You are on page 1of 16

Expert Review of Clinical Immunology

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/ierm20

Autoimmune hemolytic anemia: causes and


consequences

B Fattizzo & W Barcellini

To cite this article: B Fattizzo & W Barcellini (2022) Autoimmune hemolytic anemia:
causes and consequences, Expert Review of Clinical Immunology, 18:7, 731-745, DOI:
10.1080/1744666X.2022.2089115

To link to this article: https://doi.org/10.1080/1744666X.2022.2089115

© 2022 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group.

Published online: 14 Jun 2022.

Submit your article to this journal

Article views: 7108

View related articles

View Crossmark data

Citing articles: 5 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=ierm20
EXPERT REVIEW OF CLINICAL IMMUNOLOGY
2022, VOL. 18, NO. 7, 731–745
https://doi.org/10.1080/1744666X.2022.2089115

REVIEW

Autoimmune hemolytic anemia: causes and consequences


a,b a
B Fattizzo and W Barcellini
a
Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy; bDepartment of Oncology and Hemato-oncology,
University of Milan, Milan, Italy

ABSTRACT ARTICLE HISTORY


Introduction: Autoimmune hemolytic anemia (AIHA) is classified according to the direct antiglobulin Received 29 January 2022
test (DAT) and thermal characteristics of the autoantibody into warm and cold forms, and in primary Accepted 09 June 2022
versus secondary depending on the presence of associated conditions. KEYWORDS
Areas covered: AIHA displays a multifactorial pathogenesis, including genetic (association with con­ Warm autoimmune
genital conditions and certain mutations), environmental (drugs, infections, including SARS-CoV-2, hemolytic anemia; cold
pollution, etc.), and miscellaneous factors (solid/hematologic neoplasms, systemic autoimmune dis­ agglutinin disease;
eases, etc.) contributing to tolerance breakdown. Several mechanisms, such as autoantibody produc­ rituximab; direct
tion, complement activation, monocyte/macrophage phagocytosis, and bone marrow compensation antiglobulin test;
are implicated in extra-/intravascular hemolysis. Treatment should be differentiated and sequenced complement system;
according to AIHA type (i.e. steroids followed by rituximab for warm, rituximab alone or in association monocyte/macrophage
system; thrombosis;
with bendamustine or fludarabine for cold forms). Several new drugs targeting B-cells/plasma cells,
infection
complement, and phagocytosis are in clinical trials. Finally, thrombosis and infections may complicate
disease course burdening quality of life and increasing mortality.
Expert opinion: Beyond warm and cold AIHA, a gray-zone still exists including mixed and DAT negative
forms representing an unmet need. AIHA management is rapidly changing through an increasing
knowledge of the pathogenic mechanisms, the refinement of diagnostic tools, and the development
of novel targeted and combination therapies.

1. Introduction erythrocytes at 0–4°C), but a huge gray zone of mixed and


overlapping conditions still exists and deserves separate con­
Autoimmune hemolytic anemia (AIHA) is a rare disease with
sideration from both a pathogenic and diagnostic point of
an incidence of 0.8 to 3/100,000 people per year and is caused
view [4]. Additionally, the advent of novel therapies targeting
by an autoimmune attack against erythrocyte antigens [1,2].
specific pathogenic steps, further advocates for better disease
While accumulated knowledge exists about disease pathogen­
classification. In this review, we will address AIHA pathogen­
esis, the etiology of AIHA is still largely obscure and consid­
esis and treatment and try to shed light on the current and
ered ‘multifactorial.’ Similar to other autoimmune conditions,
future understanding of the ‘causes and consequences’ of
several risk factors for AIHA development have been described
anti-red cell autoimmunity.
including endogenous and exogenous noxae such as drugs,
infections, systemic autoimmune diseases, hematologic and
solid neoplasms, and congenital syndromes [3]. Additionally,
2. Pathogenic mechanisms
emotional stressors and environmental pollution may possibly
represent under-investigated risk factors for autoimmunity Autoimmunity can arise through several mechanisms, includ­
that may deserve dedicated studies in the future. All these ing a cross-reaction between antigens of infectious agents and
elements may contribute to the derangement of immune self-molecules (named ‘molecular mimicry’), a drug-induced
tolerance and favor the emergence of anti-red cell autoanti­ modification of erythrocyte membrane by adsorption or bind­
bodies with different abilities to cause clinically relevant ing of drugs, and the production of autoantibodies by the so-
hemolysis. The severity of the latter, the underlying condition, called ‘forbidden clones’ during B lymphoproliferative syn­
and the therapies used to treat AIHA may all lead to several dromes [1,5]. Additionally, the breakage of ‘self-tolerance’
complications, including infections, thrombosis, and mortality, may be favored by the presence of germinal mutations impair­
but also result in an increasingly recognized burden for ing immune system maturation, such as those of congenital
patients’ quality of life. Recently, treatment of AIHA has been immunodeficiencies, or by the continuous exposure of ery­
more clearly defined for ‘warm’ (wAIHA, where antibodies are throcyte self-antigens as happens in patients with ineffective
mainly IgG with highest affinity to the antigen at 37°C) and erythropoiesis (i.e. congenital anemias and myelodysplastic
‘cold’ forms, where cold agglutinins are IgM binding to syndromes) [6]. Overall, AIHA pathogenesis is complex

CONTACT B Fattizzo bruno.fattizzo@unimi.it Hematology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, via Francesco Sforza 35, Milan
20100, Italy
This article has been republished with minor changes. These changes do not impact the academic content of the article.
© 2022 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
732 B. FATTIZZO AND W. BARCELLINI

activated macrophages, and increased levels of IL-2 and


Article highlights IL-12 that promote Th1 differentiation [7]. Transforming
● AIHA is a rare and heterogeneous disease with an incidence of 0.8-3/
growth factor (TGF)-β is another regulatory cytokine
100,000 people year. whose levels were found to be elevated in AIHA. TGF-
● AIHA recognizes several pathogenic mechanisms including autoanti­ β favors the differentiation of Th17, which produce IL-
body production, complement activation, monocyte/macrophage
phagocytosis, and bone marrow compensation.
17 that amplifies the pro-inflammatory and autoim­
● Standard therapies should be differentiated between warm AIHA mune responses and decreases T-regs [11]. Finally, folli­
(steroids followed by rituximab) and cold AIHA (rituximab alone or cular T-helper cells, which contribute to generate
in association with bendamustine or fludarabine as the first line).
● Several new drugs targeting B-cells/plasma cells (B-cell receptor
memory B cells and long-lived plasma cells, have also
inhibitors, anti-CD38 monoclonal antibodies), complement (anti-C1s, been implied in autoantibodies production in AIHA [7].
C1q, C3 monoclonal antibodies), and phagocytosis (spleen tyrosine (c) Hemolysis mediated by the monocytic/macrophage sys­
kinase inhibitor and anti-neonatal Fc receptor monoclonal antibodies)
are under study.
tem is typical of IgG wAIHA and mainly occurs extra-
● Several complications, including thrombosis and infections, may vascularly in the spleen [12]. The mechanism includes
worsen disease course burdening quality of life and increasing the recognition of the Fc fragment of IgG by cells of the
mortality.
monocyte-macrophage system with consequent ery­
throcyte phagocytosis and antibody-dependent cellular
cytotoxicity (ADCC). Additionally, other immune cells
such as natural killer (NK) and neutrophils that express
(Figure 1) and mainly involves antibody production by (a) the receptors for the IgG Fc fragment may also mediate an
B-cell compartment and through (b) the alteration of ADCC. Notably, the spleen is also a lymphatic organ able
T-lymphocytes homeostasis, and final erythrocyte destruction to produce autoantibodies. The monocytic/macrophage
through (c) the mononuclear phagocytic system and (d) the system is also involved in complement-mediated (C3b)
activation of the complement cascade. Additionally, (e) the extravascular hemolysis, mainly occurring in lymphoid
efficacy of bone marrow compensatory response is a major organs and liver [5,6].
determinant of AIHA pathogenesis and severity [7]. (d) Complement system is activated through the classical
pathway [13], mainly by pentameric IgM (mainly cold
(a) Anti-erythrocyte autoantibodies are mainly produced by reactive, rarely warm), but also by abundant monomeric
B lymphocytes at different stages of maturation. IgG, also depending on the subclasses (i.e. IgG3 more
Interestingly, warm IgG autoantibodies are mainly poly­ than IgG1; IgG2 is a weak activator; no evidence for
clonal and produced by self-reactive nonmalignant lym­ IgG4). This results in the phagocytosis of C3b-coated
phocytes residing in bone marrow and spleen, cells (extravascular hemolysis) or, to a lesser extent, in
while IgM from CAD is more often monoclonal and the terminal complement activation, with formation of
sustained by a CD20+ CD5-lymphoid bone marrow infil­ the membrane attack complex and intravascular
trate. This also happens in AIHA secondary to lympho­ hemolysis.
proliferative disorders, such as chronic lymphocytic (e) Hematopoietic bone marrow compensates anemia by
leukemia (CLL), where autoantibodies are mainly poly­ increasing erythropoiesis as mirrored by the increased
clonal IgG (90%) produced by non-CLL lymphocytes and number of reticulocytes (immature RBCs with nuclear
rarely IgM directly released by the malignant cells [8]. residue) in the peripheral blood. Reticulocytosis indi­
Similar to this mechanism is polyclonal B-cell activation, cates an efficient bone marrow compensation,
occurring in different conditions such as infections with while reticulocytopenia, present in up to 20% of
EBV, CMV, hepatotropic virus, and HIV. adults and 40% of children, correlates with greater
(b) T-cell compartment is also involved through an imbal­ clinical severity and slower recovery [14,15] and may
ance among T helper (Th)1, Th2, Th17, and regulatory be due to autoimmunity against bone marrow pre­
T-cells (Tregs), and the production of several cytokines cursors [7].
that favor immune tolerance breakage [5]. Results from
various studies are sometimes conflicting, highlighting Finally, it should be noted that anti-RBC autoantibodies are
the heterogeneity of immune response in AIHA not necessarily pathogenic since they can be detected in
patients, often tested at different disease stages healthy individuals (including blood donors); the lack of
(onset, remission, and relapse) and under various thera­ pathogenicity may be due to different Ig subclasses, avidity,
pies. Specifically, Th2 response appears overactive, with ability to fix complement, and thermal amplitude (i.e. cold
a consistent increase of interleukin (IL)-4, IL-6, and IL-10 autoantibodies reacting at 4°C are less likely to induce clini­
in AIHA patients versus healthy controls [9,10], and cally significant hemolysis) [1,2].
a decrease of Interferon (IFN)-γ, promoting humoral
autoimmunity. Additionally, IL-10 has anti-
3. Diagnosis and classification of the different types
inflammatory and regulatory properties and its dysre­
of AIHA
gulation likely contributes to loss of self-tolerance to
RBC autoantigens and autoantibody production. The first step is a thorough medical history and physical
Cellular immunity is also involved, with elevated activity examination for signs and symptoms of acute (symptomatic
of cytotoxic CD8 + T lymphocytes, natural killer cells, anemia possibly accompanied by abdominal pain and
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 733

