Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Warm Autoimmune Hemolytic Anemia: Recent Progress in

Understanding the Immunobiology and the Treatment


Melca M.O. Barros, Morris A. Blajchman, and José O. Bordin

Autoimmune hemolytic anemia (AIHA) is defined as a ameliorating the hemoglobin level with such transfu-
condition associated with the increased destruction of sions particularly in critically ill patients with warm
red blood cells (RBCs) associated with the presence of AIHA. Glucocorticoids are the first-line treatment for
IgG anti-RBC autoantibodies. The etiology underlying patients with warm AIHA resulting in an 80% clinical
the pathogenesis of such autoantibodies is still un- response after 3 weeks of treatment. The latter,
certain. In the present article, we will discuss the however, also may cause adverse events such as
postulated mechanisms that produce a breakdown of excessive weight gain, neuropsychiatric disorders,
immunologic tolerance leading to warm AIHA including endocrine, or cardiovascular events. Splenectomy
the possible roles of RBC autoantigens and the should be considered for patients who do not show a
complement system, the lack of effective presentation satisfactory response to glucocorticoids and may offer
of autoantigens, functional abnormalities of B and T a success rate of up to 70% in patients with idiopathic
cells resulting in polyclonal lymphocyte activation and warm AIHA. Rituximab treatment in patients with
alteration of cytokine production, and the role of refractory warm AIHA has been well tolerated with an
immunoregulatory T cells. Because warm AIHA is a overall median response rate of approximately 60%.
relatively rare clinical entity, current recommended Danazol, intravenous immunoglobulin, alemtuzumab, as
therapeutic strategies for patients with warm AIHA well as other immunosuppressive drugs have also been
are mainly based on results from small cohort studies. successfully used in patients with warm AIHA, refrac-
Clinicians must also balance the risk of withholding tory to glucocorticoids, splenectomy, and rituximab.
RBC transfusions against the possible benefit of © 2010 Elsevier Inc. All rights reserved.

UTOIMMUNE HEMOLYTIC ANEMIA disorder. Idiopathic warm AIHA represents ap-


A (AIHA) is defined as the increased destruc-
tion of red blood cells (RBCs) in the presence of
proximately 50% of reported cases of AIHA,
whereas secondary cases present associated with
anti-RBC autoantibodies.1 Immune hemolytic ane- conditions or agents (microbiological or chemical)
mias have been classified in different ways, but including lymphoproliferative disorders (20%),
typically they are subdivided into 4 major types connective tissue diseases (20%), solid tumors,
including warm AIHA, the cold agglutinin syn- infections, or drugs. Most patients with idiopathic
drome, mixed-type AIHA, and paroxysmal cold warm AIHA are women in their fourth or fifth
hemoglobinuria. Autoimmune hemolytic anemia is decade of life.2,3
uncommon with an annual incidence of about 1 to 3 The diagnosis of warm AIHA requires the
per 100 000 individuals. Autoimmune hemolytic demonstration of clinical and/or laboratory findings
anemia is often classified also by the optimal of hemolysis and serologic evidence of an autoan-
temperature at which the autoantibodies bind to a tibody (usually IgG) to RBCs.2-4 Typically, such
patient's RBCs in vivo. Warm AIHA comprises anemias are macrocytic because of the presence of
80% to 90% of all cases. In warm AIHA, the marked reticulocytosis, but early in the presenta-
autoantibodies demonstrable in the serum react tion, transient reticulocytopenia may occur despite
optimally with human RBCs at 37°C. In patients the presence of a normal or even a hyperplastic
with the other types of AIHA, the AIHA is bone marrow. Reticulocytopenia may be seen
mediated by the presence of cold autoantibodies because of the in vivo hemolysis of the reticulo-
that exhibit affinity for RBCs at temperatures lower cytes or related to compromised marrow function.5
than 37°C.2
Cases of warm AIHA have also been subclassi-
fied by the presence or absence of various From the Universidade Federal São Paulo, São Paulo, Brazil;
pathophysiologically associated underlying condi- and McMaster University, Hamilton, Ontario, Canada (MAB).
tions or diseases. The term primary or idiopathic Address reprint requests to José O. Bordin, MD, Universidade
warm AIHA is applied when there is no recogniz- Federal de São Paulo Rua Botucatu, 740 São Paulo-SP, Brazil.
E-mail: jobordin@hemato.epm.br
able underlying or associated condition present. 0887-7963/$ - see front matter
Secondary warm AIHA can also be seen as a © 2010 Elsevier Inc. All rights reserved.
manifestation of an underlying pathophysiologic doi:10.1016/j.tmrv.2010.03.002

Transfusion Medicine Reviews, Vol 24, No 3 (July), 2010: pp 195-210 195


196 BARROS ET AL

The blood film often reveals important features for patients with warm AIHA are based on results
such as marked anisocytosis and polychromasia from relatively small cohort studies.
reflecting the increased reticulocyte count. Spher-
ocytosis is a usual feature of warm AIHA, THE IMMUNOBIOLOGY AND PATHOGENESIS
particularly in patients with moderate or severe OF WARM AIHA
warm AIHA. Upon presentation, the hemolysis The pathogenesis of warm AIHA is likely a
may be severe and on occasion, may even be life- complex process in which many factors play an
threatening. Typically, patients with AIHA exhibit essential role. The mechanisms that have been
a mild leukocytosis with neutrophilia. Occasional- proposed to explain the spontaneous appearance of
ly, patients present with leukopenia, neutropenia, or AIHA include the role of autoantigens, the role of
thrombocytopenia, the latter often due to associated the complement system, the lack of effective
immune-mediated platelet destruction. presentation of autoantigens, and functional abnor-
The laboratory parameters in patients with malities of B and T cells. Immunologic data that
AIHA, which indicates the presence of hemolysis support these pathogenetic theories on autoimmu-
include a reduced serum haptoglobin level, an nity derive from experimental studies in both
increase in the indirect bilirubin concentration, and experimental animal models as well as from
an elevated lactate dehydrogenase level.2-4 human studies. These postulated mechanisms are
The presence of positive direct antiglobulin tests summarized in Table 1 and discussed in further
(DATs) indicates that RBC autoantibodies and/or detail below.
complement proteins, or both, are bound to the
circulating RBCs in vivo. A polyspecific antiglob- The Role of RBC Autoantigens
ulin reagent that contains antibodies directed Studies in humans have indicated that Rhesus
against human IgG and complement proteins (Rh) polypeptides are the commonest targets for the
(usually C3) is used to screen the patient's RBCs. pathogenic autoantibodies in patients with warm
If positive, monospecific antisera with antibodies to AIHA. Other blood group antigen specificities have
IgG or complement are used to define the pattern of been identified in a minority of patients and include
the DAT. Monospecific antisera to IgM or IgA may the glycophorins, Band 3 proteins, and the RBC
also be used in selected cases. In most cases of anion channel protein. In canine AIHA, the
warm AIHA, no autoantibody specificity is appar- glycophorins are the dominant RBC autoantigen
ent, but in such circumstances, antibodies bound to targets, whereas in mice, the RBC autoantigens are
the patient's RBCs can be eluted off the RBC located on RBC membrane Band 3.6-9 In man,
surface and potentially identified in the eluate several studies have shown that Rh-like proteins are
obtained. Generally, autoantibodies are antigen able to stimulate the in vitro proliferation of
nonspecific and react with all panel RBCs tested. mononuclear cells from peripheral blood or spleen
Occasionally, a broad specificity, usually in the Rh but only in patients with AIHA.10,11 The mecha-
system, will be seen. On occasion, autoantibody nism that has most often been proposed to explain
specificities to other blood group antigen systems the role of autoantigens as a cause of apparently
have been reported. These have included the Kell, spontaneous autoimmune disease is cross-reactivi-
Kidd, Duffy, and Diego blood group systems.1,4 ty, or molecular mimicry, between environmental
The etiology underlying the pathogenesis of antigens and autoantigens. Such cross-reactivity
warm autoantibodies is uncertain. Recent studies in may offer amplified levels of particular antigenic
both humans and animals have been designed to sequences, or greater avidity of antigen recognition,
explain not only immunologic tolerance to self- to overcome self-tolerance. This molecular mimicry
antigens in warm AIHA but also possible mechan- may enhance immunogenicity by innate stimuli.10-12
isms of the increased RBC destruction. In the A recent study with New Zealand Black (NZB)
present article, we review the various mechanisms mice demonstrated that the development of AIHA
that produce a breakdown of immunologic toler- is not dependent on the expression of the major
ance and discuss current treatment options, as well autoantigen and that a defect in self-tolerance is
as some still emerging new therapeutic approaches. directed to the RBC type.13 On the other hand, the
It is important to note particularly with regard to the tendency to RBC autoimmunity in NZB mice may
latter that many recently recommended treatments be focused on Band 3 by weak helper T cell (Th
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 197

