
1 Daniels G: Blood group antibodies in of the disease. Prenat Diagn 2010; 18 Han P, Kiruba R, Ong R, et al.: Hae-
haemolytic disease of the fetus and 30:668–673 matolytic disease due to ABO incom-
newborn; in: Hadley A, Soothill P 9 Vaughan JI, Manning M, Warwick patibility: incidence and value of
(eds): Alloimmune Disorders of Preg- RM, et al.: Inhibition of erythroid screening in an Asian population.
nancy. Anaemia, Thrombocytopenia progenitor cells by anti-Kell antibod- Aust Paediatr J 1988; 24:35–38
and Neutropenia in the Fetus and ies in fetal alloimmune anemia. N 19 Bhat YR, Kumar CG: Morbidity of
Newborn. Cambridge, Cambridge Uni- Engl J Med 1998; 338:798–803 ABO haemolytic disease in the new-
versity Press, 2002:21–40 10 Daniels G, Hadley A, Green CA: born. Paediatr Int Child Health 2012;
2 Franklin IM: Prevention of rhesus Causes of fetal anemia in hemolytic 32:93–96
haemolytic disease of the fetus and disease due to anti-K. Transfusion 20 Lin M, Broadberry RE: ABO hemo-
newborn. Lancet 2009; 373:1082 2003; 43:115–116 lytic disease of the newborn is more
3 Moise KJ: Fetal anemia due to non- 11 Heathcote DJ, Carroll TE, Flower RL: severe in Taiwan than in white popu-
Rhesus-D red-cell alloimmunization. Sixty years of antibodies to MNS sys- lations. Vox Sang 1995; 68:136
Semin Fetal Neonatal Med 2008; tem hybrid glycophorins: what have 21 Toy PT, Reid ME, Papenfus L, et al.:
13:207–214 we learned? Transfus Med Rev 2011; Prevalence of ABO maternal-infant
4 Mollison PL, Engelfriet CP, Contreras 25:111–124 incompatibility in Asians, Blacks,
M: Hemolytic disease of the fetus and 12 Dajak S, Stefanovic V, Capkun V: Hispanics and Caucasians. Vox Sang
the newborn; in: Mollison PL, Engelf- Severe hemolytic disease of fetus and 1988; 54:181–183
riet CP, Contreras M (eds): Blood newborn caused by red blood cell 22 Daniels G, Finning K, Martin P, et al.:
Transfusion in Clinical Medicine, antibodies undetected at first-trimes- Noninvasive prenatal diagnosis of
11th edn. Oxford, UK, Oxford Black- ter screening (CME). Transfusion fetal blood group phenotypes: current
well Science, 2005:496–545 2011; 51:1380–1388 practice and future prospects. Prenat
5 Moise KJ: Management of rhesus al- 13 NICE: NICE issues guidance for RhD- Diagn 2009; 29:101–107
loimmunization in pregnancy. Obstet negative women during pregnancy. 23 Daniels G, van der Schoot CE, Olsson
Gynecol 2008; 112:164–176 2002. Report No.: 2002/024 ML: Report of the fourth Interna-
6 Koelewijn JM, de Haas M, Vrijkotte 14 Hackney DN, Knudtson EJ, Rossi KQ, tional Workshop on molecular blood
TG, et al.: One single dose of 200 et al.: Management of pregnancies group genotyping. Vox Sang 2011;
microg of antenatal RhIG halves the complicated by anti-c isoimmuniza- 101:327–332
risk of anti-D immunization and tion. Obstet Gynecol 2004; 103:24–30 24 Finning K, Martin P, Summers J, et al.:
hemolytic disease of the fetus and new- 15 Joy SD, Rossi KQ, Krugh D, et al.: Fetal genotyping for the K (Kell) and
born in the next pregnancy. Transfu- Management of pregnancies compli- Rh C, c, and E blood groups on cell-
sion 2008; 48:1721–1729 cated by anti-E alloimmunization. free fetal DNA in maternal plasma.