Figure 1. Pathogenic mechanisms involved in autoimmune hemolytic anemia (AIHA). Several predisposing conditions are involved in AIHA development including genetic
background, presence of underlying immunodeficiencies, infections, solid and hematologic neoplasms. The main immune mechanisms responsible for tolerance breakdown are
molecular mimicry, emergence of forbidden clones, and polyclonal B-cell activation. Additionally, drugs and hematologic treatments, such as checkpoint inhibitors (CPI) and
hematopoietic stem cell transplant (HSCT) may induce immune dysregulation or generation of neoantigens. The resulting immunologic activation involves antigen presenting
cells (APC), T- and B-cells, and cytokines and leads to the production of autoantibodies of several classes. B-cells interact with T helper (Th) cells via CD40 and its ligands but also
present cognate antigens, acting as professional APC. This interaction also contributes to generate class-switched autoantibodies. Concerning cytokines, interleukin (IL)-4, IL-6,
and IL-10 levels are increased and interferon gamma (IFN-γ) decreased favoring Th2 response and boosting humoral autoimmunity; the IL-2 and IL-12 levels are also increased
contributing to Th1 differentiation and cellular autoimmunity. Transforming growth factor beta (TGF-β) is elevated favoring Th17 differentiation and production of IL-17 that
decrease T-regulatory (Treg) levels. Once produced, IgG autoantibodies may weakly activate the complement cascade, and mainly destroy erythrocytes via antibody dependent
cellular cytotoxicity and phagocytosis primarily in the spleen. IgM autoantibodies strongly activate complement system and cause erythrocyte destruction via C3b opsonization
and phagocytosis primarily in the liver. These mechanisms are both called extravascular hemolysis. IgM may lead to terminal complement activation until the formation of the
membrane attack complex (MAC) with consequent intravascular hemolysis.

hemoglobinuria) or chronic hemolysis (milder fatigue, jaun­ monospecific antisera (anti-IgG, -IgA, -IgM, anti-complement,
dice, gallstones, splenomegaly, peripheral cold-induced cya­ -C), allowing the classification of AIHA into wAIHA (DAT posi­
nosis, etc.); furthermore, recent infections, drug exposure, tive with IgG or IgG+C at low titer, <64), and cold forms (DAT
chronic diseases, tumors, and transplant should be evaluated. positive with anti-C only and presence of cold agglutinins
Laboratory workup shows anemia of various degrees with usually of high titer). The latter are further classified in cold
elevated reticulocytes, increased indirect bilirubin and LDH agglutinin diseases (CAD), where monoclonal IgM is sustained
(more typical of intravascular hemolysis), and decreased hap­ by a clonal lymphoproliferative bone marrow disease similar
toglobin [1,2,16]. The direct antiglobulin test (DAT) is the to low-grade non-Hodgkin lymphoma, termed ‘CA-associated
mainstay of AIHA diagnosis and should be performed with lymphoproliferative bone marrow disorder’ [17–19], and cold
734 B. FATTIZZO AND W. BARCELLINI

agglutinin syndrome (CAS), occurring secondary to infections, upon specific clinical suspects. Finally, the determination of
such as Mycoplasma pneumoniae, EBV, CMV, and COVID-19, or endogenous erythropoietin (EPO) levels is suggested in
malignancies (aggressive B-cell lymphoma). Rarer forms patients with inadequate reticulocytosis, particularly if relap­
include mixed AIHA [20], where DAT is positive for IgG and sing and heavily treated [7].
C and cold agglutinins with high thermal amplitude are
detected. Additionally, some Authors suggest that the titer
of cold agglutinins, along with positive autoagglutination at 4. Conditions associated with AIHA development
20°C, may be helpful to distinguish mixed AIHA from IgG+C and secondary forms
wAIHA [2,4]. The very rare paroxysmal cold hemoglobinuria
Slightly more than 50% of AIHA cases are secondary to asso­
(PCH) should also be considered, the latter is sustained by
ciated conditions, i.e. chronic lymphocytic leukemia, systemic
a biphasic IgG hemolysin called Donath-Landsteiner antibody
lupus erythematosus (SLE), common variable immune deficiency,
[4] that binds to erythrocytes at low temperatures but acti­
or other immunologic or lymphoproliferative disorders [8,23,24].
vates complement at 37°C resulting in intravascular hemolysis.
Furthermore, several endogenous and exogenous noxae may
The disease is nowadays mainly temporary, self-limiting, and
increase the risk of autoimmunity as detailed thereafter.
occurring in children after viral infections [4]. Other atypical
cases include warm IgA driven AIHA, the very severe and
potentially fatal warm IgM cases, characterized by huge intra­
4.1. Endogenous noxae associated with AIHA
vascular hemolysis due to massive complement activation at
development
body temperature, and DAT negative AIHA [1,21]. DAT may be
falsely negative due to a low number of erythrocyte-bound 4.1.1. Genetics and congenital conditions associated with
autoantibodies or to low-affinity ones. In these cases, more AIHA
sensitive second-level tests may be performed in reference Several studies highlighted the association of AIHA with altera­
centers. However, DAT remains negative in up to 10% of tions of genes involved in antigen recognition, T- and B-cell
AIHAs and the diagnosis is based on the exclusion of other maturation and proliferation, and cytokine homeostasis. The
hemolytic conditions. Specifically, other acquired forms should formers include specific HLA loci (HLA-B8, BW6, DR, and DQ)
be ruled out, including mechanical and toxic noxae (i.e. ana­ [25,26] and stereotyped rearrangements of genes encoding the
mnesis for intravascular devices, prosthetic valves, angiomas, variable region of the immunoglobulin heavy and light chains
radiation, and drugs), paroxysmal nocturnal hemoglobinuria (IGHV4-34, IGHV3, and IGKV3-20) [27–30]. Additionally, the
(high-resolution cytofluorimetry), thrombotic microangiopa­ G polymorphism of cytotoxic T-lymphocyte-associated protein
thies (platelets values, coagulation parameters, blood smear 4 (CTLA-4, a negative regulator of T-cell responses) and the AG
for schistocytes), and infections (i.e. malaria); subsequently, configuration of lymphotoxin-α (LT-α) gene showed a higher
congenital hemolytic anemias (i.e. RBC membrane and frequency among patients with AIHA [31,32]. A stronger genetic
enzyme defects) should be considered through the evaluation predisposition was recently demonstrated in pediatric patients
of family history, blood smear, osmotic fragility tests, and with Evans syndrome, where AIHA occurs concomitantly with
enzyme activities [3,7]. Thereafter, response to steroid therapy autoimmune thrombocytopenia and/or neutropenia. By next-
allows the ex juvantibus diagnosis of DAT negative AIHA. generation sequencing, a genetic lesion was detected in 65%
In all patients, the clinical evaluation is completed by the of cases, mainly involving genes related to primary immunode­
investigation of underlying alloantibodies in patient’s serum ficiencies (TNFRSF6, CTLA4, STAT3, PIK3CD, CBL, ADAR1, LRBA,
(indirect agglutinin test, IAT), reported in about one-third of RAG1, and KRAS). These pathogenic hints have prognostic and
AIHA patients [22]. The latter may cause severe hemolytic therapeutic importance, since mutated patients diagnosed with
reactions in the case of transfusion. In complex cases, allo- primary immunodeficiencies are more prone to catastrophic
and autoantibody may be distinguished by immune- infections after immunosuppression [33] and may be candidates
absorbance techniques and extended RBC genotyping [3]. for new biological drugs used in these conditions. Mutations
Additionally, autoantibodies may be eluted from the washed typical of primary immunodeficiencies include those of KMT2D
patient’ RBCs to determine the class, specificity, titer, and and CARD11, which have been described in 69% and 31% of CAD
thermal range. As regards specificity, in wAIHA autoantibodies patients, respectively [34], and are the hallmark of Kabuki syn­
are mainly directed against RBC membrane proteins including drome. The latter is a congenital disorder characterized by mal­
Band 3, glycophorin A, and Rh system, while in CAD the formations, immune-deficiency, and autoimmune phenomena,
polysaccharide regions of glycoproteins, such as I/i or Pr anti­ including AIHA in 4% of cases. Other primary immunod
gen, are the most common target. eficiencies include common variable immunodeficiency (CVID)
Bone marrow evaluation is useful to evaluate compensa­ [35], IgA deficiency [36], and autoimmune lymphoproliferative
tory erythroid hyperplasia, features suggestive of underlying syndromes (ALPS) [37]. Finally, AIHA may develop in patients
bone marrow failure or lymphoma, and the presence and type with congenital anemia, including beta-thalassemia (up to 6%),
of lymphoid infiltrate that may be differently targeted with sickle cell disease, and hereditary spherocytosis, where erythro­
available therapy. It is advised in CAD at diagnosis, in poiesis is overstimulated and largely ineffective [38]. With more
relapsed/refractory cases of wAIHA, and in cases of suspected sensitive methods, such as mitogen-stimulated DAT (MS-DAT),
underlying hematologic diseases [2,4]. Similarly, whole-body anti-erythrocyte autoantibodies were demonstrated in up to
CT scan, serology for infections and autoimmune diseases, and 61% of patients with hereditary spherocytosis, although with
testing of anti-phospholipid antibodies are recommended doubtful pathogenic significance [39].
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 735