Table 1. Postulated Mechanisms of Autoimmunity in AIHA with RBC Rh proteins in a multiprotein complex
I. The role of RBC autoantigens: with many other proteins.14 Experimental studies
1. Rh-related polypeptides are the commonest targets for the have suggested that CD47 functions as a marker of
pathogenic autoantibodies in patients with warm AIHA. self on murine RBCs; thus, CD47 null murine
2. In NZB mice, the tendency to RBC autoimmunity may be
RBCs have been shown to be rapidly eliminated
focused toward RBC Band 3 by weak cross-reactivity
between the T-helper cell dominant epitope and an from the circulation of wild-type mice, by splenic
exogenous antigen. macrophages. This mechanism is based on the
3. CD47 functions as a marker of self on murine RBCs. binding of CD47 RBCs to signal regulatory protein
4. The interaction SIRP-α and CD47 is not responsible for RBC α (SIRP-α) on macrophages resulting in the
clearance in humans.
inhibition of macrophage activation and phagocy-
II. The role of the complement system:
1. Altered levels of expression of CR1 are observed in patients tosis.14,15 The human data available show that the
with autoimmune diseases, AIDS, and lepromatous leprosy. expression of CD47 on RBCs and SIRP-α,β on the
2. CR1/DAF-deficient murine RBCs are eliminated more rapidly monocytes of patients with active warm AIHA in
by complement than by the Fc receptor pathway when the remission are not different from those seen in
RBCs are opsonized with autoantibody.
healthy individuals.16,17 These data thus suggest
3. Patients with autoimmune cytopenias, including warm
AIHA, may have a deficiency of CD59 expression. that signaling by CD47 in man may be qualitatively
III. Lack of effective presentation of autoantigens: different from that seen in mice and imply that the
1. The presence of immature DCs leads to the transient interaction between SIRP-α and CD47 is not
induction of antigen-specific T cell activation followed by responsible for RBC clearance in humans.
T cell deletion and unresponsiveness.
2. Many self-epitopes are inefficiently processed and The Role of the Complement System in the
presented by APCs; thus, self-reactive naive T cells
Maintenance of Self-Tolerance
remain in the peripheral T cell repertoire leading
to autoimmunity. The role of complement in the maintenance of
IV. Functional abnormalities of B and T cells result in: self-tolerance is supported by several lines of
1. Polyclonal lymphocyte activation
evidence. Altered levels of the expression of
(a) Several virus or parasite infections are followed by the
increased production of autoantibodies, which in turn is complement receptor1 (CR1) has been observed
linked to B cell polyclonal activation. in patients with autoimmune disease such as
(b) In chronic graft-vs-host disease, activation of host systemic lupus erythematosus (SLE), AIDS, and
B cells by donor T cells induces autoimmunity, lepromatous leprosy.18 Thus, patients with AIDS
including warm AIHA.
have a progressive decrease of CR1 on their RBCs,
2. Alteration of cytokine production
(a) Prevalence of Th1 cytokines in NZB mice. and genetic studies showed unequivocally that this
(b) IL-4 and IL-10 treatment ameliorates the anemia defect is acquired.19 Data from SLE patients have
in NZB mice. been more difficult to interpret, but genetic studies
(c) Imbalance between IL-10 and IL-12, in NZB mice and indicate an acquired loss of CR1.20 A recent study
humans with AIHA.
in mice indicates that mouse RBCs deficient in CR1
(d) In nonstimulated PBMC cultures from AIHA patients,
there is an in vitro increase of IL-10 and IL-4 production are spontaneously eliminated in vivo by the
with a reduction of IFN-γ and IL-12. complement system.21 These studies demonstrate
(e) PBMCs from patients with AIHA stimulated in vitro with that RBCs in CR1/decay-accelerating factor
RhD protein produce IFN-γ and IL-10 but not IL-4. (DAF)-deficient mice are cleared spontaneously
3. Treg cells have an important role in the maintenance of
by the alternative pathway of complement, and
peripheral tolerance
(a) Autoimmune diseases are associated with abnormalities when opsonized with an immunoglobulin IgG2a
in Treg cell number and/or function. autoantibody, CR1/DAF-deficient mouse RBCs
(b) Older CD25 knockout mice develop AIHA. were eliminated more rapidly by complement than
(c) Treg cells from patients with AIHA are capable by the Fc receptor pathway. These findings suggest
of inhibiting the Th1 effector responses in vitro by
secretion of IL-10.
that CR1 (CD35) and DAF (CD55) influence the
hemolytic pathway in AIHA.22 However, RBC
CD55 expression levels are normal in patients with
cells) cross-reactivity between the helper dominant primary warm AIHA17 as well as in patients with
epitope and an exogenous antigen.13 warm AIHA secondary to SLE.23 On the other
CD47 is a transmembrane glycoprotein (integrin- hand, it has been reported that patients with either
associated protein) that has been closely associated primary or secondary AIHA may have a deficiency
198 BARROS ET AL

of CD59 expression on their RBCs.17,23 CD59 escaping thymic deletion.28 These epitopes would
deficiency also occurs in various conditions thus be generated as a result of the alteration of
characterized by complement activation, such as endosomal enzymes with consequent changes in the
SLE-associated anemia and lymphopenia. In addi- cleavage of proteins during antigen processing or
tion, CD3+ lymphocytes from SLE patients with weak binding to accessible major histocompatibility
lymphopenia show a diminished expression of complex (MHC) molecules that would compete
CD55 and CD59 when compared with lymphocytes ineffectively with other peptides for presentation.12,29
from healthy individuals.24 Relevantly, it has been Relevantly, Liu et al29 demonstrated in mice with
reported that 36% of patients with autoimmune experimentally induced allergic encephalomyelitis
thrombocytopenia have deficient expression of that naive T cells are potentially autoaggressive and
CD59, whereas 16% present with deficient expres- can cause autoimmunity when stimulated.
sion of CD55 compared to that seen in healthy
individuals. Moreover, CD59 deficiency has been Functional Abnormalities of B and T Cells
reported to be associated with severity of thrombo- To identify the immunologic factors contributing
cytopenia in such individuals.25 to autoimmune onset and maintenance, several
murine strains have been studied extensively.
Lack of Effective Presentation of Autoantigens Functional abnormalities of B and T cells in such
One of the hypotheses put forward to explain the mice have been investigated, particularly their
development of autoimmune diseases such as warm lymphocyte activation, cytokine production, and
AIHA is that the clinical entity might be triggered their role in the onset of their autoimmunity.
by changes in the way autoantigens on target cells Playfair and Marshall-Clarke 30 described one
are presented, so that they overcome self-tolerance. murine model of AIHA, in which the alloimmuni-
Normally, antigen-presenting cells (APCs), includ- zation of mice with cross-reacting rat RBCs
ing dendritic cells (DCs) and macrophages, capture resulted in the development of RBC autoantibodies
self-antigens from other cells and present them to as well as rat-specific alloantibodies, suggesting
autoreactive T cells to induce T cell tolerance by that the AIHA may have been provoked by the
either deletion or anergy.26 There are several exposure to foreign antigens that cross-react with
hypotheses to explain the lack of effective presen- self-antigens. In NZB mice, treatment with anti-
tation of autoantigens. One postulate is that the CD4 monoclonal antibody retarded IgG autoanti-
APCs remain relatively immature and therefore body production demonstrating that the generation
tolerate rather than activate self-reactive cells. This of IgG autoantibody is T cell dependent. 31
type of maturation of APCs thus occurs as a Depletion of T cells from such mice prevents the
consequence of signals received by the other cells induction of AIHA in response to immunization by
of the innate immune system, with the presence of rat RBCs.32 In human AIHA, Rh polypeptides,
pathogens leading to the more effective autoantigen including glycophorins, and Band 3 proteins cause
presentation to T cells to induce an immune T cell activation and their proliferation.8-12
response to self-antigens.26 Using antigens targeted
to immature DCs in vivo, Hawiger et al 27 Polyclonal Lymphocyte Activation
demonstrated that in the absence of additional One explanation for the autoaggressive damag-
stimuli, DCs lead to the induction transient antigen- ing tissue responses are the polyclonal activators
specific T cell activation followed by T cell deletion such, as superantigens or mitogens, that are
and subsequent unresponsiveness. However, if the associated with a failure to control lymphoproli-
antigen is targeted to DCs together with anti-CD40 feration. Studies with NZB/W mice thus have
antibodies, which promote APC maturation, there is described excessive B cell proliferation with
prolonged T cell activation and immunity.27 increased amounts of IgM-secreting cells, particu-
A second explanation for the lack of the effective larly in the early life of such animals.33 Thus,
presentation of autoantigens is that many self-epitopes polyclonal activation of B cells has been suggested
may be inefficiently processed and presented by as a possible mechanism by which viruses trigger
APCs. This would result in minimal cross presenta- autoimmune disease. Several virus or parasite
tion and as a result, the self-reactive naive T cells infections are known to be followed by the
would remain as part of the peripheral T cell repertoire increased production of autoantibodies, which is
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 199