7 Koelewijn JM, Vrijkotte TG, van der Obstet Gynecol 2005; 105:24–28 Transfusion 2007; 47:2126–2133
Schoot CE, et al.: Effect of screening 16 Hughes LH, Rossi KQ, Krugh DW, et al.: 25 Legler TJ, Muller SP, Haverkamp A,
for red cell antibodies, other than Management of pregnancies compli- et al.: Prenatal RhD testing: a review
anti-D, to detect hemolytic disease of cated by anti-Fy(a) alloimmunization. of studies published from 2006 to
the fetus and newborn: a population Transfusion 2007; 47:1858–1861 2008. Transfus Med Hemother 2009;
study in the Netherlands. Transfusion 17 Pate LL, Myers JC, Palma JP, et al.: 36:189–198
2008; 48:941–952 Anti-Ge3 causes late-onset hemolytic 26 Muller SP, Bartels I, Stein W, et al.:
8 Illanes S, Soothill P: Management of disease of the newborn: the fourth Cell-free fetal DNA in specimen from
red cell alloimmunisation in preg- case in three hispanic families. Trans- pregnant women is stable up to 5 days.
nancy: the non-invasive monitoring fusion 2013; 53:2152–2157 Prenat Diagn 2011; 31:1300–1304

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Haemolytic disease of fetus and newborn 111

27 Scheffer PG, van der Schoot CE, assay in the management of Rh D al- uterine transfusion for hemolytic
Page-Christiaens GC, et al.: Noninva- loimmunization. Am J Obstet Gynecol disease of the fetus/newborn: the
sive fetal blood group genotyping of 2001; 184:1015–1020 LOTUS study. Am J Obstet Gynecol
rhesus D, c, E and of K in alloimmun- 39 de Haas M, Koelewijn JM, Bilgin K, 2012; 206:141–148
ised pregnant women: evaluation of a et al.: Diagnostic performance of 48 Connan K, Kornman L, Savoia H,
7-year clinical experience. BJOG laboratory monitoring to predict et al.: IVIG – is it the answer? Mater-
2011; 118:1340–1348 severe haemolytic disease of the nal administration of immunoglobu-
28 De Haas M, Finning K, Massey E, fetus and newborn in non-RhD-al- lin for severe fetal red blood cell
Roberts DJ. Anti-D prophylaxis: past, loimmunised pregnancies; in: Koe- alloimmunisation during pregnancy:
present and future. Transfus Med. lewijn JM (ed.): Detection and as case series. Aust N Z J Obstet
2014; 24:1–17 Prevention of Pregnancy Immunisa- Gynaecol 2009; 49:612–618
29 Scheffer PG, de Haas M, van der tion. The OPZI Study. Groningen, 49 Fox C, Martin W, Somerset DA, et al.:
Schoot CE: The controversy about Drukkerij Van Denderen, 2008: Early intraperitoneal transfusion and
controls for fetal blood group geno- 73–85 adjuvant maternal immunoglobulin
typing by cell-free fetal DNA in 40 Alfirevic Z, Sundberg K, Brigham S: therapy in the treatment of severe red
maternal plasma. Curr Opin Hematol Amniocentesis and chorionic villus cell alloimmunization prior to fetal
2011; 18:467–473 sampling for prenatal diagnosis. intravascular transfusion. Fetal Diagn
30 Guidelines for blood grouping and Cochrane Database Syst Rev 2003; Ther 2008; 23:159–163
red cell antibody testing during preg- (3):CD003252 50 Ruma MS, Moise KJ, Kim E, et al.:
nancy. British Committee for Stan- 41 Schonewille H, Klumper FJ, van de Combined plasmapheresis and intra-
dards in Haematology, Blood Watering LM, et al.: High additional venous immune globulin for the
