Anda di halaman 1dari 11

review

How I manage Evans Syndrome and AIHA cases in children

Maurizio Miano

Clinical and Experimental Haematology Unit, Department of Haematology/Oncology, IRCCS Istituto Giannina Gaslini, Genoa, Italy

direct antiglobulin test (DAT) showing positivity without


Summary
haemolysis.
The management of Evans Syndrome in children is challeng- Front-line treatment is based on steroid administration,
ing due to the lack of evidence-based data on treatment. the first reported therapy (Dameshek et al, 1951), although it
Steroids, the first-choice therapy, are successful in about 80% has never been validated by any clinical trial. Very little data
of cases. For children who are resistant, relapse or become is available on second and later-line approaches for the
steroid-dependent, rituximab is considered a valid second- relapsing/resistant disease, which is a very challenging issue,
line treatment, with the exception of those with an underly- particularly in children who are at risk of severe long-term
ing diagnosis of autoimmune lymphoproliferative syndrome steroid-related side effects. In the last few years, new
who may benefit from other options such as mycophenolate immunosuppressive drugs have provided encouraging results
mofetil and sirolimus. Better knowledge of the immunologi- in controlling the immune cytopenias and/or in acting as
cal mechanisms underlying cytopenias and the availability of steroid-sparing tools.
new immunosuppressive drugs can be helpful in the choice We focus here on the diagnosis and management of chil-
of more targeted therapies that would enable the reduction dren with ES with particular attention to second and third
of the use of long-term steroid administration or other more line treatment.
aggressive options, such as splenectomy or stem cell trans-
plantation. This manuscript provides an overview of the
Incidence and prevalence
pathogenic background of the disease, and suggests a clinical
approach to diagnosis and treatment with a particular focus Data on the incidence and prevalence of ES is scarce and is
on the management of relapsing/resistant disease. essentially derived from cohorts of patients with AIHA in
which cell lineages additional to red cells was involved. AIHA
Keywords: Evans syndrome, autoimmune haemolytic anae- has an estimated incidence of 04 cases/100 000 children per
mia, immune thrombocytopenia, children, autoimmune year. No data is available on the exact prevalence in paedi-
lymphoproliferative syndrome, immunosuppression. atric patients. Between 13% and 73% of patients with AIHA
are reported to have multi-lineage involvement (Pui et al,
Evans Syndrome (ES) is a rare disorder that was initially 1980; Wang, 1988; Mathew et al, 1997). In the largest
described as the presence of autoimmune haemolytic anae- available study of 265 children with AIHA, ES, defined as the
mia (AIHA) and immune thrombocytopenia with unknown involvement of both red cells and platelets, was reported
aetiology (Evans et al, 1951). Over time, the definition of the to account for 37% of cases (Aladjidi et al, 2011). In the
diseases has changed and currently ES is defined as the same study, the mortality rate was reported to be as high
destruction of at least two blood cell lineages in the absence as 10%.
of other diagnoses (Wang, 1988; Mathew et al, 1997; Savasßan
et al, 1997). However, due to the recently acquired knowl- Pathogenesis
edge of the novel immune deregulatory syndromes underly-
ing most cases, ES might also be considered as a Although the diagnosis of ES may not necessarily include the
manifestation of different immune-mediated disorders. Red immune destruction of red cells, AIHA remains the most
blood cell involvement might also be expressed by the pres- important component of the disease. Immune-mediated
ence of laboratory signs of the disease, such as an isolated haemolysis can be driven by different mechanisms. In most
(60–70%) cases (Aladjidi et al, 2011), auto-reactive
immunoglobulin (Ig) G binds red blood cell (RBC) antigens
and causes their destruction, mainly in the extra-vascular
Correspondence: Maurizio Miano, Clinical and Experimental compartment, through the antibody-dependent cellular cyto-
Haematology Unit, IRCCS Istituto Giannina Gaslini, Largo G. toxicity mechanism. More rarely, both IgA and IgM can tar-
Gaslini 5, 16148-Genoa, Italy. get the RBCs as well. The maximal activity of IgG is usually
E-mail: mauriziomiano@gaslini.org at 37°C, and for this reason they are classified as warm