linked to B lymphocyte polyclonal activation. levels of the Th1 cytokine IFN-γ but little or no
Thus, after the intracerebral inoculation of the IL-4, IL-5, or IL-10.39 Interleukin-12, a cytokine
lymphocytic choriomeningitis virus, the CH3cB/ produced primarily by APCs, promotes the devel-
FeJ mouse develops AIHA and hypergammaglo- opment of Th1 cells in vitro and in vivo and may
bulinemia.34 Similarly, we have reported that the inhibit IL-4-induced antibody production. Interfer-
prevalence of a positive DAT on the RBCs of HIV- on-γ amplifies the IL-12 activation of Th1 cells and
infected persons was 16.7% (40/239) probably suppresses Th2 cell expansion and promotes B cell
because B cells are polyclonally activated and maturation and pathologic antibody generation.40
secrete large amounts of immunoglobulin that can Mice treated with IFN-γ develop accelerated SLE,
appear on the surface of RBCs.35 However, one of whereas the administration of TNF-α has the
the HIV-positive patients we investigated exhibited opposite effect.41 Interleukin-4 and IL-10 inhibit
reticulocytosis with a decreased hemoglobin level, the development of Th1 cell clones through the
unconjugated hyperbilirubinemia, and sphero- down-regulation of IL-12 production by mono-
cytes.35 We have also investigated whether the cytes.37 In NZB mice, the intranasal administration
RBCs of 67 patients with visceral leishmaniasis of soluble peptide containing the dominant helper
(kala-azar) were coated with IgG and found that the epitope that encourage the autoimmune response
prevalence of a positive DAT before any treatment toward a Th2 profile resulted in the amelioration of
was 32.8%. Interestingly, a positive DAT and disease.42 In support of the hypothesis that Th2
positive enzyme-linked direct antiglobulin test cytokines protect against autoimmune disease, it
correlated with the presence of either rheumatoid has been shown that IL-4 and IL-10 treatment
factor or circulating immune complexes.36 ameliorates the anemia in NZB mice.43 On the
In human chronic graft-vs-host-disease after other hand, minimal IL-10 administration acceler-
bone marrow transplantation, alloreactive donor T ates the onset of the autoimmune syndrome. In
cells that recognize host histocompatibility antigens contrast, the continuous administration of anti–IL-
cause, by direct interaction between T cells and B 10 antibodies delays the onset of autoimmunity in
cells, a selective stimulation of host self-reactive B NZB/W mice, owing to the augmentation of the
cell clones, which produces a wide spectrum of TNF-α level.44
autoantibodies, including those against RBCs that Patients with primary or secondary AIHA not
can give rise to AIHA.37 subjected to immunosuppressive therapy show high
serum levels of IL-1α, IL-2, IL-4, and IL-2R.45
Cytokine Production Peripheral blood mononuclear cells (PBMCs) from
The T cell population comprises distinct subsets patients with SLE have been shown to have a
that are characterized by surface markers of helper decreased production of IFN-γ, TNF-α, IL-12, and
T cells (CD4+, Th) and cytotoxic T cells (CD8+). high levels of IL-10. Decreased production of Th1
Th cells have played an important role in the cytokines in SLE may be secondary to the
development of animal models of autoimmunity. imbalance between IL-10 and IL-12, whereas the
Th cell clones can be classified into different deficiency in IL-12 production may be the effect of
functional types on the basis of the cytokines they excessive IL-10 production because recombinant
secrete. Thus, Th1 cells synthesize interleukin-2 IL-10 strongly inhibits in vitro production of IL-12
(IL-2), interferon-γ (IFN-γ), and tumor necrosis in SLE. Peripheral blood mononuclear cell cultures
factor-β (TNF-β), and promote cell-mediated im- from normal controls and anti-IL10 reverse IL-12
munity, delayed-type hypersensitivity, macrophage deficiency of SLE PBMCs. These data suggest that
activation, and opsonizing antibody production. In IL-10 and IL-12 can potentially build an immuno-
contrast, Th2 cells secrete IL-4, IL-5, IL-6, IL-10, regulatory circuit that can regulate the development
and IL-13 and promote both humoral and allergic and maintenance of an autoimmune disease.46,47
responses.38 The role of the Th1 and Th2 cytokines The AIHA patients have been shown to exhibit
in the pathogenesis of autoimmune diseases is a increased basal synthesis of IL-4 and also have
field of on-going investigation. Thus, T cells from decreased levels of IFN-γ produced by AIHA
NZB mice stimulated with the RBC Band 3 protein PBMCs, compared with control subjects. These
spectrin, from the RBC membrane skeleton and results suggest that there is a basal decrease of Th1
mycobacterial heat-shock protein produce high cytokines and an increase of the Th2 cytokines.
200 BARROS ET AL

Unstimulated PBMCs from AIHA patients have CD4+CD25+ Treg cell subset is distinguished by a
shown a constitutively increase of IL-10 production transcription factor known as the forkhead box
in vitro compared to those from normal controls. protein P3 (FOXP3) and also their expression of
After lipopolysaccharide (LPS) stimulation of CD25. Naturally occurring Treg cells comprise
PBMCs, an increased IL-10 production in both approximately 6% of the CD4+ T cell population in
AIHA patients and controls was observed, compared healthy individuals. These cells seem to be anergic
to basal levels. Furthermore, IL-12 basal levels are to proliferative responses but have the capacity to
higher in AIHA patients than in controls; however, a suppress proliferation of effector T cells in
significant increase in IL-12 production is seen in vitro.51,52 The inducible Treg cells include T-helper
LPS-stimulated PBMC cultures from healthy con- 3 cells, which originate from naive T cells that are
trols but not from those from AIHA patients. The either CD4+ or CD8+, and type 1 Treg (Treg1) cells
latter could be explained as the consequence of an IL- derived from CD4+ precursor cells. These inducible
10 inhibitory effect. These data thus support the Treg cells may or may not express FOXP3. These
hypothesis that the decreased production of Th1 and cells nonetheless can secrete the inhibitory cyto-
the prevalent Th2-type cytokines leading to auto- kines TGF-β or IL-10.51
antibodies production in AIHA may be secondary to Several studies have analyzed the number and/or
the imbalance between IL-10 and IL-12 production function of Treg cells in human autoimmune
by PBMCs.35,48 These observations contrast with diseases providing evidence that autoimmune
results from parallel studies of PBMCs from patients diseases are often associated with abnormalities in
with AIHA stimulated in vitro with a major RBC Treg cells number or function compared to healthy
autoantigen. The RhD protein was thus found to controls.51 In this context, it has been shown that
cause proliferation of Th1 cells that produces IFN-γ CD25 knockout mice, as they age develop
and the regulatory cytokine IL-10 but not IL-4. An autoimmune disease, including hemolytic ane-
analysis of all the samples obtained from a AIHA mia.53 In antibody depletion studies using anti-
patient panel revealed that high IL-10 responses were CD25 antibody, the incidence of AIHA in C57/Bl6
associated with less severe anemia, supporting the mice increased from 30% to 90%. Adoptive transfer
idea that the balance between autoantigen specific Th of purified Treg but not CD4+CD25− cells from
cells subsets secreting different patterns of cytokines immunized mice into naive recipients prevented the
is an important mechanism for regulating autoim- induction of the AIHA. These findings thus
mune responses because Th1 responses appear to be establish a critical role for Treg cells for the control
pathogenic, whereas Th2 responses might mediate of AIHA.54
protection from such disease.49 The peripheral blood of patients with warm
AIHA has been found to contain Treg cells
The Role of Immunoregulatory T Cells specific for the target Rh autoantigens capable of
For several decades, it had been postulated that inhibiting Th1 effector responses in vitro by the
there exists a subset of lymphocytes capable of secretion of IL-10.49 Studies to characterize
suppressing and “managing” the various immune phenotype, origin, and the regulatory function of
responses. Several pieces of evidence have revealed Treg cells in human warm AIHA demonstrated
that active suppressive phenomena, including the that Treg cells shared phenotypic characteristics
control of immune-mediated pathologic conditions, with both natural and inducible Treg cells and
are mediated by the recently defined CD4+ T expressed high levels of CD25 and the lympho-
regulatory (Treg) cells. Treg CD4+ cell populations cyte activation gene-3 when expanding nonspe-
are characterized by the expression of very high cifically, but the Treg cells expressed FOXP3 after
levels of the IL-2 receptor (CD25), and relevantly, activation by the recognized autoantigen. In
these cells have been shown to be CD4+CD25+. addition, it has been shown that Treg cells
These Treg cells have an important role to play in expressed the Th1 signature transcription factor
the generation and maintenance of peripheral T-bet, a transcription factor associated with the
tolerance.50 T regulatory cells are classified accord- Th1 cells phenotype, suggesting that the Treg
ing to their surface phenotype and cytokine cells derive from Th1 cells. These data suggesting
secretion profile and also whether they are naturally the existence of such cells specific for autoanti-
occurring or inducible. The naturally occurring gens of pathogenic relevance in human patients
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 201

show the importance of studying autoreactive heterogeneous patient samples or from single case
Treg cells in the context of known specificity. reports. A proposed algorithm for the treatment of
Analysis of T-bet expression suggests that the warm AIHA is summarized in Figure 1.
Treg cells derive from Th1 cells and may have
arisen as a result of effector T cell stimulation Red Blood Cell Transfusions
associated with the chronic autoimmune response The RBC transfusions often may be necessary in
underlying AIHA.55 the management of patients with warm AIHA,
especially in symptomatic older patients, particu-
TREATMENT OF WARM AIHA larly in patients with coronary artery disease, taking
Because warm AIHA is a rare disease, treatment into account the potential vital risk of the degree of
recommendations for patients with warm AIHA are anemia to their clinical situation. Such patients
often based mainly on results from retrospective should receive RBC transfusions to maintain their
data and/or relatively small cohort studies including hemoglobin levels at a clinically acceptable level, at

Fig 1. Proposed algorithm for the treatment of warm AIHA.