Transfusion Task Force. Transfus Med maternal red cell alloimmunization treatment of severe maternal red cell
1996; 6:71–74 after Rhesus- and K-matched intra- alloimmunization. Am J Obstet Gyne-
31 American College of Obstetricans and uterine intravascular transfusions for col 2007; 196:138.e1–e6
Gynecologists ACOG Practice Bulletin hemolytic disease of the fetus. Am J 51 Maisels MJ, McDonagh AF: Photo-
No. 75: management of alloimmuni- Obstet Gynecol 2007; 196:143–146 therapy for neonatal jaundice. N Engl
zation during pregnancy. Obstet 42 Mari G, Deter RL, Carpenter RL, J Med 2008; 358:920–928
Gynecol 2006; 108:457–464 et al.: Noninvasive diagnosis by 52 American Academy of Pediatrics
32 Moise KJ: Red blood cell alloimmuni- Doppler ultrasonography of fetal ane- Subcommittee on Hyperbilirubinemia
zation in pregnancy. Semin Hematol mia due to maternal red-cell alloim- Management of hyperbilirubinemia in
2005; 42:169–178 munization. Collaborative Group for the newborn infant 35 or more weeks
33 McKenna DS, Nagaraja HN, Doppler Assessment of the Blood of gestation. Pediatrics 2004; 114:
O’Shaughnessy R: Management of Velocity in Anemic Fetuses. N Engl J 297–316
pregnancies complicated by anti-Kell Med 2000; 342:9–14 53 De Boer IP, Zeestraten EC, Lopriore E,
isoimmunization. Obstet Gynecol 43 Oepkes D, Seaward PG, Vandenbus- et al.: Pediatric outcome in Rhesus
1999; 93 (5 Pt 1):667–673 sche FP, et al.: Doppler ultrasonogra- hemolytic disease treated with and
34 Leggat HM, Gibson JM, Barron SL, phy versus amniocentesis to predict without intrauterine transfusion. Am
et al.: Anti-Kell in pregnancy. Br J fetal anemia. N Engl J Med 2006; J Obstet Gynecol 2008; 198:54
Obstet Gynaecol 1991; 98:162–165 355:156–164 54 Rath ME, Smits-Wintjens VE, Linden-
35 van Wamelen DJ, Klumper FJ, de 44 Bettelheim D, Panzer S, Reesink HW, burg IT, et al.: Exchange transfusions
Haas M, et al.: Obstetric history and et al.: Monitoring and treatment of and top-up transfusions in neonates
antibody titer in estimating severity anti-D in pregnancy. Vox Sang 2010; with Kell haemolytic disease com-
of Kell alloimmunization in preg- 99:177–192 pared to Rh D haemolytic disease.
nancy. Obstet Gynecol 2007; 45 van Kamp IL, Klumper FJ, Oepkes D, Vox Sang 2011; 100:312–316
109:1093–1098 et al.: Complications of intrauterine 55 Russel G, Goulden N: Neonatal ther-
36 Hadley AG: Laboratory assays for intravascular transfusion for fetal apy for haemolytic disease of the
predicting the severity of haemolytic anemia due to maternal red-cell al- newborn; in: Hadley A, Soothill P
disease of the fetus and newborn. loimmunization. Am J Obstet Gynecol (eds): Alloimmune Disorders of Preg-
Transpl Immunol 2002; 10:191–198 2005; 192:171–177 nancy. Cambridge, Cambridge Univer-
37 Engelfriet CP, Ouwehand WH: ADCC 46 Weisz B, Rosenbaum O, Chayen B, sity Press, 2000:203–218
and other cellular bioassays for pre- et al.: Outcome of severely anaemic 56 Mundy CA: Intravenous immuno-
dicting the clinical significance of red fetuses treated by intrauterine trans- globulin in the management of
cell alloantibodies. Baillieres Clin fusions. Arch Dis Child Fetal Neona- hemolytic disease of the newborn.