First published online 2 December 2015 ª 2015 John Wiley & Sons Ltd
doi: 10.1111/bjh.13866 British Journal of Haematology, 2016, 172, 524–534
Review

antibodies. In a minority of children (25–30%), IgM destroys Table I. Causes of secondary Evans syndrome and autoimmune hae-
RBCs by activating the complement cascade, mainly in the molytic anaemia in children.
intra-vascular compartment, when exposed to cold tempera- Infections Epstein-Barr virus
tures (cold antibodies) (Petz & Garratty, 2004). Sometimes, Cytomegalovirus
both mechanisms coexist. Immune-mediated thrombocytope- Human Immunodeficiency Virus
nia is due to platelet opsonization with anti-platelet antibod- Helicobacter Pylorii
ies and/or immune-complexes (Cines & Blanchette, 2002; Hepatitis C virus
Mycoplasma pneumoniae
Provan & Newland, 2002) that leads to their premature
Parvovirus B19
removal by the reticuloendothelial system. Peripheral
Immunodeficiencies Common-Variable Immuno-Deficiency
destruction of neutrophils is usually mediated by antibodies and Lymphoproliferative (CVID)
that recognize membrane antigens, mostly located on disorders Severe Combined Immuno-Deficiency
immunoglobulin G Fc receptor type 3b (FccIIIb receptor), (SCID)
(Farruggia & Dufour, 2015). Di George syndrome
Selective IgA deficiency
Autoimmune lymphoproliferative
Secondary forms syndrome (ALPS)
AHIA and ES can be primary or secondary to other diseases, Lipopolysaccharide-responsive and
mainly infections, lymphoproliferative disorders, autoim- beige-like anchor (LRBA) deficiency
Cytotoxic T-Lymphocyte Antigen-4
mune diseases and immunodeficiencies (Table I). The recent
(CTLA4) deficiency
new findings of different genetic defects involving the critical
Phosphoinositide 3-kinase delta
pathways of immune-regulation led to the identification of (PI3KD) mutations
specific disorders, which explain some cases of ES previously Castleman disease
reported as idiopathic. Autoimmune and Systemic lupus erythematosus
Rheumatology Anti-phospholipid syndrome
Autoimmune lymphoproliferative syndrome (ALPS) and other disorders: Rheumatoid arthritis
lymphoproliferative disorders. The first studies with ES Giant-cell hepatitis
patients showed a decreased CD4/CD8 ratio and increased Malignancies Lymphoma
production of interleukin 10 and c-interferon (Wang et al, Leukemia
1983; Karakantza et al, 2000). After the scenario of ALPS Myelodysplasia
Other Drugs
emerged, some patients reported as ES were actually shown
Vaccination
to suffer from ALPS (Teachey et al, 2005), caused by a defect
Stem cell transplantation
of the FAS apoptotic pathway, resulting in abnormal lym-
phocytes survival that leads to lymphoproliferation and
autoimmunity (Bleesing et al, 2002). Cytopenia is the most through a drug-induced hypersensitivity or through its role
common manifestation of autoimmunity and may also fea- in the transcription of lymphokine genes (Clipstone & Crab-
ture the first sign of the disease. ALPS patients are character- tree, 1992).
ized by chronic lymphoproliferation, an increased number of Incomplete immune reconstitution or immune dysregula-
a specific T cell population, termed ‘double negative’ T-cells, tion may also be the cause of ACs occurring after stem cell
and an increased risk of developing malignancies. transplantation (SCT). In the largest report on post-SCT ACs
Similar clinical features overlapping with more specific in children, the use of alternative donors was reported to be
symptoms have been recently described in other disorders associated with a higher incidence, probably due to dysregu-
caused by different genetic defects involving other crucial lation caused by anti-thymocyte globulin and alemtuzumab
pathways of lymphocytes activation and survival, such as the given in the conditioning regimen. Patients who underwent
cytotoxic T-lymphocyte antigen-4 (CTLA4), lipopolysaccha- SCT because of a non-malignant disorder were also found to
ride-responsive deige-like anchor deficiency (LRBA), phos- have a higher incidence of ACs (Faraci et al, 2014).
phoinositide 3-kinase delta (PI3KD) mutations, or CTLA4
deficiencies (Seidel, 2014). Human immunodeficiency virus (HIV). Thanks to antiviral
treatment, nowadays HIV-related AC is no longer a major
Post-transplant forms. Cases of ES and AHIA have been problem in children. Although an overt haemolysis is very
reported after both solid organ and stem cell transplanta- rare, a positive DAT has been reported in 20–40% of
tions. In the first cases, autoimmune cytopenias (ACs) occur HIV-infected patients (Toy et al, 1985). This high incidence
in the early post-transplant phases, often following viral may be related to abnormal B-cell regulation by the infected
infections. In these particular patients, alloimmunity seems T-cells, a direct activation of B-cells by the virus, or an
to play an important role in the pathogenesis, as does tacro- abnormal response to other viruses or other HIV-associated
limus-based immunosuppression, which may act either opportunistic agents (Saif, 2001).