202 BARROS ET AL

least until other treatments come into play that transfusion against the possible benefit of correcting
curtail the degree of hemolysis.56 the hemoglobin level in a seriously ill patient.56
Transfusions of RBCs to patients with warm
AIHA presents a particular set of potential Use of Glucocorticoids as First-Line Therapy
problems. One obvious and often underappreciated First-line treatment of a patient with warm AIHA
issue is the relative short survival time of the usually involves the use of glucocorticoids. Gluco-
transfused RBCs. Experience has demonstrated that corticoids influence hemolysis in patients with warm
when incompatibility is due only to the presence of AIHA by several mechanisms. Patients who respond
an RBC autoantibody, the survival of transfused to glucocorticoids show a decreased strength of the
RBCs is approximately the same as that of the DAT and indirect antiglobulin test, suggesting that
patient's autologous RBCs. The RBC transfusions improved RBC survival results from a decrease in the
can thus be thought to be able to provide only some synthesis of anti-RBC autoantibodies.1,62 On the
temporary benefit.2,56,57 Patients with warm AIHA other hand, this mechanism does not completely
usually have broadly reactive RBC autoantibodies explain the often seen early improvement in
that are generally panagglutinins that can react with hemolysis seen within 24 to 72 hours after the
the alloantigens on all potential donor RBCs. Such onset of glucocorticoid therapy. This is often well
autoantibodies can often mask the presence of RBC before any alteration in strength of the DAT is
alloantibodies that may be present. The RBC observed. Glucocorticoids are also able to suppress
alloantibodies can develop as a result of previous sequestration of RBCs by splenic macrophages. This
transfusions or pregnancies and are the most may be the result of the reduced number of Fcγ
important technical problem faced by the transfu- receptors observed on blood monocytes of AIHA
sion service regarding the RBC transfusion of patients during glucocorticoid therapy.1,63
patients with warm AIHA. This concern relates to Therapy with glucocorticoids is usually started
the fact that these alloantibodies are capable of with oral prednisone, at an initial daily dose of 1 to
causing serious acute hemolytic transfusion reac- 1.5 mg/kg or its equivalent for the first 2 to 4 weeks.
tions in recipients.56 Patients with particularly rapid hemolysis may
Reports of RBC alloantibodies in warm AIHA require intravenous methylprednisolone at a dose
patients range from 15% to 45% (mean, 32%).58-60 of 250 to 1000 mg/d for 1 to 3 days. Once the
These data thus indicate the need for a method to hematocrit begins to rise, the amount of glucocor-
identify such antibodies to prevent alloantibody- ticoid can be slowly reduced, and when a dose of 20
induced acute hemolytic transfusion reactions. to 30 mg daily of prednisone in an adult is achieved,
Selection of the best unit of RBCs to be transfused with a persistent remission, as indicated by the
to patients with warm AIHA patients thus usually stable hemoglobin level and decreasing reticulocyte
involves the need for complex compatibility test count, it is recommended that alternate day
procedures. Numerous approaches are available for prednisone doses be commenced. The advantage
selecting RBC units for transfusion for patients who of an alternate-day schedule is that it is associated
have warm autoantibodies. These include (a) the with fewer corticosteroid long-term side effects.
routine testing of the patient's serum against an Relevantly, the total duration of glucocorticoid
RBC panel; (b) the use of a dilution technique, therapy can vary from 3 to 12 months, after
which will dilute the patient's serum before doing obtaining a remission, is usually required in patients
RBC compatibility testing; (c) the use of adsorption with warm AIHA.1,2,4
procedures (a warm autoadsorption technique or After 3 weeks of treatment with glucocorticoids,
allogeneic adsorption) that removes the autoanti- approximately 80% of all warm AIHA patients have
body but not the alloantibodies from the patient's a clear clinical response (ie, hemoglobin level N 10.0
serum; and (d) extended phenotyping of the g/dL). Patients who do not have a clinical response
patient's RBCs or molecular analyses for RBC at that time and patients who present with recurrent
genotyping the patient and supplying pheno- hemolysis may require second-line treatment, such
typically compatible RBC units.56,60,61 Adequate as a splenectomy or immunosuppression.2-4
testing for alloantibodies may take several hours to Adverse events such as excessive weight gain,
be completed. Thus, in an urgent clinical situation, skin thinning, neuropsychiatric disorders, sleep
the clinician must balance the risk of withholding disturbances, increased risk of cardiovascular
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 203

events, cataracts, and myopathy may occur in autoantibody seen in some patients. Apropos of the
patients taking high doses of glucocorticoids. latter, it has been reported that the levels of RBC-
Glucocorticoid-induced osteoporosis is a potential- bound IgG necessary to maintain a given rate of
ly preventable complication; thus, patients on hemolysis are 6 to 10 times higher in splenecto-
prolonged courses of glucocorticoids should also mized than in nonsplenectomized subjects.1
be given calcium and vitamin D supplementation. In many centers, laparoscopic splenectomy is
Patients at high risk for glucocorticoid-induced the procedure of choice because the surgical risk
osteoporosis should also be considered for treatment of laparoscopic splenectomy is lower than that
with bisphosphonates or other antiosteoporosis seen with conventional surgery, particularly if the
medications, particularly when they are receiving size of the spleen is not extensive. In large series
glucocorticoid therapy. High-dose glucocorticoids of open splenectomies, mortality is approximately
are well-known to also increase the risk of infectious 6%, whereas in recent series reporting on
complications. Other comorbidities that can be laparoscopic splenectomy, the mortality rate is
exacerbated with the use of glucocorticoids include approximately 1.6%.67-69
diabetes, hypertension, hyperlipidemia, heart fail- The most feared complication after splenectomy
ure, glaucoma, and peptic ulcer disease.64 is overwhelming sepsis due to infections with
It is suggested that low-dose folate also be given encapsulated bacteria. This complication is fortu-
to patients with AIHA as long as the warm AIHA nately uncommon but is associated with a high
persists because patients with chronic hemolytic mortality rate and occurs mainly in the first 2 years
anemia often have folate deficiency.2 The use of after splenectomy. This serious risk can be
folic acid will often improve the reticulocyte considerably reduced by the preoperative vaccina-
response particularly in folate deficient patients. tion for Streptococcus pneumoniae, Haemophilus
influenzae type B, and Neisseria meningitidis
Use of Splenectomy serogroup C.1,70 Such vaccinations ideally should
A splenectomy should be considered in patients be done at least 2 weeks before the splenectomy. If
who have not had a satisfactory response to initial not done before surgery, it should be done
corticosteroids, who relapse after having approximately 2 weeks after surgery, with revacci-
responded, or require the equivalent of more than nation recommended, particularly for the pneumo-
10 to 15 mg prednisone per day to maintain an coccus organism every 5 to 6 years. The role and
acceptable hemoglobin level. Although the efficacy efficacy of antibiotic prophylaxis in this regard
of splenectomy for the treatment of warm AIHA remains unclear, and not all investigators recom-
patients has never been compared to that of other mend it.1,70
second-line therapies, splenectomy is probably the
most effective second-line treatment. 1-4,65 The Use of Danazol
results of splenectomy are variable, with a reported Danazol is a synthetic derivative of ethinyl
success rate estimated at 38% to 70% in patients testosterone that has mild androgenic properties.
with idiopathic warm AIHA.2,66-69 Moreover, there The mechanism of action of danazol in the
are no long-term data on remission duration after a treatment of warm AIHA is not completely
splenectomy. In recent studies on patients with understood. Danazol can be used in association
idiopathic warm AIHA or in patients with warm with prednisone as first-line therapy in patients
AIHA secondary to chronic lymphocytic leukemia with warm AIHA, allowing for a shorter duration
(CLL), approximately 70% of patients were of prednisone therapy. The synergistic effects of
reported to have a complete or prolonged response danazol and glucocorticoids have been demon-
with a follow-up of 3 to 20 years.68,69 Patients with strated in 2 studies.71,72 The best results (80%
persistent or recurrent hemolysis after a splenecto- complete response) were observed in patients
my often will require lower doses of glucocorti- receiving a combination of danazol and glucocor-
coids than they had required previously.1 ticoids as first-line therapy. The outcomes have
The beneficial effect of splenectomy may be been much less impressive in relapsed or refrac-
related to several factors. First, splenectomy tory AIHA patients (43% complete response and
removes the primary site of RBC sequestration 14% partial responses).71,72 A retrospective study,
and also decreases the quantity of RBC-bound reported from France, did not observe any
204 BARROS ET AL