4.1.2. AIHA in the context of connective tissue diseases HCV, HBV, and HAV, and more rarely HIV [3]. In particular, HCV-
and other autoimmune conditions related AIHA was mainly sustained by cold agglutinins (CAS)
AIHA belongs to the criteria for the diagnosis of SLE and may and had a mortality rate as high as 56% in a large study [50].
represent a mild manifestation of the disease or be its leading The risk of AIHA in HCV patients consistently increased from
feature, with overt hemolysis and treatment requirements. The 2.8- to 11.6-fold in cases treated with interferon [51].
frequency of AIHA was about 14% in children and 3% in adults Additionally, AIHA may develop in up to 3% of patients with
with SLE, again underlining the likely contribution of a more infectious mononucleosis, within 1–2 weeks of onset, and
immature immune system in the former [40]. Other associated more frequently as a CAS [3]. Regarding bacteria, CAS may
autoimmune diseases include inflammatory bowel diseases follow Mycoplasma pneumoniae infection [52], as well as
(complicated by AIHA in about 4% of cases), Sjogren's syn­ tuberculosis, and usually recovers after anti-tuberculosis treat­
drome (in about 2.8% of cases), autoimmune liver disorders (in ment [53]. More rare associations include AIHA complicating
1.4% of cases), and case reports of systemic sclerosis, sclero­ brucellosis or undulant fever, and the above mentioned PCH
derma, CREST syndrome, and thyroid autoimmune disorders. classically preceded by syphilis and virus infections [3].
Although such associations do not necessarily imply Finally, alterations of the microbiome (i.e. the ensemble of
a causative relationship, AIHA severity is correlated with the microorganisms colonizing various districts of the body) are
degree of organ involvement and disease activity of the emergingly described through next-generation sequencing
underlying autoimmune condition [3]. (NGS) techniques and have been associated with the develop­
ment of autoimmune diseases, including autoimmune cytope­
4.1.3. AIHA associated with neoplasms nias [54]. A disrupted microbiome composition may lead to
Concerning solid neoplasms, AIHA is highly associated with autoimmunity through molecular mimicry, chronic inflamma­
ovarian teratoma and thymoma (up to 9% of cases), mainly tion, alteration of epithelial barriers, and dissequestration of
occurs during disease progression, and tends to ameliorate self-antigens. On the other hand, the microbiome perturba­
after tumor resection, thymectomy, and/or chemotherapy [41]. tions may also be an effect of the autoimmune disease and its
An association has also been reported with carcinomas [42]. treatment. For instance, in immune thrombocytopenia and
Very recently, a Japanese group described ectopic band 3 chronic idiopathic neutropenia, a peculiar microbiota compo­
expression and erythrocyte membrane-bound IgG (by flow cyto­ sition has been identified, possibly predictive of response to
metry) in 97% of 50 colorectal cancer patients, with a DAT therapy, and future studies will possibly clarify its role in other
positivity in 10 of them. The presence of anti-band three auto­ conditions including AIHA.
antibodies was confirmed in patients’ sera and mouse experi­
ments suggested that ectopic band 3 expression occurs in
4.2.2. Drugs associated with AIHA
tumor cells under hypoxic conditions [43]. Regarding hematolo­
gical diseases, AIHA is frequently associated with lymphoproli­ Drug-induced AIHAs [1,23,55–57] are generally categorized
ferative disorders, such as CLL (5–10% of cases), non-Hodgkin according to the pathogenic mechanism in forms due to (1)
lymphoma (NHL, 2–3%), and rarely HL (0.2%), which may pre­ drug-dependent antibodies that activate an immune response
cede or follow the hemolytic disease [3,8]. AIHA prevalence only while the drug is present and (2) drug-independent anti­
increases in specific NHL subtypes, being maximal in angioim­ bodies. The first encompasses hapten-mediated antibodies,
munoblastic T-cell lymphoma [44] and in marginal zone lym­ recognizing a mixed epitope composed of erythrocyte parts
phoma [45]. Notably, CAD is associated with the above- and drug non-covalently bound to RBC, and includes penicillin
mentioned CAD-associated lymphoproliferative disease that (DAT positive for IgG only) and ceftriaxone (DAT positive for
may be difficult to distinguish from other NHLs but usually IgG, IgG + C, or C only). Drug-independent antibodies induce
presents with a 10–15% isolated bone marrow infiltrate [18]. AIHA via adsorption, immune dysregulation, or unknown
Similar to solid cancers, CLL/NHL treatment usually induces mechanisms and may occur after cladribine, methyldopa,
AIHA recovery. As described for congenital anemias, the positiv­ and fludarabine [2,3]. The latter, a purine analogue used in
ity of anti-erythrocyte autoantibodies by MS-DAT is frequent in the CLL treatment, further induces an imbalance between
CLL [46], but even in myelodysplastic syndromes, and in primary Th17 and T-regs [7], that is restored by the association of
myelofibrosis [47,48], again with only a few cases developing cyclophosphamide and rituximab that kill autoreactive T-cells
clinical hemolysis. Finally, AIHA may rarely complicate multi­ and markedly reduce AIHA [58]. Concerning novel oral B-cell
centric Castleman disease and large granular lymphocyte (LGL) receptor inhibitors, increased incidence of autoimmune hepa­
lymphoproliferative disorders and should be included in the titis, colitis, and pneumonitis has been reported after the PI3K
differential diagnosis of anemia in this setting [49]. inhibitor idelalisib, and a single case report of AIHA post- the
BCL-2 inhibitor venetoclax has been published. Contrarily,
Bruton’s tyrosine kinase inhibitor ibrutinib seems safe and
favored the resolution of primary and secondary AIHAs, pos­
4.2. Exogenous noxae associated with AIHA
sibly through the inhibition of autoantibodies producing
development
B-cells and restoration of T-cell homeostasis [59–61]. Finally,
4.2.1. Infections associated with AIHA and the role of novel immune-activating anti-cancer drugs whose therapeutic
microbiome effect is based on enhancing host immune response against
AIHA may follow several infections, particularly Parvovirus B19, tumor cells may favor AIHA development. Immune checkpoint
in up to 20% of cases, hepatotropic virus infections, including inhibitors (CPIs) that reactivate T-cell mediated
736 B. FATTIZZO AND W. BARCELLINI

immunosurveillance are the main example. They inhibit pro­ more events conferred a 70% increased risk for hospitalization
grammed death receptor 1 and its ligand (PD1 and PDL1) or for autoimmune diseases [70]. Although only nine AIHAs were
CTLA-4 and may evoke severe autoimmune reactions, includ­ reported in this study, this remains an open field for further
ing fulminant AIHA [62,63]. Recent studies reported a total of investigation.
82 cases mainly occurring after nivolumab, followed by pem­
brolizumab, ipilimumab, and atezolizumab. All cases displayed 4.2.5. Possible association of pollution with autoimmunity
severe hemolysis, with transfusion requirement (80%), high Several epidemiology studies linked air pollution (a heteroge­
prevalence of DAT negativity (38%), relapse in about 50% of neous mixture including carbon monoxide, nitrates, sulfur
patients, and mortality as high as 17% [62,63]. A last mention dioxide, ozone, lead, toxic by-product of tobacco smoke and
deserves the occurrence of cytopenias in about 30% of particulate matter) to the occurrence of autoimmune diseases
patients with acute lymphoblastic leukemia and NHL treated through various mechanisms including systemic inflammation,
with chimeric antigen T-cell infusions; these cases may be life- oxidative stress, epigenetic modifications, and chronic airway
threatening and steroid-refractory and are likely to be immune damage [73]. In this view, air pollutants have been shown to
mediated [64]. bind to the aryl hydrocarbon receptor (AHR) in the lung to
regulate Th17 and Tregs balance. Furthermore, pollutants trig­
4.2.3. Transplant associated AIHA ger inducible bronchus associated with lymphoid tissue,
AIHA complicates 5–10% of liver and/or intestinal transplanta­ resulting in the production of pro-inflammatory cytokines.
tion and 2.5% of pancreas transplants [2]. In this context, it is The latter stimulates B- and dendritic cells thus favoring the
worth mentioning the passenger lymphocyte syndrome that is production of antibodies and self-reactive T lymphocytes.
sustained by immune competent lymphocytes transferred Clinical examples are the effect of smoking and occupational
with the donor organ [65]. Onset is between 3- and 24-days exposure to silica on rheumatoid arthritis and SLE develop­
post-transplant, the risk is proportional to the burden of ment, and the association of inhalation of chemical solvents,
transplanted lymphocytes, ranging from 9 to 70% of cases herbicides, and silica with scleroderma and ANCA associated
(kidney < liver < heart-lung transplants) [66], and hemolysis vasculitis [74]. Although a direct effect on AIHA development
is generally transient. Regarding hematopoietic stem cell has not been reported yet, air pollutants have been recently
transplant (HSCT), 2–4% of patients may experience AIHA shown to affect erythropoiesis, and particulate matter (PM2.5)
after 3–10 months, and mortality increases with infections and nitrogen dioxide (NO2) levels have been associated with
[67,68]. Some risk factors for AIHA developing post HSCT lower hemoglobin in older Americans and were also related to
include unrelated donor, HLA-mismatch, graft-versus-host dis­ higher C-reactive protein levels as markers of systemic inflam­
ease (GVHD), cord blood as HSC source, age <15 years, CMV mation [75].
reactivation, alemtuzumab use, and nonmalignant diagnosis
pre-HSCT. The passenger lymphocyte syndrome may also
5. Standard therapy
occur after HSCT and has been linked to HLA mismatches,
use of peripheral blood as stem cell source, cyclosporine Based on the above-mentioned differences in physiopathol­
alone for GVHD prophylaxis, reduced-intensity conditioning, ogy, the treatment of warm and cold AIHA forms differs in the
and female donors, while cord blood use appears protective. choice of the type and sequence of immunosuppressive
Careful transfusion procedures are warranted in transplanted agents [1; 2,4,6]. In both conditions, the utility of supportive
patients, particularly in mismatched cases [69]. measures, including transfusions, nutrient supplementation,
erythropoiesis stimulating agents, and anticoagulant and anti-
4.2.4. Possible effect of emotional stress on the infective prophylaxis should be considered. Finally, secondary
development of autoimmunity forms may necessitate treatment for the underlying disease,
Several hematologic patients, including those with autoim­ such as chemo-immunotherapy for lymphomas and solid
mune conditions, report that their diagnosis had been pre­ tumors, intravenous immunoglobulin (IVIG) in primary immu­
ceded by a variety of traumas/stressors either physical nodeficiencies, cytotoxic immunosuppressants in systemic
(accidents, infections, etc.) or psychological (bereavement, dis­ autoimmune diseases (Table 1).
missal, divorce, etc.). Their contribution to disease develop­
ment is difficult to establish, but it has been speculated that
5.1. Primary warm-AIHA
they may interact with the immune system by altering the
adrenergic/glucocorticoid axis thus interfering with disease Predniso(lo)ne 60–100 mg or 1 mg/kg body weight per day for
course [70]. Some evidence come from pediatric studies eval­ 2–3 weeks is the recommended first-line therapy, followed by
uating the long-term effects of childhood traumatic stress, a slow tapering in 3–6 months. About 80% respond initially,
including increased prevalence of altered inflammatory mar­ but only 30–40% experience sustained remission after 1 year
kers and higher risk of ischemic heart disease [71,72]. A recent [76]. Rituximab addition in the first line, which prolonged
analysis of 15,357 adults enrolled in the Adverse Childhood response duration in two prospective studies [77], should be
Experiences (ACEs) Study from 1995 to 1997 in California, and considered in severe settings (i.e. Hb<8 g/dL, presence of
followed-up until 2005, evaluated the effect of physical, emo­ atypical AIHA or Evans syndrome). In steroid refractory
tional, or sexual abuse on the frequency of hospitalizations for patients, second-line rituximab is the preferred choice, after
autoimmune diseases. The Authors found that 64% of patients re-assessment for secondary AIHA forms and bone marrow
reported at least one ACE and that the occurrence of 2 or evaluation, with response rates of 70–80% in about 3–
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 737

Table 1. Standard therapies for warm AIHA (wAIHA) and cold agglutinin disease (CAD).
Response
Treatment Route and dose rates % Comments
wAIHA
1st line
Steroids Oral or IVPrednisone or 75–80 Curative in 20–30% only; short-term side effects (mood swings, psychosis); long-term side
methylprednisolone 1 mg/Kg/ effects (diabetes, hypertension, infections, osteoporosis, cushingoid syndrome)
day
2nd line
Rituximab IV 375 mg/m2 or 100 mg/week 80–90 Effective even at low-dose; re-treatment equally effective; well-established safety profile
x 4 weeks
Further lines
Splenectomy / 80 Potentially curative; unsuitable for elderly; surgical complications; thrombotic risk; life-long
immune suppression; infection prophylaxis required
Azathioprine Oral 50–100 mg/day 40–60 Steroid-sparing agent; myelosuppression; infections; secondary malignancy; hepatic
toxicity
Cyclophospamide Oral 50 mg day 40–60 Steroid-sparing; myelosuppression; infections; urotoxicity; secondary malignancy; fertility
problems/teratogenic
Cyclosporin A Oral 3–5 mg/Kg/day 40–60 Steroid-sparing; possible dose adjustment; hypertension; arrhythmia; myelosuppression;
infections; nephrotoxicity
Mycophenolate Oral 1–1.5 g/day 80–100 Steroid-sparing; particularly effective in children; good safety profile; mild
Mofetil myelosuppression
Danazol Oral 200–600 mg/day 40 Steroid-sparing agent; low effectiveness in refractory cases; long-term hepatotoxicity
High dose IV boluses 800 mg/m2 70 Response in small series; high toxicity (myelosuppression; infections; urotoxicity; secondary
cyclophosphamide malignancy; infertility)
Additional
therapies
Intravenous IV 0.4 g/Kg x 5 days or 1 g/Kg 40 Effective in AIHA secondary to infections; particularly effective in pediatric settings (60%);
immunoglobulin x 2 days low toxicity
Recombinant Subcutaneous 40,000 IU/week 50–70 Effective in multi-refractory AIHA particularly with reticulocytopenia; useful in very severe
erythropoietin presentation
CAD
Watch and wait - - keeping patient and room warm and transfusion of pre-warmed to 37°C blood
First line
Steroids Oral or IV prednisone or 15–30 Effective only at high dose; short-term side effects (mood swings, psychosis); long-term
methylprednisolone 1 mg/Kg/ side effects (diabetes, hypertension, infections, osteoporosis, cushingoid syndrome)
day
Rituximab IV 375 mg/m2/week x 4 weeks 50 Re-treatment equally effective; well-established safety profile
Further lines
Rituximab/ Oral 40 mg/m2 days 1–5, 29–34, 75 Sustained remissions (median response duration 6.5 years); hematologic and infectious
fludarabine 57–61 and 85–89 toxicity mostly in older and frail patients
Rituximab/ IV 90 mg/m2 days 1–2 for 4 70 Sustained remissions; infectious complications
Bendamustine cycles of 28 days
Bortezomib IV or subctuaneous 1.3 mg/m2 30 Side effects (neuropathy, neutropenia, thrombocytopenia, diarrhea, fatigue and rash)
days 1,4, 8,11
Additional
therapies
Recombinant Subcutaneous 40,000 IU/week 50–70 Effective in multi-refractory cases particularly with reticulocytopenia
erythropoietin