Haematol 1990; 3:321–337 tal Ed 2009; 94:F201–F204 Neonatal Netw 2005; 24:17–24
38 Oepkes D, van Kamp IL, Simon MJ, 47 Lindenburg IT, Smits-Wintjens VE, 57 Smits-Wintjens VE, Walther FJ, Lo-
et al.: Clinical value of an antibody- van Klink JM, et al.: Long-term neu- priore E: Rhesus haemolytic disease
dependent cell-mediated cytotoxicity rodevelopmental outcome after intra- of the newborn: postnatal manage-

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
112 M. de Haas et al.

ment, associated morbidity and long- 69 Gooch A, Parker J, Wray J, et al.: 80 Filbey D, Hanson U, Wesstrom G: The
term outcome. Semin Fetal Neonatal Guideline for blood grouping and prevalence of red cell antibodies in
Med 2008; 13:265–271 antibody testing in pregnancy. Trans- pregnancy correlated to the outcome
58 Rath ME, Smits-Wintjens VE, Linden- fus Med 2007; 17:252–262 of the newborn: a 12 year study in
burg I, et al.: Top-up transfusions in 70 Branch DR, Shabani F, Lund N, et al.: central Sweden. Acta Obstet Gynecol
neonates with Rh hemolytic disease Antenatal administration of Rh- Scand 1995; 74:687–692
in relation to exchange transfusions. immune globulin causes significant 81 Wong KF, Tse KT, Lee AW, et al.: Is
Vox Sang 2010; 99:65–70 increases in the immunomodulatory antenatal antibody screening worth-
59 Mainie P: Is there a role for erythro- cytokines transforming growth fac- while in Chinese? Br J Haematol
poietin in neonatal medicine? Early tor-beta and prostaglandin E2. Trans- 1997; 97:917–919
Hum Dev 2008; 84:525–532 fusion 2006; 46:1316–1322 82 Howard H, Martlew V, McFadyen I,
60 Rath ME, Smits-Wintjens VE, Linden- 71 Brinc D, Le-Tien H, Crow AR, et al.: et al.: Consequences for fetus and
burg IT, et al.: Postnatal outcome in IgG-mediated immunosuppression is neonate of maternal red cell allo-im-
neonates with severe Rhesus c com- not dependent on erythrocyte clear- munisation. Arch Dis Child Fetal
pared to Rhesus D hemolytic disease. ance or immunological evasion: Neonatal Ed 1998; 78:F62–F66
Transfusion 2012; 53:1580–1585 implications for the mechanism of 83 Rothenberg JM, Weirermiller B, Dirig
61 Alcock GS, Liley H: Immunoglobulin action of anti-D in the prevention of K, et al.: Is a third-trimester antibody
infusion for isoimmune haemolytic haemolytic disease of the newborn? screen in Rh+ women necessary? Am
jaundice in neonates. Cochrane Data- Br J Haematol 2007; 139:275–279 J Manag Care 1999; 5:1145–1150
base Syst Rev 2002; (3):CD003313 72 Kumpel BM: On the immunologic 84 Andersen AS, Praetorius L, Jorgensen
62 Gottstein R, Cooke RW: Systematic basis of Rh immune globulin (anti-D) HL, et al.: Prognostic value of screen-
review of intravenous immunoglobu- prophylaxis. Transfusion 2006; ing for irregular antibodies late in
lin in haemolytic disease of the new- 46:1652–1656 pregnancy in rhesus positive women.