reduction in the response rate nor in the reduction deposition, thus, reducing the degree of inflamma-
in the duration of the prednisone therapy required tion and tissue damage.79
in patients treated with danazol.4 Rituximab has a very favorable benefit-to-risk
ratio in the treatment of patients with refractory
Other Immunotherapy Modalities to Treat Patients warm AIHA. Published reports indicate that
With AIHA rituximab (375 mg/m2 weekly for 2-4 weeks) is
effective in treating warm AIHA with an overall
Intravenous immunoglobulin. The treatment of
response rate ranging from 40% to 100% (median,
warm AIHA with intravenous immunoglobulin
60%), with patients of all ages responding. Initial
(IVIG) alone or in combination with prednisone is
studies in pediatric patients reported particularly
controversial, primarily because its use has mainly
promising results, with rates of complete responses
been limited to small case series.73 In a retrospec-
varying from 87% to 100%. 80,84 In adults,
tive study of 73 patients, a response to IVIG was
rituximab shows rates of complete responses
observed in 39.5% of patients.74 Another study
varying from 25% to 100%.85-93 Such results with
showed that a good response occurred in 5 patients
rituximab has been observed both in patients with
with recurrent warm AIHA associated with CLL.
idiopathic warm AIHA and in patients with warm
In this study, the recovery of the hemoglobin level
AIHA secondary to a range of conditions, including
was seen to be faster when prednisone and high-
lymphoproliferative disorders, autoimmune dis-
dose IVIG were combined.75 Published reports of
ease, and also in patients having received bone
IVIG use in warm AIHA suggest that it may be
marrow transplants. The duration of the remission
necessary to use a high dose of IVIG similar to that
achieved has been reported to range from 2 to 95+
used for the treatment of patients with immune
months. The rapidity with which warm AIHA
thrombocytopenia (400-1000 mg/kg per day for 5
patients reached a response to rituximab varied
days). Intravenous immunoglobulin is also recom-
considerably, with some patients responding very
mended for use in patients with warm AIHA
quickly and others taking weeks or even months to
refractory to conventional therapy with corticos-
achieve their maximum response.80-93 Rituximab
teroids. Intravenous immunoglobulin has been
has usually been used in patients with warm AIHA
recommended as a possible temporizing measure
as monotherapy; however, rituximab has also been
before doing a splenectomy.73-77 The mechanism
used in combination with corticosteroids, other
of the IVIG effect in AIHA appears to be related to
immunosuppressive drugs, and IFN-α, particularly
the saturation of Fc phagocyte receptors in
in patients with underlying lymphoproliferative
reticuloendothelial system that is amplified in
disorders.81,88-90 Preliminary data suggest that
AIHA patients.73,77
rituximab retreatment may also be effective in
Rituximab. For the past several years, the AIHA relapsed patients.80,81,84 A summary of the
effectiveness of rituximab has been evaluated in data from the various studies using rituximab for
several autoimmune diseases and in particular in the treatment of warm AIHA is shown in Table 2.
autoimmune hematologic disorders. Rituximab is a Rituximab treatment in patients with warm
monoclonal antibody directed against the CD20 AIHA has been well tolerated, but some adverse
antigen expressed on B cells in the blood, lymph events have been reported including hypotension,
nodes, and bone marrow. The mechanism of action fever, chills, rigors, hypertension, and broncho-
of rituximab in patients with warm AIHA is probably spasm. The occurrence of such side effects seems
through the depletion of the B cells that produce the to be reduced with subsequent rituximab infu-
autoantibodies against the RBCs; however, clinical sions. The prolonged impairment of antibody
experience with rituximab in autoimmune diseases production also increases the risk of viral and
suggests that autoantibody levels are not always bacterial infections; thus, some infectious events
affected by the rituximab treatment.78 This may be have been described related to the use of
explained by that the binding of rituximab to B cells rituximab therapy.80-93
generates immune complexes that may serve as The use of alemtuzumab in warm AIHA.
decoys to attract and bind Fcγ receptors expressed Alemtuzumab is a monoclonal antibody directed
on monoyctes and macrophages, diminishing the against the CD52 antigen expressed on the cell
recruitment of these cells at sites of immune complex membrane of virtually all normal as well as malignant
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 205

Table 2. Reports on the Use of Rituximab for the Treatment of Warm AIHA
Complete Partial No
References Study n Age (y) Treatment protocol Type of AIHA response (%) response (%) response (%)

Quartier et al80 Prospective 6 0.6-2.9 375 mg/m2 weekly Primary 100 – –


for 4 wk
Gupta et al85 Prospective 8 46-70 375 mg/m2 weekly Secondary to CLL 87.5 12.5 –
for 2-5 wk
Shanafelt et al87 Retrospective – 21-79 375 mg/m2 weekly Primary (3) 40 – 60
for 3-8 wk Secondary (2)
Trapè et al86 Prospective 5 44-66 375 mg/m2 weekly Secondary to 60 40 –
for 4 wk lymphoproliferative
disorder
Zaja et al88 Prospective 4 42-84 375 mg/m2 weekly Secondary to CLL 25 – –
for 4 wk
Zecca et al81 Prospective 14 0.3- 375 mg/m2 weekly Primary (10) 71.4 14.3 14.3
13.8 for 2-4 wk Secondary (4)
Narat et al89 Retrospective 11 18-81 375 mg/m2 weekly Primary (2) 27 36 36
for 4 wk Secondary (9)
D'Arena et al90 Retrospective 14 48-87 375 mg/m2 weekly Secondary to CLL 21 50 39
for 3-4 wk
D'Arena et al91 Retrospective 11 23-81 375 mg/m2 weekly Primary 73 27 –
for 4 wk
Rao et al82 Prospective 6 5-17 375 mg/m2 weekly Primary 67 17 –
for 4 wk
Bussone et al92 Retrospective 27 15-81 375 mg/m2 weekly Primary (17) 30 63 7
for 2-4 wk Secondary (10)
Kaufman et al93 Retrospective 20 – 375 mg/m2 weekly Secondary to CLL 100 – –
for 3 wk
Kumar et al83 Retrospective 4 13-16 350-500 mg/m2 Secondary to SLE 100 – –
weekly for 2-4 wk
Svahn et al84 Prospective 4 0.1-0.9 375 mg/m2 weekly Primary (3) 75 25 –
for 4 wk Secondary (1)

lymphocytes, whether they are B or T cells. warm AIHA. Alemtuzumab should therefore be
Alemtuzumab has the ability to cause the lysis of considered even before rituximab, if warm AIHA is
such cells by a host-effector mechanism such as accompanied by progressive CLL in patients also in
complement fixation, antibody-dependent cell-medi- need of cytoreductive therapy.98-100
ated cytotoxicity, and the induction of apoptosis.
Alemtuzumab therapy can result in a prolonged and The Use of Other Immunosuppressive Approaches
profound peripheral blood lymphopenia particularly in Patients With AIHA
affecting T cells.94 Alemtuzumab is very active in the Immunosuppressive drugs may also be indicated
treatment of B-CLL, but the experience with the use mainly in patients refractory to prednisone, sple-
of alemtuzumab in patients with warm AIHA has nectomy, and rituximab, or for those patients
been very limited. Preliminary results indicate that resistant to these therapies and who may be unfit
alemtuzumab may induce responses in patients with to undergo splenectomy. Cytotoxic drugs such as
severe warm AIHA, who failed to respond to azathioprine, cyclophosphamide, cyclosporine,
conventional immunosuppression.95-97 Interestingly, mycophenolate mofetil (MMF) have been used to
one patient who was refractory to rituximab treat refractory cases of warm AIHA in an attempt
responded to alemtuzumab.97 In small series of to suppress autoantibody synthesis. Although
patients with warm AIHA secondary to CLL and authentic successes have been reported with the
refractory to conventional therapy such as corticos- use of such agents, their relative effectiveness
teroids, rituximab, or splenectomy, complete or compared to other treatments is unclear.1-4
partial responses were achieved in all treated patients, Cyclophosphamide and Azathioprine. Many
suggesting that alemtuzumab may be highly effective patients with refractory warm AIHA have been
in the treatment of severe, resistant B-CLL secondary treated with small oral doses of cyclophosphamide
206 BARROS ET AL