6 weeks. The conventional dose is 375 mg/m2 weekly for 4 dexamethasone, high-dose cyclophosphamide, or stem cell
weeks, but a fixed low dose of 100 mg weekly for 4 weeks transplantation in ultra-refractory cases [2].
yielded equal efficacy [78]. Splenectomy is currently recom­
mended in patients unresponsive to/relapsing after rituximab,
with responses of about 70%. The risk of severe infection and 5.2. Cold agglutinin disease
thrombosis should be taken into account and prevented (see In a proportion of patients with mild anemia or compensated
the dedicated section) [79]. Further options include azathiopr­ hemolysis without circulatory symptoms and fatigue, non-
ine, cyclophosphamide, cyclosporine, mycophenolate mofetil, pharmacologic management with thermal protection is
or bortezomib, basing mostly on small retrospective series and advised [2,4,80]. In symptomatic cases, glucocorticoids are
case reports. For emergencies, high-dose intravenous methyl­ effective only at unacceptably high doses, so that rituximab
prednisolone and IVIG may be appropriate, and some reports has become the preferred first-line therapy with response
or retrospective series of plasma exchange, emergency sple­ rates of 45–60%, mainly partial. A majority of responders
nectomy, or partial splenic embolization have been published. relapse within 12–15 months, and repeated rituximab courses
Finally, limited evidence regards the use of bortezomib plus have a fair chance to succeed. The addition of fludarabine
738 B. FATTIZZO AND W. BARCELLINI

has yielded higher responses, but with more toxicity, 6. New drugs
while the association with bendamustine seems safer and
Novel treatments mainly target autoantibody production by
even more effective. The overall response exceeded 70%
the B-cell/plasma-cell compartment or the final erythrocyte
and deepened overtime, with an estimated median response
breakdown by either complement or mononuclear phagocyte
duration of >88 months [81]. For refractory patients, borte­
system (Table 2) [6]. The formers include monoclonal antibo­
zomib monotherapy (1 cycle of 1.3 mg/sm iv for total 4
dies mainly used in secondary AIHA, such as ofatumumab
biweekly doses) may be considered, with responses in 32%
(anti-CD20), alemtuzumab (anti-CD52), and daratumumab
in a small prospective study [82]. Finally, a recent case series
(anti-CD38). The latter targets long-lived plasma cells that do
of 10 CAD patients (primary or secondary) treated with ibru­
not express CD20 and may cause rituximab refractoriness.
tinib, described responses in all of them, suggesting the need
Isatuximab, another anti-CD38 MoAb is under investigation
of future investigation [83]. Complement inhibitors represent
in wAIHA in phase 1 study [NCT04661033]. Oral B-cell receptor
a promising approach that will be addressed in the following
inhibitors parsaclisib [NCT03538041 and NCT05073458], ibru­
paragraph.
tinib [NCT03827603], and rilzabrutinib [NCT05002777] are also
being studied in clinical trials in wAIHA and CAD with promis­
ing results.
5.3. Other AIHAs
Complement modulation is the most promising drug under
In mixed AIHA cases [20], which are generally more severe and study for CAD: sutimlimab, a monoclonal antibody against
refractory/relapsing, clinical-laboratory features (monospecific complement protein C1s, demonstrated short time to
DAT, cold agglutinin titers, autoagglutination, presence of cold response, rapid normalization of hemolysis, and good safety
agglutinin symptoms) should be evaluated at each relapse to profile in a phase 3 trial [86]; pegcetacoplan, a pegylated
assess the prevailing form (warm versus cold). Glucocorticoids peptide that inhibits C3 [6] also showed good activity in
should be given generously, and rituximab considered as an CAD and wAIHA with IgG+C DAT positivity. In the latter,
early second line, particularly in the presence of CAD features, a novel C1q inhibitor ANX005 is also being developed
while splenectomy is discouraged [4]. [NCT04691570]. Notably, complement inhibitors do not elim­
The very rare PCH is usually self-remitting and may be inate antibody production and will probably have to continue
handled with steroids. Recently, the efficacy of a single dose indefinitely. However, the very short time to response may be
of the complement C5 inhibitor eculizumab in a child was particularly helpful in severely anemic patients and acute
reported [84]. Treatment for drug-induced hemolytic anemia crises. The reticuloendothelial system may be targeted by
mainly consists of discontinuation of suspected medications, inhibiting the spleen tyrosine kinase with fostamatinib,
and transfusions and steroids were required in severe cases which also inhibits the B-cell receptor downstream pathway
such as those occurring post-CPIs. For the latter, early ritux­ and is now in phase 2 studies in wAIHA [NCT02612558].
imab may be required given the high rate of steroid-refractory Finally, the safety/efficacy of several inhibitors of the neonatal
cases. Fc receptor (FcRn) such as intravenous nipocalimab
[NCT03075878] and subcutaneous RVT-1401 [NCT04253236],
are under investigation. The FcRn, which is homologous to
5.4. Supportive measures the MHC Class I receptor family, is widely expressed, and
mediates IgG recycling conferring to the antibodies their
Transfusions must not be denied in cases of life-threatening
long half-life. Blocking FcRn increases IgG clearance including
anemia (i.e. Hb<6 g/dL or conditions of increased O2
that of pathogenic IgG autoantibodies.
requests), but over-transfusion should be avoided. If time
permits, an extended phenotyping should be performed
with respect to Rh subgroups, Kell, MNS, Kidd, S/s, and Duffy 7. Consequences of AIHA and its treatment
antigens to avoid alloimmunization that may occur in up to
one-third of cases [2,7,22]. Genotyping can be considered but The detrimental events that may complicate AIHA clinical
will often be too time-consuming. In CAD, compatible erythro­ course include disease relapse, the above-mentioned associa­
cytes by crossmatching at 37°C are usually available and may tion with other autoimmune cytopenias, the occurrence of
be safely transfused provided adequate warming of the thrombosis and infections, and the rare evolution into other
patient and the extremity chosen for infusion. Vitamin (B12, lymphoid or myeloid hematologic conditions. All these may
folic acid, vitamin D) and iron supplementation should be have an impact on patients’ mortality and overall quality of life
tailored on specific deficiencies/need of osteoporosis preven­ and are discussed thereafter [3].
tion. Anticoagulant and anti-infective prophylaxis should also
be considered. Finally, recombinant erythropoietin (i.e. epoe­
7.1. Risk of AIHA relapse
tin alpha 40,000 UI/week subcutaneously) may improve hemo­
globin in w-AIHA as well as CAD and can be used as The severity of anemia at onset constituted the major determi­
a supportive strategy in patients with inadequate reticulocy­ nant of disease relapse in two recent large multicenter studies
tosis [85]. Plasma exchange (with the use of albumin substitu­ [15,87]. Relapse risk augmented with Hb decrease, by about 7%
tion instead of plasma to avoid complement supplementation) increased risk per each gram of reduction of Hb. Concerning
may be successfully used in refractory life-threatening cases, AIHA type, CAD, mixed and atypical forms were at higher risk of
while specific immunosuppressive therapy is instituted [2,6]. relapse and required multi-treatment, while wAIHA had a more
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 739

Table 2. New drugs in warm AIHA (wAIHA) and cold agglutinin disease (CAD). [92], and the close interplay among autoimmunity/autoinflam­
Route of mation, complement aactivation,and the coagulation cascade.
Mechanism Administration Study Phase Setting Moreover, the presence of underlying diseases, such as anti-
B-cell directed therapies phospholipid syndrome, active infections, and neoplasms may
Ofatumumab Anti-CD20 IV Case report Secondary
AIHA favor the occurrence of thrombotic complications [91,93]. On
Alemtuzumab Anti-CD52 SC Case reports Secondary the whole, the use of anticoagulant prophylaxis should be
AIHA considered in hyper-hemolytic, infected AIHA patients, pro­
Daratumumab Anti-CD38 IV Case reports wAIHA/CAD/
Secondary vided safe platelet counts. A comprehensive evaluation of all
AIHA risk factors is advised, including thrombophilia screening (par­
Isatuximab Anti-CD38 SC Phase 1 wAIHA ticularly lupus anticoagulant, LAC) in selected cases and before
Ibrutinib BTKi Oral Case reports, Secondary
Phase 2 plus AIHA splenectomy [2,88,91]. Finally, an ongoing prospective rando­
rituximab mized study will evaluate the efficacy of prolonged oral antic­
Rilzabrutinib BTKi Oral Phase 1 wAIHA oagulation with apixaban in preventing thrombosis during the
Venetoclax Bcl2i Oral Case reports Secondary
AIHA 12-weeks after AIHA diagnosis [NCT05089227].
Parsaclisib PI3Kδi Oral Phase 2, Phase wAIHA/CAD
3
Complement inhibitors
Eculizumab Anti-C5 IV Case reports CAD/Mixed 7.4. Risk of infections
and Phase 2 AIHA
Sutimlimab Anti-C1s IV Phase 1b and 3 CAD Infections may be both a trigger for AIHA development and
Pegcetacoplan Anti-C3 SC Phase 1/2 and CAD/wAIHA
/C3b Phase 3 relapse and be caused by immunosuppressive treatment in
ANX005 Anti-C1q IV Phase 1 wAIHA IgG+C a vicious circle. They complicate about 20% of AIHA cases,
IgG-mediated with a frequency increase along with the number of therapy
phagocytosis inhibitors
Fostamatinib * Syki Oral Phase 2 wAIHA lines. Interestingly, beyond splenectomy, which is associated
Orilanolimab Anti-FcRn IV Phase 1b wAIHA with a known risk of sepsis by capsulated bacteria, repeated
Nipocalimab Anti-FcRn IV Phase 2 wAIHA steroid therapy at each relapse seemed to have a prominent
RVT-1401 Anti-FcRn SC Phase 2 wAIHA
role in recent studies [87,88,94]. In thrombosis, underlying
*Fostamatinib is also a B-cell receptor inhibitor; BTKi: Bruton tyrosine kinase
inhibitor; Bcl2: B-cell lymphoma 2; δPI3Ki: phosphatidylinositol 3-kinase-δ diseases such as primary immunodeficiencies and malignan­
inhibitor; MoAb: monoclonal antibody; mTORi: mammalian target of cies and their therapies may lead to increased infectious risk
Rapamycin inhibitor; Syki: Spleen tyrosine kinase inhibitor; FcRn: neonatal Fc deserving special attention. On the whole, testing HBV/HCV
receptor
status, quantiferon for tuberculosis, is highly advised in patient
candidates to immunosuppression with high-dose steroids,
rituximab, or cytotoxic drugs to prompt adequate prophylaxis.
benign course. Overall, complement activation seems to be an Cotrimoxazole is advisable in patients receiving >25 mg day
additional risk factor for relapse, particularly after rituximab. prednisone (or equivalent) for >4 weeks, anti-viral agents (i.e.
lamivudine) and surveillance in those at risk for HBV reactiva­
tion, referral to an infective disease specialist for anti-
7.2. Evans syndrome (ES)
tubercular treatment if tuberculosis is suspected. HCV positive
The association of immune thrombocytopenia and/or neutro­ subjects should be referred for possible eradication.
penia to AIHA has a significant impact on prognosis. Vaccinations against capsulated bacteria (Meningococcus,
Particularly, relapse rate exceeded 70% in these patients, and Haemophilus, and Pneumococcus) are mandatory before sple­
more than 50% required ≥3 therapy lines in a recent analysis nectomy [95].
[88]. If cumulated with Hb < 8 g/dL or forms other than
WAIHA, the presence of ES was associated wit 3-/4-folds
higher than other cases [87]. ES patients had also a higher
7.5. Steroid induced side effects
incidence of infections (33%) and thrombotic complications
(21%) as compared with primary AIHA cases, correlating with In a recent survey, it has been calculated that AIHA patients
the number of therapy lines and resulting in a mortality rate received a cumulative dose of 8.2 ± 2.1 kg (mean± standard
approaching that of low-grade hematologic malignancies, error) in a mean follow-up of 12 ± 3.5 months [94]. This may
despite young age. be associated with well known but sometimes under recog­
nized side effects including arterial hypertension, diabetes,
gastrointestinal dysfunction, neurological disorders, and
7.3. Risk of thrombosis
osteoporosis. Concerning the latter, assessment of bone den­
Thrombotic events occur in about 15–20% of patients sity scan and supplementation with calcium and vitamin
[17,89,90] and were mainly observed in patients with very D may aid prompt recognition/prevention of harmful sponta­
severe anemia (i.e. Hb levels <6 g/dL) at onset, intravascular neous fractures. Additionally, vitamin D levels were found
hemolysis (LDH > 1.5 x upper limit of normality ULN), previous reduced in AIHA patients versus controls, particularly in
splenectomy, and presence of ES [87,91]. The pathogenesis is those who had received more therapy lines; furthermore, the
still unclear and likely includes the contribution of nitric oxide addition of vitamin D exerted a dose-dependent inhibition on
depletion by free Hb, the release of erythrocyte microparticles in vitro production of anti-erythrocyte antibodies [96]. These
740 B. FATTIZZO AND W. BARCELLINI