born. Arch Dis Child Fetal Neonatal 73 Wilson JMG, Jungner G: Principles Acta Obstet Gynecol Scand 2002;
Ed 2003; 88:F6–F10 and Practice of Screening for Disease. 81:407–411
63 Elalfy MS, Elbarbary NS, Abaza HW: Geneva, World Health Organization; 85 Lee CK, Ma ES, Tang M, et al.: Preva-
Early intravenous immunoglobin Public Health Papers, 1968 lence and specificity of clinically sig-
(two-dose regimen) in the manage- 74 Koelewijn JM, Vrijkotte TG, de Haas nificant red cell alloantibodies in
ment of severe Rh hemolytic disease of M, et al.: Women’s attitude towards Chinese women during pregnancy – a
newborn-a prospective randomized prenatal screening for red blood cell review of cases from 1997 to 2001.
controlled trial. Eur J Pediatr 2011; antibodies, other than RhD. BMC Transfus Med 2003; 13:227–231
170:461–467 Pregnancy Childbirth 2008; 8:49 86 Jovanovic-Srzentic S, Djokic M, Tij-
64 Girish G, Chawla D, Agarwal R, et al.: 75 Hardy J, Napier JA: Red cell antibod- anic N, et al.: Antibodies detected in
Efficacy of two dose regimes of ies detected in antenatal tests on rhe- samples from 21,730 pregnant
intravenous immunoglobulin in Rh sus positive women in South and Mid women. Immunohematology 2003;
hemolytic disease of newborn-a ran- Wales, 1948–1978. Br J Obstet 19:89–92
domized controlled trial. Indian Pedi- Gynaecol 1981; 88:91–100 87 Ameen R, Al-Eyaadi O, Al-Shemmari
atr 2008; 45:653–659 76 Bowell PJ, Brown SE, Dike AE, et al.: S, et al.: Frequency of red blood cell
65 Smits-Wintjens VE, Walther FJ, Rath The significance of anti-c alloimmu- alloantibody in Kuwaiti population.
ME, et al.: Intravenous immunoglob- nization in pregnancy. Br J Obstet Med Princ Pract 2005; 14:230–234
ulin in neonates with rhesus Gynaecol 1986; 93:1044–1048 88 Lurie S, Eliezer E, Piper I, et al.: Is
hemolytic disease: a randomized con- 77 Belfrage P, Thomassen P, Floberg J, antibody screening in Rh (D)-positive
trolled trial. Pediatrics 2011; 127: et al.: Allo-immunization during pregnant women necessary? J Matern
680–686 pregnancy. Clinical results from 1983 Fetal Neonatal Med 2003; 14:404–406
66 Santos MC, Sa C, Gomes SCJ, et al.: to 1989 in a Scandinavian University 89 Adeniji AA, Fuller I, Dale T, et al.:
The efficacy of the use of intravenous Hospital. Acta Obstet Gynecol Scand Should we continue screening rhesus
human immunoglobulin in Brazilian 1992; 71:273–277 D positive women for the develop-
newborns with rhesus hemolytic dis- 78 Heddle NM, Klama L, Frassetto R, ment of atypical antibodies in late
ease: a randomized double-blind trial. et al.: A retrospective study to deter- pregnancy? J Matern Fetal Neonatal
Transfusion 2013; 53:777–782 mine the risk of red cell alloimmuniza- Med 2007; 20:59–61
67 Canadian Blood services: Clinical tion and transfusion during pregnancy. 90 Gottvall T, Filbey D: Alloimmuniza-
Guide to Transfusion. Toronto, Cana- Transfusion 1993; 33:217–220 tion in pregnancy during the years
dian Blood Services, 2007 79 Gottvall T, Selbing A, Hilden JO: 1992–2005 in the central west region
68 Dutch Society of Obstetrics and Evaluation of a new Swedish protocol of Sweden. Acta Obstet Gynecol
Gynaecology (in Dutch: NVOG): for alloimmunization screening dur- Scand 2008; 87:843–848
Erythrocyte Immunisation and Preg- ing pregnancy. Acta Obstet Gynecol 91 Koelewijn JM, de Haas M, Vrijkotte