(100 mg/d) or azathioprine (100-150 mg/d). Most of cells. Mycophenolate mofetil potently suppresses
the reported data indicate an efficacy of 25% to 40% lymphocyte proliferation and has been successfully
in such patients. Such treatments should be used in organ transplantation to prevent graft
continued for up to 6 months before being rejection. There has been only limited data reported
considered as failed because the response may be with the use of MMF in patients with refractory
somewhat slow in coming.1-4,65 In one small series, warm AIHA. Most of the patients receiving MMF
high-dose cyclophosphamide (50 mg/kg per day for have been reported to achieve partial responses, and
4 days) was used in 8 patients with highly refractory continuous treatment was required to maintain the
warm AIHA. Five patients achieved a complete response.106-108 Mycophenolate mofetil has been
response, and 3 achieved a partial response, not shown to be particularly effective in the treatment
requiring RBC transfusion support. No relapses of the refractory autoimmune cytopenias in children
were seen after a median follow-up of 15 months with the autoimmune lymphoproliferative syn-
(range, 4-29 months); however, most of these drome. In a study of 13 autoimmune lymphopro-
patients had prolonged cytopenias requiring granu- liferative syndrome patients with warm AIHA, it
locyte colony-stimulating factor support. The use of was shown that 12 responded to MMF with
high-dose cyclophosphamide was associated with maintenance of an adequate blood count and/or
nonfatal adverse effects including nausea, vomiting, reduction, or cessation, of the need for other
transient alopecia, and neutropenic fever.101 immunosuppressive drugs. The median follow-up
Vincristine-Loaded Platelets. One old but not time in responders was 49 weeks. The MMF
forgotten treatment modality available for the treatment was well tolerated in all patients.109
treatment of patients with warm AIHA is the
infusion of vincristine-loaded platelets. Recently, Hematopoietic Stem Cell Transplantation in
this form of treatment was used in 3 refractory warm Warm AIHA
AIHA patients and in 3 subjects with refractory There is very limited information on the use of
Evan syndrome. All 6 patients with both immune hematopoietic stem cell transplantation (HSCT) in
thrombocytopenia and AIHA initially achieved a patients with warm AIHA. In 2008, the registry of
complete response, but 4 patients subsequently the European Group of Blood and Marrow
relapsed. Two other patients achieved remissions Transplantation reported data on 65 transplants
lasting for 8+ and 9+ years, respectively.102 (37 autologous and 28 alogeneic) in 59 patients
Cyclosporin A. Cyclosporin A (CsA) has been including subjects with AIHA, Evan syndrome,
used effectively in a few patients with refractory idiopathic thrombocytopenic purpura, pure red cell
warm AIHA. In a comparative study to test the aplasia, pure white cell aplasia, or thrombotic
effectiveness of CsA in the treatment of warm thrombocytopenic purpura. A complete response
AIHA and Evan syndrome, a high response rate was achieved in 2 of 2 patients with warm AIHA
(89%) was seen in 18 patients treated with a subjected to allogeneic HSCT, whereas a partial
combination consisting of CsA, prednisone, and response was seen in only 1 of 5 patients treated
danazol. In contrast, in 26 patients treated with only with an autologous HSCT.110
prednisone and danazol, the complete response rate
was only 57.7%. In addition, the patients who Therapeutic Plasma Exchange in AIHA Patients
received CsA had a lower relapse rate compared to The role of therapeutic plasma exchange (TPE)
those who did not (3.3% vs 70%, respectively).103 in patients with warm AIHA remains unclear. The
In another report, complete responses were ob- indication for TPE in warm AIHA is considered as
served in 3 of 3 patients with refractory warm a category III intervention (may be beneficial, but
AIHA treated with CsA.104 A good response was insufficient evidence exists to establish efficacy) by
also reported in 56% of patients with warm AIHA the American Society for Apheresis. Therapeutic
associated with CLL treated with CsA.105 plasma exchange has been used sporadically in
Mycophenolate mofetil. Mycophenolate mofe- patients with severe cold and warm AIHA for many
til is the prodrug of mycophenolic acid that inhibits years in an attempt to remove circulating auto-
inosine monophosphate dehydrogenase, a rate- antibodies. Most of the published data have
limiting enzyme acting on the de novo synthesis indicated reductions in the autoantibody titer, but
of guanosine nucleotides, especially in T and B clinical responsiveness has been inconsistent. Some
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 207

authors believe that TPE increases the efficiency of case-control study in 5 patients indicates that the
RBC transfusions in patients with fulminant use of TPE before RBC transfusions does not
hemolysis.111 In contrast, a recent retrospective increase the efficiency of the RBC transfusions.112

REFERENCES
1. Packman CH: Hemolytic anemia due to warm auto- 18. Yazdanbakhsh K: Controlling the complement system
antibodies. Blood Rev 22:17-31, 2008 for prevention of red cell destruction. Curr Opin Hematol
2. Petz LD, Garratty G: Immune hemolytic anemia. ed 2. 12:117-122, 2005
Philadelphia, PA, Elsevier Inc; 2004 19. Cohen JH, Geffriaud C, Caudwell V, et al: Genetic
3. Gehrs BC, Friedberg RC: Autoimmune hemolytic analysis of CR1 (the C3b complement receptor, CD35)
anemia. Am J Hematol 69:258-271, 2002 expression on erythrocytes of HIV-infected individuals. AIDS
4. Genty I, Michel M, Hermine O, et al: Caractéristiques 3:397-399, 1989
des anémies hémolytiques auto-immunes de l'adulte: Analyse 20. Birmingham DJ: Erythrocyte complement receptors.
rétrospective d'une série de 83 patients. Rev Méd Interne Crit Rev Immunol 15:133-154, 1995
23:901-909, 2002 21. Miwa T, Zhou L, Hilliard B, et al: Crry, but not CD59
5. Conley CL, Lippman SM, Ness P: Autoimmune and DAF, is indispensable for murine erythrocyte protection in
hemolytic anemia with reticulocytopenia. A medical emergency. vivo from spontaneous complement attack. Blood 99:3707-3716,
JAMA 244:1688-1690, 1980 2002
6. Barker RN, Casswell KM, Reid ME, et al: Identification 22. Molina H, Miwa T, Zhou L, et al: Complement-
of autoantigens in autoimmune haemolytic anaemia by a non- mediated clearance of erythrocytes: Mechanism and delineation
radioisotope immunoprecipitation method. Br J Haematol of regulatory roles of Crry and DAF. Blood 100:4544-4549,
82:126-132, 1992 2002
7. Barker RN, Elson CJ: Red blood cell glycophorins as B 23. Richaud-Patin Y, Pérez-Romano B, Carrillo-Maravilla
and T-cell antigens in canine autoimmune haemolytic anaemia. E, et al: Deficiency of red cell bound CD55 and CD59 in patients
Vet Immunol Immunopathol 47:225-238, 1995 with systemic lupus erythematosus. Immunol Letters 88:95-99,
8. Barker RN, Casswell KM, Elson CJ: Identification of 2003
murine erythrocyte autoantigens and cross-reactive rat antigens. 24. García-Valladares I, Atisha-Fregoso Y, Richaud-Patin Y,
Immunology 78:568-573, 1993 et al: Diminished expression of complement regulatory proteins
9. Leddy JP, Falany JL, Kissel GE, et al: Erythrocyte (CD55 and CD59) in lymphocytes from systemic lupus erythe-
membrane proteins reactive with human (warm-reacting) matosus patients with lymphopenia. Lupus 15:600-605, 2006
anti-red cell autoantibodies. J Clin Invest 91:1672-1680, 25. Ruiz-Argüelles A, Llorente L: The role of complement
1993 regulatory proteins (CD55 and CD59) in the pathogenesis of
10. Barker RN, Elson CJ: Multiple self epitopes on the autoimmune hemocytopenias. Autoimmun Rev 6:155-161,
Rhesus polypeptides stimulate immunologically ignorant human 2007
T cells in vitro. Eur J Immunol 24:1578-1582, 1994 26. Ohashi PS, DeFranco AL: Making and breaking
11. Barker RN, Hall AM, Standen GR, et al: Identification tolerance. Curr Opin Immunol 14:744-759, 2002
of T-cell epitopes on the Rhesus polypeptides in autoimmune 27. Hawiger D, Inaba K, Dorsett Y, et al: Dendritic cells
hemolytic anemia. Blood 90:2701-2715, 1997 induce peripheral T cell unresponsiveness under steady state
12. Elson CJ, Barker RN, Thompson SJ, et al: Immunolog- conditions in vivo. J Exp Med 194:769-779, 2001
ically ignorant autoreactive T cells, epitope spreading and 28. Barker RN, Vickers MA, Ward FJ: Controlling
repertoire limitation. Immunol Today 16:71-76, 1995 autoimmunity-Lessons from the study of red blood cells as
13. Hall AM, Ward FJ, Shen CR, et al: Deletion of the model antigens. Immunol Lett 108:20-26, 2007
dominant autoantigen in NZB mice with autoimmune hemolytic 29. Liu GY, Fairchild PJ, Smith RM, et al: Low avidity
anemia: Effects on autoantibody and T-helper responses. Blood recognition of self-antigen by T cells permits escape from central
110:4511-4517, 2007 tolerance. Immunity 3:407-415, 1995
14. Oldenborg PA, Zheleznyak A, Fang YF, et al: Role of 30. Playfair JHL, Marshall-Clarke S: Induction of red cell
CD47 as a marker of self on red blood cells. Science autoantibodies in normal mice. Nature New Biol 243:213-214,
288:2051-2054, 2000 1973
15. Oldenborg PA, Gresham HD, Chen Y, et al: Lethal 31. Oliveira GG, Hutchings PR, Roitt IM, et al: Production
autoimmune hemolytic anemia in CD47-deficient nonobese of erythrocyte autoantibodies in NZB mice is inhibited by CD4
diabetic (NOD) mice. Blood 99:3500-3504, 2002 antibodies. Clin Exp Immunol 96:297-302, 1994
16. Ahrens N, Pagenkopf C, Kiesewetter H, et al: CD47 is 32. Naysmith JD, Ortega-Pierres MG, Elson CJ: Rat
expressed at normal levels in patients with autoimmune erythrocyte-induced antierythrocyte autoantibody production
haemolytic anaemia and/or immune thrombocytopenia. Transfus and control in normal mice. Immunol Rev 55:55-87, 1981
Med 16:397-402, 2006 33. Theophilopoulos AN, Dixon FJ: Murine models of
17. Barros MM, Yamamoto M, Figueiredo MS, et al: systemic lupus erythematosus. Adv Immunol 37:269-273, 1985
Expression levels of CD47, CD35, CD55, and CD59 on red 34. Musaji A, Meite M, Detalle L, et al: Enhancement of
blood cells and signal-regulatory protein-alpha,beta on mono- autoantibody pathogenicity by viral infections in mouse models
cytes from patients with warm autoimmune hemolytic anemia. of anemia and thrombocytopenia. Autoimmun Rev 4:247-252,
Transfusion 49:154-160, 2009 2005
208 BARROS ET AL