findings further suggest preventive administration of vitamin their own words. However, communication skills of both the
D in patients with AIHA. patient and the caregiver are highly heterogeneous and
impede an objective evaluation of quality of life, and the
use of structured surveys is helpful. The UK group recently
7.6. AIHA evolving into other hematologic diseases administered a semi-structured interview to 12 adult
Various reports described the possible evolution of AIHA into patients with AIHA and found that the disease had
clonal hematologic diseases and bone marrow failure. In par­ a significant impact on QoL for most patients [103]. On
ticular, lymphoproliferative diseases may become clinically a 0–10 scale – where 10 was the ‘worst I’ve ever felt’ –
overt in patients followed for AIHA, more frequently as the 77% of subjects scored 5–10, and 31% scored 9 or 10.
evolution of an underlying misdiagnosed lymphoid indolent Patients described that the worst aspects of AIHA included
infiltrate into aggressive forms, particularly in CAD [17]. the impact of investigation or treatment, symptoms (mainly
Concerning myeloid diseases (from idiopathic cytopenia/dys­ fatigue), and anxiety over outcome. Multiple QoL domains
plasia of unknown significance to myelodysplastic syndromes), were affected, including daily living, working, leisure, and
we previously described the presence of hypercellularity, dys­ even sleep. Finally, patients and their partners’ emotional
erythropoiesis, and increased reticulin fibrosis in about one- wellbeing were adversely affected. Available questionnaires
third of AIHA cases, associated with inadequate reticulocyto­ are being actively used in the afore mentioned clinical trials
penia and chronic/relapsing course. Furthermore, we reported to assess the beneficial impact of novel drugs on QoL [86].
two cases of AIHA and ES with anti-erythroblast antibodies, However, an effort to design disease-specific surveys will be
which showed a clear-cut bone marrow erythrocyte precursor expected in the next future.
hyperplasia at diagnosis but evolved into idiopathic cytope­
nia/dysplasia of uncertain significance (ICUS/IDUS) and aplas­
tic anemia (AA) after several years [97]. It may be hypothesized 8. COVID-19 infection and AIHA
that refractory/relapsing AIHAs may shift from a predominant
‘peripheral’ pattern toward a ‘central’ autoimmunity over time, 8.1. AIHA associated with SARS-CoV-2 infection
with a likely contribution of chronic inflammation, aging, and In the last two years, SARS-CoV-2 has become one of the
accumulation of somatic mutations [6]. In this sense, small major triggers for the development and exacerbations of
clones of paroxysmal nocturnal hemoglobinuria have been autoimmune cytopenias, including AIHA and Evans syndrome
described in AIHA patients, possibly evolving to clinically [104–107]. The latter may in turn increase the need of immu­
overt disease [98]. nosuppressive treatment and, consequently, the frailty of this
patient population. Pathogenically, molecular mimicry among
7.7. Mortality of AIHA SARS-CoV-2 antigens and red cells’ epitopes seems the promi­
nent mechanism [108], followed by the over-inflammation
In a large retrospective series, mortality of AIHA patients was triggered by the virus that may result in cross activation of
around 10–20% and mainly associated with very severe ane­ the many arms of the immune system (humoral and cellular
mia. Patients presenting with Hb <6 g/dL had an overall immunity, complement, coagulation cascade, etc.) against self-
mortality of 24% versus 18% of those with Hb >6 g/dL. Age, antigens in an ‘innocent bystander’ fashion. The latter may also
multi-treatment, comorbidities, and the occurrence of compli­ favor the oxidative stress of erythrocytes and platelets with
cations, further impacted patients outcome [99,100]. exposure of phosphatidyl serine and consequent clearance by
Specifically, mortality risk was predicted by ES, acute renal the mononuclear phagocyte system [109,110]. COVID-19-
failure, and infections (hazard risk, HR 8, 95% CI 2.5–26; HR associated AIHA also displays some epidemiologic peculiari­
6.3, 1.4–29; HR 4.8, 1.5–15, respectively) [15,87]. In a series of ties. In fact, a higher incidence of ES was noted, while it usually
13 very severe multitreated primary AIHA mortality reached complicates only one-fifth of AIHAs [15], and mainly in adults
57%, despite transfusions, steroid boli, IVIG, rituximab, EPO, that are more frequently infected by the virus. Additionally,
and plasma-exchange [101]. More recently, Lafarge et al. a higher prevalence of cold-reactive complement-activating
described a mortality rate of up to 30% in 44 AIHA patients autoantibodies (up to 50%) was found as compared to primary
admitted to the intensive care unit for severe anemia [102]. In AIHA [111]. From a therapeutic point of view, SARS-CoV-2
CAD, a recent large multicenter study reported a disease- associated AIHA and ES were mainly handled with
related mortality of 11% and a median estimated survival of a combination of steroids and intravenous immunoglobulins,
16 years from diagnosis (5-year survival 83%) [17]. Finally, irrespective of AIHA type. Rituximab was administered in only
secondary AIHAs may also impact on disease severity and one case displaying cold reactive autoantibodies [104]. This
prognosis and higher mortality was observed in AIHA second­ drug represents a concern during septic states, including
ary to lymphoma and cancer [2]. SARS-CoV-2 infection, and has been associated with a more
severe COVID19 in rheumatic disorders [112]. Supportive treat­
ments, including rEPO and IVIG may be also considered.
7.8. Quality of life in AIHA
Finally, the thrombotic risk of AIHA adds to that of COVID19
Clinicians are used to evaluate patients’ quality of life (QoL) thus advocating for anticoagulant prophylaxis with low mole­
by simply asking the patients to communicate the percep­ cular weight heparin in these patients, provided a safe platelet
tion of health status and disease impact on daily living in count [113].
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 741

8.2. AIHA associated with SARS-CoV-2 vaccine the clinical course. The latter may be affected by several com­
plications including the occurrence of thrombosis and infec­
Several reports highlighted the possible impaired immune
tions, and the rare evolution into other lymphoid or myeloid
response to SARS-CoV-2 vaccines in patients requiring immu­
hematologic conditions. Finally, SARS-CoV-2 and its vaccines
nosuppression, including autoimmune cytopenias. Specifically,
represent novel triggers of AIHA development/exacerbations,
recent reports showed that hematological patients treated
including warm and cold forms, as well as cases of Evans syn­
with monoclonal antibodies (such as rituximab, daratumumab,
drome with several diagnostic and therapeutic pitfalls.
and the anti-B cell early maturation antigen belantamab) do
not mount humoral response to vaccines. Additionally, it has
been shown that steroids reduce both humoral and T cell 10. Expert opinion
response to COVID19 vaccine in hematologic patients and in
Anti-erythrocyte autoimmunity has long been recognized as the
those with autoimmune diseases [114]. On the other hand,
most suitable scholastic example to teach autoimmunity, since it
SARS-CoV-2 vaccine may trigger autoimmune phenomena by
involves different classes of antibodies with variable thermal
activating ADCC as well as the classical complement cascade.
characteristics, complement, cytokines, and cellular immune
In the literature, various AIHA, both warm and cold, ITP, and
effectors, such as T-lymphocytes and monocyte-macrophages.
Evans syndromes developing/reactivating after SARS-CoV-2
Additionally, AIHA is unusual among autoimmune diseases in
vaccine have been reported [115–118], irrespective of vaccine
that the target autoantigens are well characterized (i.e. band 3,
type and dose. All patients have been successfully managed
glycophorin A, and Rh system in wAIHA and I/i system in CAD).
with steroids, adjustment of ongoing treatment, or supportive
Notwithstanding the simplicity of the cell targeted by the
therapy [118]. Interestingly, two large cohort studies showed
immune attack, and the easiness of monitoring the degree of
that post-vaccine ITP has an incidence inferior to that
the immune activation (as compared to other more complex
expected for primary ITP in the general population (0.8 versus
systemic autoimmune diseases), anti-erythrocyte autoimmunity
3.3 cases per 100,000 adults per year) [119,120]. Overall, it is
highlights the complex interaction of several immune effectors
suggested to perform vaccinations as far as possible from
and the existence of gray zones that elude classification. For
rituximab treatment and at the lowest steroid dose and to
example, complement activation is the well-established key
strictly monitor Hb and hemolytic markers for possible exacer­
mechanisms in CAD; however, the disease is undoubtedly due
bations since they are manageable if promptly recognized.
to an antibody (mainly IgM, usually monoclonal). Thus, the ther­
apeutic strategy would better benefit from a combination of
B-cell/plasma cell-directed drugs and complement inhibition.
9. Conclusions
Additionally, thermal characteristics are not trivial: while an IgM
In conclusion, this review highlights the multifaceted nature of reacting only at cold temperature will result in minimal/negligible
AIHA including a multifactorial etiology, encompassing genetic clinical consequences, the very rare IgM with thermal range close
predispositions and environmental exposures leading to toler­ to body temperature may be the most severe and potentially
ance breakage. The distinction between cold and warm forms fatal forms. Paradoxically, these cases have not been included in
by DAT with monospecific antisera is pivotal since they show CAD trials, even if they would probably benefit from early com­
diverse hemolytic patterns (i.e. IgG mediated extravascular plement inhibition. On the other hand, in wAIHA, it is assumed
hemolysis in the spleen versus complement mediated hemolysis that complement plays a secondary role, and most clinical trials
in the liver or intravascularly) and should be treated differently. are focused on targeting B-cells/plasma cells, inhibiting ADCC, or
Furthermore, a prompt recognition/exclusion of the associated removing pathologic antibodies via the Fc-Rn. This strategy may
conditions (infections, drugs, tumors, hematologic conditions, exclude the severe wAIHA with DAT positivity for IgG and C,
systemic autoimmune diseases, congenital immunodeficiencies, where complement inhibition may have a definite reason to be
etc.) is fundamental, since they may complicate AIHA diagnosis, performed. Finally, the strict and scholastic distinction between
and may require specific management (i.e. CT scan, bone mar­ wAIHA and CAD leaves an unmet need for mixed forms, which
row evaluation, genetic testing). Treatment of wAIHA mainly are generally more severe and relapsing than other AIHAs, and
relies on steroids as the first line, followed by rituximab in have no therapeutic options after steroids and rituximab failure.
refractory/relapsing cases. Splenectomy is generally deferred to Altogether, these considerations underline the need to identify
the three line in young low-comorbid patients. Asymptomatic not only the main mechanism involved in erythrocyte destruction
CAD patients may be handled with cold temperatures avoid­ but also all the complex interactions of the different immune
ance, while those subjects displaying moderate-to-severe ane­ effectors. This is also pivotal for a disregarded mechanism, i.e.
mia or disabling peripheral symptoms require frontline bone marrow compensation, which may be optimal in early
rituximab (since steroids are effective only at high unacceptable disease, but progressively decreasing with patient’s age, disease
doses). For the rarer mixed and atypical AIHA forms, which may duration/relapses, and presence of anti-erythroblast autoimmu­
be more severe and difficult to diagnose, there is great uncer­ nity. Furthermore, in clinical practice, strict classifications may be
tainty and a close interaction with the laboratory physicians is difficult to apply, and tests’ positivities may vary overtime and
pivotal. The role of bone marrow compensation and the efficacy according to the clinical setting (host immunity, previous treat­
of recombinant erythropoietin in patients with inadequate ery­ ments, presence of triggers). For instance, a high dose steroid
thropoiesis should be always considered. Several novel drugs treatment received for the first AIHA episode may abate/mask the
are being studied in clinical trials and will target the different ‘warm’ part of a mixed AIHA at relapse. Additionally, during
pathogenic steps of AIHA that may dynamically change during polyclonal activation, as that occurring during infections, B-cell
742 B. FATTIZZO AND W. BARCELLINI