nancy. NVOG, 2009 Scand 1993; 72:434–438 TG, et al.: Risk factors for RhD im-

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113
Haemolytic disease of fetus and newborn 113

munisation despite antenatal and design and quality. PLoS ONE 2012; and postnatal anti-D. ISBT Sci Ser
postnatal anti-D prophylaxis. BJOG 7:e30711 2012; 7:164–167
2009; 116:1307–1314 97 Clausen FB, Christiansen M, Steffensen 102 Benachi A, Delahaye S, Leticee N,
92 Koelewijn JM, Vrijkotte TG, de Haas R, et al.: Report of the first nationally et al.: Impact of non-invasive fetal
M, et al.: Risk factors for the pres- implemented clinical routine screen- RhD genotyping on management costs
ence of non-rhesus D red blood cell ing for fetal RHD in D- pregnant of rhesus-D negative patients: results
antibodies in pregnancy. BJOG 2009; women to ascertain the requirement of a French pilot study. Eur J Obstet
116:655–664 for antenatal RhD prophylaxis. Trans- Gynecol Reprod Biol 2012; 162:28–32
93 Crowther C, Middleton P: Anti-D fusion 2012; 52:752–758 103 Szczepura A, Bonsel G, Krauth C,
administration after childbirth for 98 Finning K, Martin P, Summers J, et al.: Fetal RHD typing: is fetal RHD
preventing Rhesus alloimmunisation. et al.: Effect of high throughput RHD typing in all RhD negative women
Cochrane Database Syst Rev 2000; typing of fetal DNA in maternal cost effective? BMJ 2008; 336:906
(2):CD000021 plasma on use of anti-RhD immuno- 104 Hawk AF, Chang EY, Shields SM,
94 Engelfriet CP, Reesink HW, Judd WJ, globulin in RhD negative pregnant et al.: Costs and clinical outcomes of
et al.: Current status of immunopro- women: prospective feasibility study. noninvasive fetal RhD typing for tar-
phylaxis with anti-D immunoglobin. BMJ 2008; 336:816–818 geted prophylaxis. Obstet Gynecol
Vox Sang 2003; 85:328–337 99 Muller SP, Bartels I, Stein W, et al.: 2013; 122:579–585
95 Crowther CA, Middleton P, McBain The determination of the fetal D sta- 105 Cardo L, Garcia BP, Alvarez FV: Non-
RD: Anti-D administration in preg- tus from maternal plasma for decision invasive fetal RHD genotyping in the
nancy for preventing Rhesus alloim- making on Rh prophylaxis is feasible. first trimester of pregnancy. Clin Chem
munisation. Cochrane Database Syst Transfusion 2008; 48:2292–2301 Lab Med 2010; 48:1121–1126
Rev 2013; (2):CD000020 100 van der Schoot CE, Soussan AA, Koe- 106 Akolekar R, Finning K, Kuppusamy
96 Turner RM, Lloyd-Jones M, Anumba lewijn J, et al.: Non-invasive antena- R, et al.: Fetal RHD genotyping in
DO, Smith GC, Spiegelhalter DJ, tal RHD typing. Transfus Clin Biol maternal plasma at 11–13 weeks of
Squires H, Stevens JW, Sweeting MJ, 2006; 13:53–57 gestation. Fetal Diagn Ther 2011;
Urbaniak SJ, Webster R, Thompson 101 de Haas M, van der Ploeg CPB, 29:301–306
SG: Routine antenatal anti-D Scheffer PG, Verlinden DA, Hirsch- 107 Wikman AT, Tiblad E, Westgren M:
prophylaxis in women who are berg H, Abbink F, van der Schoot CE: Noninvasive prenatal screening for
Rh(D) negative: meta-analyses A nation-wide fetal RHD screening RHD: the Stockhom study. ISBT Sci
adjusted for differences in study programme for targetted antenatal Ser 2012; 7:168–172

© 2015 International Society of Blood Transfusion


Vox Sanguinis (2015) 109, 99–113

You might also like