35. Bordin JO, Kerbauy J, Souza-Pinto JC, et al: Quantita- 53. Willerford DM, Chen J, Ferry JA, et al: Interleukin-
tion of red cell-bound IgG by an enzyme-linked antiglobulin test 2 receptor alpha chain regulates the size and content of the
in human immunodeficiency virus-infected persons. Transfusion peripheral lymphoid compartment. Immunity 3:521-530,
32:426-429, 1992 1995
36. Vilela RB, Bordin JO, Chiba AK, et al: RBC-associated 54. Mqadmi A, Zheng X, Yazdanbakhsh K: CD4+CD25+
IgG in patients with visceral leishmaniasis (kala-azar): A regulatory T cells control induction of autoimmune hemolytic
prospective analysis. Transfusion 42:1442-1447, 2002 anemia. Blood 105:3746-3748, 2005
37. Toriani-Terenzi C, Fagiolo E: IL-10 and the cytokine 55. Ward FJ, Hall AM, Cairns LS, et al: Clonal regulatory T
network in the pathogenesis of human autoimmune hemolytic cells specific for a red blood cell autoantigen in human
anemia. Ann N Y Acad Sci 1051:29-44, 2005 autoimmune hemolytic anemia. Blood 111:680-687, 2008
38. Mosmann TR, Coffman RL: TH1 and TH2 cells: 56. Petz LD: A physician's guide to transfusion in autoim-
Different patterns of lymphokine secretion lead to different mune haemolytic anaemia. Br J Haematol 124:712-716, 2004
functional properties. Annu Rev Immunol 7:145-173, 1989 57. Salama A, Berghöfer H, Mueller-Eckhardt C: Red blood
39. Shen CR, Mazza G, Perry FE, et al: T-helper 1 cell transfusion in warm-type autoimmune haemolytic anaemia.
dominated responses to erythrocyte Band 3 in NZB mice. Lancet 340:1515-1517, 1992
Immunology 89:195-199, 1996 58. Branch DR, Petz LD: Detecting alloantibodies in
40. Adorini L: The pathogenic role of IL12 in TH1- patients with autoantibodies. Transfusion 39:6-10
mediated autoimmune diseases. Immunol Immunopharm 59. Wallhermfechtel MA, Pohl BA, Chaplin H: Alloimmu-
20:29-35, 2000 nization in patients with warm autoantibodies. A retrospective
41. Jacob CO, van der Meide PH, McDevitt HO: In vivo study employing three donor alloabsorptions to aid in antibody
treatment of (NZB × NZW)F1 lupus-like nephritis with detection. Transfusion 24:482-485, 1984
monoclonal antibody to gamma interferon. J Exp Med 60. Shirey RS, Boyd JS, Parwani AV, et al: Prophylactic
166:798-803, 1987 antigen-matched donor blood for patients with warm autoanti-
42. Shen CR, Youssef AR, Devine A, et al: Peptides bodies: an algorithm for transfusion management. Transfusion
containing a dominant T-cell epitope from red cell band 3 have in 42:1435-1441, 2002
vivo immunomodulatory properties in NZB mice with autoim- 61. Reid ME: Overview of molecular methods in immuno-
mune hemolytic anemia. Blood 102:3800-3806, 2003 hematology. Transfusion 47:10S-16S, 2007
43. Youssef AR, Shen CR, Lin CL, et al: IL-4 and IL-10 62. Rosse WF: Quantitative immunology of immune
modulate autoimmune haemolytic anaemia in NZB mice. Clin hemolytic anemia: II. The relationship of cell-bound antibody
Exp Immunol 139:84-89, 2005 to hemolysis and the effect of treatment. J Clin Invest
44. Ishida H, Munchamuel T, Sakaguchi S, et al: Contin- 50:734-743, 1971
uous administration of anti-IL10 antibodies delay onset of 63. Fries LF, Brickman CM, Frank MM: Monocyte
autoimmunity in NZB/W F1 mice. J Exp Med 179:305-310, receptors for the Fc portion of IgG increase in number in
1994 autoimmune hemolytic anemia and other hemolytic states and
45. Fagiolo E, Vigevani F, Pozzetto U: High cytokine serum are decreased by glucocorticoid therapy. J Immunol
levels in patients with autoimmune hemolytic anemia (AIHA). 131:1240-1245, 1983
Immunol Invest 23:449-456, 1994 64. McDonough AK, Curtis JR, Saag KG: The epidemiol-
46. Liu TF, Jones BM: Impaired production of IL-12 in ogy of glucocorticoid-associated adverse events. Curr Opin
systemic lupus erythematosus. I. Excessive production of IL-10 Rheumatol 20:131-137, 2008
suppresses production of IL-12 by monocytes. Cytokine 65. Valent P, Lechner K: Diagnosis and treatment of
10:140-147, 1998 autoimmune haemolytic anaemias in adults: A clinical review.
47. Horwitz DA, Gray JD, Behrendsen SC, et al: Decreased Wien Klin Wochenschr 120:136-151, 2008
production of interleukin-12 and other Th1-type cytokines in 66. Coon WW: Splenectomy in the treatment of hemolytic
patients with recent-onset systemic lupus erythematosus. Arthri- anemia. Arch Surg 120:625-628, 1985
tis Rheum 41:838-844, 1998 67. Hill J, Walsh RM, McHam S, et al: Laparoscopic
48. Fagiolo E, Toriani-Terenzi C: Th1 and Th2 cytokine splenectomy for autoimmune hemolytic anemia in patients with
modulation by IL-10/IL-12 imbalance in autoimmune haemoly- chronic lymphocytic leukemia: A case series and review of the
tic anaemia (AIHA). Autoimmunity 35:39-44, 2002 literature. Am J Hematol 75:134-138, 2004
49. Hall AM, Ward FJ, Vickers MA, et al: Interleukin-10- 68. Pomp A, Gagner M, Salky B, et al: Laparoscopic
mediated regulatory T-cell responses to epitopes on a human red splenectomy: A selected retrospective review. Surg Laparosc
blood cell autoantigen. Blood 100:4529-4536, 2002 Endosc Percutan Tech 15:139-143, 2005
50. Sakaguchi S, Sakaguchi N, Asano M, et al: Immuno- 69. Balagué C, Targarona EM, Cerdán G, et al: Long-term
logic self-tolerance maintained by activated T cells expressing outcome after laparoscopic splenectomy related to hematologic
IL-2 receptor alpha-chains (CD25). Breakdown of a single diagnosis. Surg Endosc 18:1283-1287, 2004
mechanism of self-tolerance causes various autoimmune dis- 70. Newland A, Provan D, Myint S: Preventing severe
eases. J Immunol 155:1151-1164, 1995 infection after splenectomy. BMJ 331:417-418, 2005
51. Torgerson TR: Regulatory T cells in human autoim- 71. Pignon JM, Poirson E, Rochant H: Danazol in
mune diseases. Springer Semin Immunopathol 28:63-76, 2006 autoimmune haemolytic anaemia. Br J Haematol 83:343-345,
52. Hsieh CS, Zheng Y, Liang Y, et al: An intersection 1993
between the self-reactive regulatory and nonregulatory T cell 72. Ahn YS: Efficacy of danazol in hematologic disorders.
receptor repertoires. Nat Immunol 7:401-410, 2006 Acta Haematol 84:122-129, 1990
WARM AIHA: IMMUNOBIOLOGY AND TREATMENT 209