populations may release autoantibodies of different Ig subclasses. ORCID


This may also occur during primary IgM immune responses that B Fattizzo http://orcid.org/0000-0003-0857-8379
may be immature and less specific. Finally, the presence of W Barcellini http://orcid.org/0000-0003-1428-9944
a complement-activating trigger, such as SARS-CoV-2, may favor
complement deposition on red blood cells and further interfere
with DAT findings. References
Beyond the diagnostic issues, the study of the so-called
Papers of special note have been highlighted as either of interest (•) or of
risk factors and predisposing conditions is gaining impor­
considerable interest (••) to readers.
tance. The latter include modifiable environmental triggers 1. Petz LD, Garratty G. Immune Hemolytic Anemias. 2nd ed.
that affect the general population, such as air pollutants and Philadelphia: Churchill Livingstone; 2004.
other stressors, that might allow the establishment of pre­ 2. Jäger U, Barcellini W, Broome CM, et al. Diagnosis and treatment of
ventive measures in the future. In this view, a fascinating autoimmune hemolytic anemia in adults: recommendations from the
first international consensus meeting. Blood Rev. 2020May; 41:100648.
field is the analysis of the microbiome and its interactions
10.1016/j.blre.2019.100648. Epub 2019 Dec 5. PMID: 31839434
with disease development and treatment. Dysbiosis may in •• the first international consensus paper on management of
fact represent both a modifiable risk factor for AIHA onset autoimmune hemolytic anemia
and relapses, as well as a trigger for infectious complications 3. Barcellini W, Giannotta J, Fattizzo B. Autoimmune hemolytic anemia in
following immunosuppression. In the near future, the avail­ adults: primary risk factors and diagnostic procedures. Expert Rev
Hematol. 2020Jun;13(6):585–597. Epub 2020 Apr 29. PMID: 32274943.
ability of NGS techniques will improve our ability to analyze
4. Berentsen S, Barcellini W. Autoimmune hemolytic anemias. N Engl
these microorganisms with a likely impact on how we man­ J Med. 2021 Oct 7;385(15):1407–1419. PMID: 34614331.
age several diseases, including AIHA. Broader, molecular • an updated practical review on autoimmune hemolytic anemia
techniques will allow the definition of the genomic land­ 5. Barcellini W, Fattizzo B. The changing landscape of autoimmune
scape of this disease, such as the presence of germinal and hemolytic anemia. Front Immunol. 2020 Jun 3;11:946. 10.3389/
fimmu.2020.00946. PMID: 32655543; PMCID: PMC7325906.
somatic mutations that may predispose to autoimmunity
• a review highlighting novel aspects of autoimmune hemolytic
through an altered antigen recognition, an imbalance of B/ anemia pathogenesis and clinical features.
T populations and of cytokine milieu, and the emergence of 6. Barcellini W, Zaninoni A, Giannotta JA, et al. New insights in auto­
clonal entities. The latter may include ‘clonal myelopoiesis/ immune hemolytic anemia: from pathogenesis to therapy stage 1.
lymphopoiesis of indeterminate potential’ that may be the J Clin Med. 2020 Nov 27;9(12):3859. PMID: 33261023; PMCID:
PMC7759854.
herald of autoimmunity in hematology. The same techniques
7. Fattizzo B, Barcellini W. Autoimmune cytopenias in chronic lym­
will possibly help to clarify why some patients are healed, phocytic leukemia: focus on molecular aspects. Front Oncol. 2020
some relapse and become chronic, and some other evolve Jan 10;9:1435. DOI:10.3389/fonc.2019.01435. PMID: 31998632;
into neoplastic conditions or bone marrow failure. PMCID: PMC6967408.
Therefore, the management of AIHA will significantly 8. Abbas AK, Lichtman AH, Pillai S. Cellular and molecular immunol­
ogy. 7th ed. Philadelphia: Saunders; 2012.
change through a deepening knowledge of the various patho­
9. Barcellini W, Clerici G, Montesano R, et al. In vitro quantification of
genic mechanisms, to the refinement of diagnostic tools and anti-red blood cell antibody production in idiopathic autoimmune
disease classification, and to the development of novel tar­ haemolytic anaemia: effect of mitogen and cytokine stimulation. Br
geted and combination therapies. J Haematol. 2000;111(2):452–460.
10. Toriani-Terenzi C, Fagiolo E. IL-10 and the cytokine network in the
pathogenesis of human autoimmune hemolytic anemia. Ann
N Y Acad Sci. 2005;1051(1):29–44.
Declaration of interest 11. Xu L, Zhang T, Liu Z, et al. Critical role of Th17 cells in development
B Fattizzo received consultancy for Amgen, Alexion, Annexon, Apellis, of autoimmune hemolytic anemia. Exp Hematol. 2012;40
Momenta, Novartis, and Sobi. W Barcellini received consultancy for Agios, (12):994–1004.
Alexion, Annexon, Sanofi, Novartis, and Sobi. The authors have no other 12. Gallagher MT, Branch DR, Mison A, et al. Evaluation of reticuloen­
relevant affiliations or financial involvement with any organization or entity dothelial function in autoimmune hemolytic anemia using an in vitro
with a financial interest in or financial conflict with the subject matter or assay of monocyte-macrophage interaction with erythrocytes. Exp
materials discussed in the manuscript apart from those disclosed. Hematol. 1983Jan; 111: 82–89PMID: 6832239.
13. Berentsen S. Role of complement in autoimmune hemolytic
anemia. Transfus Med Hemother. 2015Sep;42(5):303–310. Epub
2015 Sep 7. PMID: 26696798; PMCID: PMC4678321.
Reviewer disclosures 14. Conley CL, Lippman SM, Ness PM, et al. Autoimmune hemolytic
Peer reviewers on this manuscript have no relevant financial or other anemia with reticulocytopenia and erythroid marrow. N Engl
relationships to disclose. J Med. 1982 Feb 4;306(5):281–286.PMID: 7054700.
• pioneering article on the importance of bone marrow compen­
sation in autoimmune hemolytic anemia
15. Barcellini W, Fattizzo B, Zaninoni A, et al. Clinical heterogeneity and
Author contributions predictors of outcome in primary autoimmune hemolytic anemia:
B Fattizzo and W Barcellini wrote the manuscript and revised it for a GIMEMA study of 308 patients. Blood. 2014 Nov 6;124
important intellectual content. All authors agree for the final version of (19):2930–2936. Epub 2014 Sep 16. PMID: 25232059.
the manuscript to be published. •• one of the largest series of autoimmune hemolytic anemia
ever reported
16. Fattizzo B, Giannotta JA, Serpenti F, et al. Difficult cases of auto­
Funding immune hemolytic anemia: a challenge for the internal medicine
specialist. J Clin Med. 2020 Nov 27;9(12):3858. PMID: 33261016;
This study was funded by Italian Ministry of Health - Current research IRCCS. PMCID: PMC7760866.
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 743

17. Berentsen S, Barcellini W, D’Sa S, et al., Cold agglutinin disease revis­ 36. Odineal DD, Gershwin ME. The epidemiology and clinical manifes­
ited: a multinational, observational study of 232 patients. Blood. 136 tations of autoimmunity in selective IgA deficiency. Clin Rev Allergy
(4): 480–488. 2020. Immunol. 2019;58(1):107–133.
• one of the largest series of cold agglutinin disease patients 37. Oliveira JB. The expanding spectrum of the autoimmune lympho­
ever reported proliferative syndromes. Curr Opin Pediatr. 2013;25(6):722–729.
18. Randen U, Trøen G, Tierens A, et al. Primary cold agglutinin-associated 38. Motta I, Giannotta J, Ferraresi M, et al. Autoimmune hemolytic
lymphoproliferative disease: a B-cell lymphoma of the bone marrow anemia as a complication of congenital anemias. a case series
distinct from lymphoplasmacytic lymphoma. Haematologica. 2014;99 and review of the literature. J Clin Med. 2021 Aug 2;10(15):3439.
(3):497–504. PMID: 34362222; PMCID: PMC8347040.
19. Małecka A, Delabie J, stlie I, et al. Cold agglutinin-associated B-cell 39. Zaninoni A, Vercellati C, Imperiali FG, et al. Detection of red blood
lymphoproliferative disease shows highly recurrent gains of chro­ cell antibodies in mitogen-stimulated cultures from patients with
mosome 3 and 12 or 18. Blood Adv. 2020;4(6):993–996. hereditary spherocytosis. Transfusion. 2015;55(12):2930–2938.
20. Shulman IA, Branch DR, Nelson JM, et al. Autoimmune hemolytic 40. Gormezano NW, Kern D, Pereira OL, et al. Autoimmune hemolytic
anemia with both cold and warm autoantibodies. JAMA. 1985 Mar anemia in systemic lupus erythematosus at diagnosis: differences
22-29;253(12):1746–1748. PMID: 3974053. between pediatric and adult patients. Lupus. 2017;26(4):426–430.
• pioneering evidence of mixed type autoimmune hemolytic 41. Nenova IS, Valcheva MY, Beleva EA, et al. Autoimmune phenomena
anemia in patients with solid tumors. Folia Med (Plovdiv). 2016;58
21. Sokol RJ, Booker DJ, Stamps R, et al., Direct coombs test-negative (3):195–199.
autoimmune hemolytic anemia and low-affinity IgG class 42. Sokol RJ, Booker DJ, Stamps R. Erythrocyte autoantibodies, auto­
antibodies. Immunohematology. 1997;13(4):115–118. immune haemolysis, and carcinoma. J Clin Pathol. 1994 Apr;47
• pioneering article on direct antiglobulin test negative autoim­ (4):340–343. PMID: 8027372; PMCID: PMC501938.
mune hemolytic anemia 43. Kitao A, Kawamoto S, Kurata K, et al. Band 3 ectopic expression in
22. Branch DR, Petz LD. Detecting alloantibodies in patients with colorectal cancer induces an increase in erythrocyte
autoantibodies. Transfusion. 1999 Jan;39:6–10. PMID: 9920160 membrane-bound IgG and may cause immune-related anemia.
23. Hill QA, Stamps R, Massey E, et al., Guidelines on the management Int J Hematol. 2020 May;111(5):657–666. Epub 2020 Jan 30. PMID:
of drug-induced immune and secondary autoimmune, haemolytic 31997080
anaemia. Br J Haematol. 2017;177(2):208–220. 44. Crickx E, Poullot E, Moulis G, et al. Clinical spectrum, evolution,
•• one of the first guideline article on the management of sec­ and management of autoimmune cytopenias associated with
ondary autoimmune hemolytic anemia angioimmunoblastic T-cell lymphoma. Eur J Haematol. 2019;103
24. Azizi G, Tavakol M, Rafiemanesh H, et al. Autoimmunity in a cohort (1):35–42.
of 471 patients with primary antibody deficiencies. Expert Rev Clin 45. Dasanu CA, Bockorny B, Grabska J, et al. Prevalence and pattern of
Immunol. 2017;13(11):1099–1106. autoimmune conditions in patients with marginal zone lymphoma:
25. Abdel-Khalik A, Paton L, White AG, et al. Human leucocyte antigens a single institution experience. Conn Med. 2015;79(4):197–200.
A, B, C, and DRW in idiopathic “warm” autoimmune haemolytic 46. Barcellini W, Montesano R, Clerici G, et al. In vitro production of
anaemia. Br Med J. 1980;280(6216):760–761. anti-RBC antibodies and cytokines in chronic lymphocytic
26. Wang-Rodriguez J, Rearden A. Reduced frequency of HLA-DQ6 in leukemia. Am J Hematol. 2002 Nov;713:177–183. PMID: 12410572.
individuals with a positive direct antiglobulin test. Transfusion. 47. Zaninoni A, Imperiali FG, Cattaneo A, et al. Detection of erythro­
1996;36(11–12):979–984. blast antibodies in mitogen-stimulated bone marrow cultures from
27. Silberstein LE, Jefferies LC, Goldman J, et al. Variable region gene patients with myelodysplastic syndromes. Transfusion. 2016;56
analysis of pathologic human autoantibodies to the related i and (8):2037–2041.
I red blood cell antigens. Blood. 1991;78(9):2372–2386. 48. Barcellini W, Iurlo A, Radice T, et al. Increased prevalence of auto­
28. Potter KN, Hobby P, Klijn S, et al. Evidence for involvement of immune phenomena in myelofibrosis: relationship with clinical and
a hydrophobic patch in framework region 1 of human V4-34- morphological characteristics, and with immunoregulatory cyto­
encoded Igs in recognition of the red blood cell I antigen. kine patterns. Leuk Res. 2013;37(11):1509–1515.
J Immunol. 2002;169(7):3777–3782. 49. Barcellini W, Giannotta JA, Fattizzo B. Autoimmune complications
29. Jefferies LC, Carchidi CM, Silberstein LE. Naturally occurring in hematologic neoplasms. Cancers (Basel). 2021 Mar 26;13(7):1532.
anti-i/I cold agglutinins may be encoded by different VH3 PMID: 33810369; PMCID: PMC8037071.
genes as well as the VH4.21 gene segment. J Clin Invest. 50. Ramos-Casals M, García-Carrasco M, López-Medrano F, et al. Severe
1993;92(6):2821–2833. autoimmune cytopenias in treatment-naive hepatitis C virus infec­
30. Malecka A, Trøen G, Tierens A, et al. Immunoglobulin heavy and tion: clinical description of 35 cases. Medicine (Baltimore). 2003;82
light chain gene features are correlated with primary cold aggluti­ (2):87–96.
nin disease onset and activity. Haematologica. 2016;101(9):e361–4. 51. Chiao EY, Engels EA, Kramer JR, et al. Risk of immune thrombocy­
31. Pavkovic M, Georgievski B, Cevreska L, et al. CTLA-4 exon 1 poly­ topenic purpura and autoimmune hemolytic anemia among 120
morphism in patients with autoimmune blood disorders. Am 908 US veterans with hepatitis C virus infection. Arch Intern Med.
J Hematol. 2003;72(2):14714–14719. 2009;169(4):357–363.
32. D’Abronzo LS, Barros MM, Bordin JO, et al. Analysis of polymorphisms 52. Stein B, DeCredico N, Hillman L. Evaluation of the Direct
of TNF-a, LT-a, IL-10, IL-12 and CTLA-4 in patients with warm autoim­ Antiglobulin Test (DAT) in the setting of mycoplasma pneumoniae
mune haemolytic anaemia. Int J Lab Hematol. 2012;34(4):356–361. Infection. JAMA. 2018;319(13):1377–1378.
33. Hadjadj J, Aladjidi N, Fernandes H, et al., members of the French 53. Ramagopalan SV, Goldacre R, Skingsley A, et al. Associations
reference center for pediatric autoimmune cytopenia (CEREVANCE). between selected immune-mediated diseases and tuberculosis:
Pediatric Evans syndrome is associated with a high frequency of poten­ record-linkage studies. BMC Med. 2013;11(1):97.
tially damaging variants in immune genes. Blood. 2019;134(1):9–21. 54. Fattizzo B, Cavallaro F, Folino F, et al. Recent insights into the role
•• a large series highlighting the importance of genetic testing in of the microbiome in malignant and benign hematologic diseases.
children with Evans syndrome Crit Rev Oncol Hematol. 2021Apr; 160: 103289. Epub 2021 Mar 2.
34. Malecka A, Trøen G, Tierens A, et al. Frequent somatic mutations of PMID: 33667659
KMT 2D (MLL 2) and CARD 11 genes in primary cold agglutinin 55. Garratty G. Drug-induced immune hemolytic anemia. Hematology
disease. Br J Haematol. 2018;183(5):838–842. Am Soc Hematol Educ Program. 2009;2009(1):73–79. PMID: 20008184.
35. Feuille EJ, Anooshiravani N, Sullivan KE, et al. Autoimmune cytope­ 56. Habibi B. Drug induced red blood cell autoantibodies
nias and associated conditions in CVID: a report from the USIDNET co-developed with drug specific antibodies causing haemolytic
registry. J Clin Immunol. 2018;38(1):28–34. anaemias. Br J Haematol. 1985Sep;61(1):139–143. PMID: 2864949.
744 B. FATTIZZO AND W. BARCELLINI