73. Darabi K, Abdel-Wahab O, Dzik WH: Current usage of 91. D'Arena G, Califano C, Annunziata M, et al: Rituximab for
intravenous immune globulin and the rationale behind it: The warm-type idiopathic autoimmune hemolytic anemia: A retrospec-
Massachusetts General Hospital data and a review of the tive study of 11 adult patients. Eur J Haematol 79:53-58, 2007
literature. Transfusion 46:741-753, 2006 92. Bussone G, Ribeiro E, Dechartres A, et al: Efficacy and
74. Flores G, Cunningham-Rundles C, Newland AC, et al: safety of rituximab in adults' warm antibody autoimmune
Efficacy of intravenous immunoglobulin in the treatment of haemolytic anemia: Retrospective analysis of 27 cases. Am J
autoimmune hemolytic anemia: Results in 73 patients. Am J Hematol 84:153-157, 2009
Hematol 44:237-242, 1993 93. Kaufman M, Limaye SA, Driscoll N, et al: A
75. Besa EC: Rapid transient reversal of anemia and long- combination of rituximab, cyclophosphamide and dexametha-
term effects of maintenance intravenous immunoglobulin for sone effectively treats immune cytopenias of chronic lympho-
autoimmune hemolytic anemia in patients with lymphoproli- cytic leukemia. Leuk Lymphoma 23:1-8, 2009
ferative disorders. Am J Med 84:691-698, 1988 94. Robak T: Monoclonal antibodies in the treatment of
76. Bussel JB, Cunningham-Rundles C, Abraham C: autoimmune cytopenias. Eur J Haematol 72:79-88, 2004
Intravenous treatment of autoimmune hemolytic anemia with 95. Willis F, Marsh JC, Bevan DH, et al: The effect of
very high dose gammaglobulin. Vox Sang 51:264-269, 1986 treatment with Campath-1H in patients with autoimmune
77. Majer RV, Hyde RD: High-dose intravenous immuno- cytopenias. Br J Haematol 114:891-898, 2001
globulin in the treatment of autoimmune haemolytic anaemia. 96. Marsh JC, Gordon-Smith EC: CAMPATH-1H in the
Clin Lab Haematol 10:391-395, 1988 treatment of autoimmune cytopenias. Cytotherapy 3:189-195, 2001
78. Garvey B: Rituximab in the treatment of autoimmune 97. Cheung WW, Hwang GY, Tse E, et al: Alemtuzumab
haematological disorders. Br J Haematol 141:149-169, 2008 induced complete remission of autoimmune hemolytic anemia
79. Taylor RP, Lindorfer MA: Immunotherapeutic mechan- refractory to corticosteroids, splenectomy and rituximab. Hae-
isms of anti-CD20 monoclonal antibodies. Curr Opin Immunol matologica 91:ECR13
20:444-449, 2008 98. Karlsson C, Hansson L, Celsing F, et al: Treatment of
80. Quartier P, Brethon B, Philippet P, et al: Treatment of severe refractory autoimmune hemolytic anemia in B-cell
childhood autimmune haemolytic anemia with rituximab. Lancet chronic lymphocytic leukemia with alemtuzumab (humanized
358:1511-1513, 2001 CD52 monoclonal antibody). Leukemia 21:511-514, 2007
81. Zecca M, Nobili B, Ramenghi U, et al: Rituximab for the 99. Laurenti L, Tarnani M, Efremov DG, et al: Efficacy and
treatment of refractory autoimmune hemolytic anemia in safety of low-dose alemtuzumab as treatment of autoimmune
children. Blood 101:3857-3861, 2003 hemolytic anemia in pretreated B-cell chronic lymphocytic
82. Rao A, Kelly M, Musselman M, et al: Safety, efficacy, leukemia. Leukemia 21:1819-1821, 2007
and immune reconstitution after rituximab therapy in pediatric 100. Royer B, Vaida I, Etienne A, et al: Treatment of severe
patients with chronic or refractory hematologic autoimmune autoimmune hemolytic anemia in B-cell chronic lymphocytic
cytopenias. Pediatr Blood Cancer 50:822-825, 2008 leukemia with alemtuzumab. Leukemia 21:1841-1842, 2007
83. Kumar S, Benseler SM, Kirby-Allen M, et al: B-cell 101. Moyo VM, Smith D, Brodsky I, et al: High-dose
depletion for autoimmune thrombocytopenia and autoimmune cyclophosphamide for refractory autoimmune hemolytic anemia.
hemolytic anemia in pediatric systemic lupus erythematosus. Blood 100:704-706, 2002
Pediatrics123 :159-163, 2009 102. Shvidel L, Sigler E, Shtalrid M, et al: Vincristine-loaded
84. Svahn J, Fioredda F, Calvillo M, et al: Rituximab-based platelet infusion for treatment of refractory autoimmune
immunosuppression for autoimmune haemolytic anaemia in hemolytic anemia and chronic immune thrombocytopenia:
infants. Br J Haematol 145:96-100 Rethinking old cures. Am J Hematol 81:423-425, 2006
85. Gupta N, Kavuru S, Patel D, et al: Rituximab-based 103. Liu H, Shao Z, Jing L: The effectiveness of
chemotherapy for steroid refractory autoimmune hemolytic cyclosporine A in the treatment of autoimmune hemolytic
anemia of chronic lymphocytic leukemia. Leuk 16:2092-2095, anemia and Evans syndrome. Zhonghua Xue Ye Xue Za Zhi
2002 22:581-583, 2001
86. Trapè G, Fianchi L, Lai M, et al: Rituximab chimeric 104. Emilia G, Messora C, Longo G, et al: Long-term salvage
anti-CD20 monoclonal antibody treatment for refractory hemo- treatment by cyclosporin in refractory autoimmune haematolo-
lytic anemia in patients with lymphoproliferative disorders. gical disorders. Br J Haematol 93:341-344, 1996
Haematologica 88:223-225, 2003 105. Cortes J, O'Brien S, Loscertales J, et al: Cyclosporin A
87. Shanafelt TD, Madueme HL, Wolf RC, et al: Rituximab for the treatment of cytopenia associated with chronic lympho-
for immune cytopenia in adults: Idiopathic thrombocytopenic cytic leukemia. Cancer 92:2016-2022, 2001
purpura, autoimmune hemolytic anemia, and Evans syndrome. 106. Zimmer-Molsberger B, Knauf W, Thiel E: Mycophe-
Mayo Clinic Proc 78:1340-1346, 2003 nolate mofetil for severe autoimmune haemolytic anemia. Lancet
88. Zaja F, Vianelli N, Sperotto A, et al: Anti-CD20 therapy 350:1003-1004, 1997
for chronic lymphocytic leukemia-associated autoimmune dis- 107. Howard J, Hoffbrand AV, Prentice HG, et al: Myco-
eases. Leuk Lymphoma 44:1951-1955, 2003 phenolate mofetil for the treatment of refractory auto-immune
89. Narat S, Gandla J, Hoffbrand AV, et al: Rituximab in the haemolytic anaemia and auto-immune thrombocytopenia pur-
treatment of refractory autoimmunecytopenias in adults. Hae- pura. Br J Haematol 117:712-715, 2002
matologica 90:1273-1274, 2005 108. Kotb R, Pinganaud C, Trichet C, et al: Efficacy of
90. D'Arena G, Laurenti L, Capalbo S, et al: Rituximab mycophenolate mofetil in adult refractory auto-immune cytope-
therapy for chronic lymphocytic leukemia-associated autoim- nias: A single center preliminary study. Eur J Haematol
mune hemolytic anemia. Am J Hematol 81:598-602, 2006 75:60-64, 2005
210 BARROS ET AL

109. Rao VK, Dugan F, Dale JK, et al: Use of mycophenolate 111. McLeod BC: Evidence based therapeutic apheresis in
mofetil for chronic, refractory immune cytopenias in children autoimmune and other hemolytic anemias. Curr Opin Hematol
with autoimmune lymphoproliferative syndrome. Br J Haematol 14:647-654, 2007
129:534-538, 2005 112. Ruivard M, Tournilhac O, Montel S, et al: Plasma
110. Passweg JR, Rabusin M: Hematopoetic stem cell transplan- exchanges do not increase red blood cell transfusion efficiency in
tation for immune thrombocytopenia and other refractory autoim- severe autoimmune hemolytic anemia: A retrospective case-
mune cytopenias. Autoimmunity 41:660-665, 2008 control study. J Clin Apher 21:202-206, 2006

You might also like