57. Mueller-Eckhardt C, Salama A. Drug-induced immune cytopenias: 75. Honda T, Pun VC, Manjourides J, et al. Anemia prevalence and
a unifying pathogenetic concept with special emphasis on the role hemoglobin levels are associated with long-term exposure to air
of drug metabolites. Transfus Med Rev. 1990;4(1):69–77. pollution in an older population. Environ Int. 2017
58. Fischer K, Bahlo J, Fink AM, et al. Long-term remissions after FCR Apr;101:125–132. Epub 2017 Jan 31. PMID: 28153527; PMCID:
chemoimmunotherapy in previously untreated patients with CLL: PMC5361751.
updated results of the CLL8 trial. Blood. 2016;127(2):208–215. 76. Barcellini W, Fattizzo B. How I treat warm autoimmune hemolytic
59. Autore F, Pasquale R, Innocenti I, et al. Autoimmune hemolytic anemia. Blood. 2021;137(10):1283–1294.
anemia in chronic lymphocytic leukemia: a comprehensive 77. Michel M, Terriou L, Roudot-Thoraval F, et al., A randomized and
review. Cancers (Basel). 2021 Nov 19;13(22):5804. PMID: 34830959; double-blind controlled trial evaluating the safety and efficacy of
PMCID: PMC8616265. rituximab for warm autoimmune hemolytic anemia in adults (the
60. Moreno C. Autoimmune cytopenia and CLL ride together. Blood. RAIHA study). Am J Hematol. 92(1): 23–27. 2017.
2021 Jun 24;137(25):3464–3465. PMID: 34165542. • one of the few prospective trials of rituximab in autoimmune
61. Vitale C, Salvetti C, Griggio V, et al. Preexisting and hemolytic anemia
treatment-emergent autoimmune cytopenias in patients with CLL 78. Murakhovskaya I. Rituximab use in warm and cold autoimmune
treated with targeted drugs. Blood. 2021 Jun 24;137 hemolytic anemia. J Clin Med. 2020;9(12):4034.
(25):3507–3517. PMID: 33651883. 79. Patel NY, Chilsen AM, Mathiason MA, et al. Outcomes and compli­
62. Tanios GE, Doley PB, Munker R. Autoimmune hemolytic anemia cations after splenectomy for hematologic disorders. Am J Surg.
associated with the use of immune checkpoint inhibitors for can­ 2012;204(6):1014–1019.
cer: 68 cases from the food and drug administration database and 80. Berentsen S. How I treat cold agglutinin disease. Blood. 2021;137
review. Eur J Haematol. 2019;102(2):157–162. (10):1295–1303.
63. Leaf RK, Ferreri C, Rangachari D, et al. Clinical and laboratory 81. Berentsen S, Randen U, Oksman M, et al. Bendamustine plus ritux­
features of autoimmune hemolytic anemia associated with imab for chronic cold agglutinin disease: results of a Nordic pro­
immune checkpoint inhibitors. Am J Hematol. 2019;94(5):563–574. spective multicenter trial. Blood. 2017 Jul 27;130(4):537–541. Epub
64. Schubert ML, Schmitt M, Wang L, et al. Side-effect management of 2017 May 22. PMID: 28533306.
chimeric antigen receptor (CAR) T-cell therapy. Ann Oncol. 2021;32 • prosepective trial in cold agglutinin disease
(1):34–48. 82. Pasquale R, Giannotta JA, Barcellini W, et al. Bortezomib in auto­
65. Hows J, Beddow K, Gordon-Smith E, et al. Donor-derived red blood immune hemolytic anemia and beyond. Ther Adv Hematol. 2021
cell antibodies and immune hemolysis after allogeneic bone marrow Nov 12;12:20406207211046428. PMID: 34795889; PMCID:
transplantation. Blood. 1986Jan; 67(1): 177–181.PMID: 3079641. PMC8593301.
• pioneering article on the passenger lymphocyte syndrome 83. Jalink M, Berentsen S, Castillo JJ, et al. Effect of ibrutinib treatment
66. Skeate R, Singh C, Cooley S, et al. Hemolytic anemia due to pas­ on hemolytic anemia and acrocyanosis in cold agglutinin disease/
senger lymphocyte syndrome in solid malignancy patients treated cold agglutinin syndrome. Blood. 2021 Nov 18;138(20):2002–2005.
with allogeneic natural killer cell products. Transfusion. 2013;53 PMID: 34293088.
(2):419–423. 84. Lau-Braunhut SA, Stone H, Collins G, et al. Paroxysmal cold hemo­
67. Barcellini W, Fattizzo B, Zaninoni A. Management of refractory globinuria successfully treated with complement inhibition. Blood
autoimmune hemolytic anemia after allogeneic hematopoietic Adv. 2019 Nov 26;3(22):3575–3578. PMID: 31738828; PMCID:
stem cell transplantation: current perspectives. J Blood Med. PMC6880888.
2019;10:265–278. 85. Fattizzo B, Michel M, Zaninoni A, et al. Efficacy of recombinant
68. González-Vicent M, Sanz J, Fuster JL, et al. Autoimmune hemolytic erythropoietin in autoimmune hemolytic anemia: a multicenter
anemia (AIHA) following allogeneic hematopoietic stem cell trans­ international study. Haematologica. 2021 Feb 1;106(2):622–625.
plantation (HSCT): a retrospective analysis and a proposal of treat­ PMID: 32354865; PMCID: PMC7849557.
ment on behalf of the Grupo Español De Trasplante De Medula • important evidence on the efficacy of recombinant erythro­
Osea En Niños (GETMON) and the Grupo Español de Trasplante poietin in patients with autoimmune hemolytic anemia
Hematopoyetico (GETH). Transfus Med Rev. 2018;S0887- 86. Röth A, Barcellini W, D’Sa S, et al. Sutimlimab in cold agglutinin
7963:30164–30165. disease. N Engl J Med. 2021 Apr 8;384(14):1323–1334. PMID:
• large series of patients developing autoimmune hemolytic 33826820.
anemia post-transplant • prospective trial with complement inhibitor in cold agglutinin
69. Kruizinga MD, VT MJD, Bekker V, et al., Risk factors, treatment, and disease
immune dysregulation in autoimmune cytopenia after allogeneic 87. Barcellini W, Zaninoni A, Fattizzo B, et al. Predictors of refractori­
hematopoietic stem cell transplantation in pediatric patients. Biol ness to therapy and healthcare resource utilization in 378 patients
Blood Marrow Transplant. 24(4): 772–778. 2018. with primary autoimmune hemolytic anemia from eight Italian
• large series of patients developing autoimmune hemolytic reference centers. Am J Hematol. 2018Sep;939:E243–6. Epub 2018
anemia post-transplant Aug 25. PMID: 29981267
70. Dube SR, Fairweather D, Pearson WS, et al. Cumulative childhood 88. Fattizzo B, Michel M, Giannotta JA, et al. Evans syndrome in adults:
stress and autoimmune diseases in adults. Psychosom Med. an observational multicenter study. Blood Adv. 2021 Dec 28;5
2009Feb;712:243–250. Epub 2009 Feb 2. PMID: 19188532; PMCID: (24):5468–5478. PMID: 34592758; PMCID: PMC8714709.
PMC3318917 •• largest series of adult patients with Evans syndrome highlight­
71. Felitti VJ, Anda RF, Nordenberg D, et al. The relationship of adult ing their management in the current era
health status to childhood abuse and household dysfunction. Am 89. Audia S, Bach B, Samson M, et al. Venous thromboembolic events
J Prev Med. 1998;14(4):245–258. during warm autoimmune hemolytic anemia. PLoS One. 2018;13
72. Danese A, Pariante CM, Caspi A, et al. Childhood maltreatment (11):e0207218.
predicts adult inflammation in a life-course study. Proc Nat Acad 90. Broome CM, Cunningham JM, Mullins M, et al. Increased risk of
Sci. 2007;104(4):1319–1324. thrombotic events in cold agglutinin disease: a 10-year retrospec­
73. Zhao CN, Xu Z, Wu GC, et al. Emerging role of air pollution in tive analysis. Res Pract Thromb Haemost. 2020 Apr 9;4(4):628–635.
autoimmune diseases. Autoimmun Rev. 2019Jun;186:607–614. PMID: 32548562; PMCID: PMC7292660.
Epub 2019 Apr 5. PMID: 30959217. • large study regarding thrombotic risk in patients with cold
74. Farhat SC, Silva CA, Orione MA, et al. Air pollution in autoimmune agglutinin disease
rheumatic diseases: a review. Autoimmun Rev. 2011Nov;111:14–21. 91. Fattizzo B, Bortolotti M, Giannotta JA, et al. Intravascular hemolysis
Epub 2011 Jul 6. PMID: 21763467. and multitreatment predict thrombosis in patients with
EXPERT REVIEW OF CLINICAL IMMUNOLOGY 745

autoimmune hemolytic anemia. J Thromb Haemost. 2022 May 12; 105. Osti N, Ceolan J, Piccoli P, et al. Acute haemolysis by cold antibody
Epub ahead of print. PMID: 35555857. during SARS-CoV-2 infection in a patient with Evans syndrome:
92. Barcellini W, Zaninoni A, Giannotta JA, et al. Circulating extracellu­ a case report and literature review. Blood Transfus. 2021;20
lar vesicles and cytokines in congenital and acquired hemolytic (2):168–172.
anemias. Am J Hematol. 2021 Apr 1;96(4):E129–32. Epub 2021 106. Barcellini W, Giannotta JA, Fattizzo B. Are patients with
Feb 9. PMID: 33491786. autoimmune cytopenias at higher risk of COVID-19 pneumonia?
93. Samuelson Bannow BT, Lee A, Khorana AA, et al. Management of The experience of a reference center in Northern Italy
cancer-associated thrombosis in patients with thrombocytopenia: and review of the literature. Front Immunol. 2021;11:609198.
guidance from the SSC of the ISTH. J Thromb Haemost. 2018;16 107. Haberman R, Axelrad J, Chen A, et al. Covid-19 in
(6):1246–1249. immune-mediated inflammatory diseases. Case series from New
94. Giannotta JA, Fattizzo B, Barcellini W. Infectious complications in York. N Engl J Med. 2020;383(1):85–88.
a cohort of autoimmune hemolytic anemia patients. Annual meet­ 108. Angileri F, Légaré S, Marino Gammazza A, et al. Is molecular mimi­
ing of the European Hematology Association. Hemasphere 2020. cry the culprit in the autoimmune haemolytic anaemia affecting
Abstract n. PB2401. patients with COVID-19? Br J Haematol. 2020;190(2):e92–3.
95. Giannotta JA, Fattizzo B, Cavallaro F, et al. Infectious complications 109. Chauhan AJ, Wiffen LJ, Brown TP. COVID-19: a collision of comple­
in autoimmune hemolytic anemia. J Clin Med. 2021 Jan 5;10(1):164. ment, coagulation and inflammatory pathways. J Thromb Haemost.
PMID: 33466516; PMCID: PMC7796467. 2020;18(9):2110–2117.
96. Fattizzo B, Zaninoni A, Giannotta JA, et al. Reduced 25-OH vitamin 110. Kubánková M, Hohberger B, Hoffmanns J, et al. Physical pheno­
D in patients with autoimmune cytopenias, clinical correlations and type of blood cells is altered in COVID-19. bioRxiv.
literature review. Autoimmun Rev. 2016Jul;157:770–775. Epub 2016 2021;120:2838–2847.
Mar 14. PMID: 26988993 111. Jacobs J, Eichbaum Q. COVID −19 associated with severe autoim­
97. Barcellini W, Fattizzo B, Zaninoni A, et al. Clinical evolution of mune hemolytic anemia. Transfusion. 2021;61(2):635–640.
autoimmune cytopenias to idiopathic cytopenias/dysplasias of 112. Avouac J, Drumez E, Hachulla E, et al. COVID-19 outcomes in
uncertain significance (ICUS/IDUS) and bone marrow failure patients with inflammatory rheumatic and musculoskeletal dis­
syndromes. Am J Hematol. 2017;92(3):E26–9. eases treated with rituximab: a cohort study. Lancet Rheumatol.
98. Fattizzo B, Giannotta J, Zaninoni A, et al. Small paroxysmal noctur­ 2021;10:1016/S2665-9913(21)00059–X. [Epub ahead of print.
nal hemoglobinuria clones in autoimmune hemolytic anemia: clin­ 113. Fattizzo B. Evans syndrome and infections: a dangerous cocktail to
ical implications and different cytokine patterns in positive and manage with caution. Blood Transfus. 2021;19(1):5–8.
negative patients. Front Immunol. 2020 Jun 4;11:1006. PMID: 114. Ehmsen S, Asmussen A, Jeppesen SS, et al. Antibody and T cell
32582157; PMCID: PMC7287021. immune responses following mRNA COVID-19 vaccination in
99. Barcellini W, Fattizzo B, Cortelezzi A. Autoimmune hemolytic ane­ patients with cancer. Cancer Cell. 2021;39(8):1034–1036.
mia, autoimmune neutropenia and aplastic anemia in the elderly. 115. Murdych TM. A case of severe autoimmune hemolytic anemia after
Eur J Intern Med. 2018;58:77–83. a receipt of a first dose of SARS-CoV-2 vaccine. Int J Lab Hematol.
100. Rattarittamrong E, Eiamprapai P, Tantiworawit A, et al. Clinical 2021;44(1):e10–e12. Epub ahead of print.
characteristics and long-term outcomes of warm-type autoimmune 116. Brito S, Ferreira N, Mateus S, et al. A case of autoimmune hemolytic
hemolytic anemia. Hematology. 2016;21(6):368–374. anemia following COVID-19 messenger ribonucleic acid
101. Fattizzo B, Zaninoni A, Nesa F, et al. Lessons from very severe, vaccination. Cureus. 2021;13(5):e15035.
refractory, and fatal primary autoimmune hemolytic anemias. Am 117. Pérez-Lamas L, Moreno-Jiménez G, Tenorio-Núñez MC, et al.
J Hematol. 2015;90(8):E149–51. Hemolytic crisis due to Covid-19 vaccination in a woman with
102. Lafarge A, Bertinchamp R, Pichereau C, et al., Prognosis of auto­ cold agglutinin disease. Am J Hematol. 2021;96(8):e288–91.
immune hemolytic anemia in critically ill patients. Ann Hematol. 98 118. Fattizzo B, Giannotta JA, Cecchi N, et al. SARS-CoV −2 vaccina­
(3): 589–594. 2019. tion in patients with autoimmune cytopenias: the experience of
• important evidence of mortality of autoimmune hemolytic a reference center. Am J Hematol. 2021;96(11):e413–6.
anemias in the intensive car unit 119. Simpson CR, Shi T, Vasileiou E, et al. First-dose ChAdOx1 and
103. Hill QA, Horan M, Charlton A, et al. Developing the evidence base BNT162b2 COVID-19 vaccines and thrombocytopenic, thromboem­
for the management of autoimmune haemolytic anaemia (AIHA): bolic and hemorrhagic events in Scotland. Nat Med. 2021;27
the UK experience. Br J Haematol. 2021Jan;1922:e54–7. Epub 2020 (7):1290–1297.
Nov 20. PMID: 33216965 120. Welsh KJ, Baumblatt J, Chege W, et al. Thrombocytopenia including
104. Fattizzo B. Evans syndrome in the SARS-CoV-2 era: “springing up immune thrombocytopenia after receipt of mRNA COVID-19 vac­
like mushrooms.” Blood Transfus. 2021 Dec 1; Epub ahead of print. cines reported to the Vaccine Adverse Event Reporting System
PMID: 34967726. (VAERS). Vaccine. 2021;39(25):3329–3332.

You might also like