ª 2015 John Wiley & Sons Ltd 525


British Journal of Haematology, 2016, 172, 524–534
Review

‘warm’ IgM or IgA, which should be identified by


Clinical presentation
monospecific anti-IgA sera or by low-affinity antibodies,
Anaemia is usually normocytic or macrocytic, often hyper- mediates haemolysis. In all these cases, the exclusion of other
chromic, and may develop gradually or quickly, depending non immune-mediated causes of haemolysis is strongly rec-
on the rate of haemolysis and on the efficacy of compen- ommended and, if thrombocytopenia is also present, throm-
satory response of the bone marrow. The severity of the dis- botic thrombocytopenic purpura and atypical haemolytic
ease can thus vary from mild to acutely life-threatening uraemic syndrome should be excluded by investigating
forms. Symptoms usually include pallor, fatigue, dyspnoea, ADAMS13 activity and complement levels.
tachycardia and often fever. Hepato-splenomegaly, jaundice About 40% of children (Aladjidi et al, 2011) can present
and haematuria/hemoglobinuria may also be present. The with a normal/low count of reticulocytes (Liesveld et al,
most frequent association with AIHA is thrombocytopenia, 1987; Jastaniah et al, 2004) that can also be the target of
which may present with petechiae, bruising and muco-cuta- the self-directed RBC antibodies. Earlier intra-medullary
neous bleeding. The association may either develop concomi- precursors may also be involved through the destruction
tantly or separately after a median interval of 3 years process thus explaining cases that can initially present as
(Aladjidi et al, 2011). For this reason, the diagnosis of ES is pure red cell anaemia (Miano et al, 2014a). Although the
challenging and should also be potentially considered at the sensitiveness of anti-platelets and anti-neutrophils is very
onset of a unilineage cytopenia, especially in cases with sev- low, their identification can be helpful to confirm the
ere symptoms and/or in the case of refractory/resistant dis- immunological background of thrombocytopenia and neu-
ease. Neutropenia can be asymptomatic or present through tropenia.
infection-related symptoms. Its combination with other ACs Bone marrow aspirate is not considered essential for the
(for example thrombocytopenia and neutropenia), although diagnosis of AIHA, as it usually presents normal features or
less frequent, is still compatible with the phenotypical diag- increased cellularity (Pui et al,1980; Mathew et al, 1997).
nosis of ES. Nonetheless, it should be performed in all cases with reticu-
locytopenia, and in ES, in order to exclude a proliferative
disorder or myelodysplasia. In very young children with neu-
Diagnosis
tropenia, bone marrow can also be helpful to exclude a con-
A careful patient and family history of malignancies, infec- genital neutropenia that usually shows a maturation arrest at
tions and immunological disorders is important to identify the promyelocyte stage. An immunological work-up at diag-
other underlying diseases. It is also useful to identify nosis is highly recommended, and should include the levels
recent exposure to drugs or vaccinations that might trigger of IG with subclasses, screening for other auto-immune dis-
the cytopenia. Clinical examination should focus on the eases, and lymphocytes subset count with double-negative
search for lymphadenopathy and hepato-splenomegaly. T-cells, in order to detect other underlying disorders that
Although the most recent definition (Wang, 1988; Mathew might be masked by the subsequent immunosuppressive
et al, 1997; Savasßan et al, 1997) also enables a diagnosis of treatment. Serological and molecular search for previous
ES in the absence of immune destruction of red cells, and/or on-going infections is also necessary at the onset of
AIHA is an important element for the diagnosis of the the disease to identify any infective trigger. It is of note that
disease that should also be looked for in less clinically evi- severe acute onset, chronic/relapsing phenotypes and ES as
dent cases. an expression of multi-lineage cytopenia, are more frequently
The diagnosis of an immune-mediated haemolysis is based associated with secondary forms and may represent an
on DAT positivity, reticulocytosis, increased indirect hyper- epiphenomenon of an underlying immune disorder (Teachey
bilirubinaemia, increased lactate dehydrogenase (LDH) level & Lambert, 2013).
and reduced serum haptoglobin. Sometimes, DAT may be It is worth noting that, thanks to more advanced diagnos-
the only sign of RBC involvement. A peripheral blood film tic tools and more prompt consideration by clinicians, about
may show spherocytes that suggest the presence of extravas- half of the patients that show the clinical phenotype of ES
cular haemolysis and/or aggregations of RBC in long chains are nowadays recognized to have an underlying ALPS (Tea-
(‘rouleaux’). Artifactual macrocytosis due to RBC agglutina- chey et al, 2005; Seif et al, 2010). For the other patients, the
tion can also be present. In the event of a warm antibody- search for gene defects related to recently identified novel
AIHA (WA-AIHA), DAT is usually IgG+ or IgG/C3d+. immunodysregulatory syndromes and known to underlie
Patients with cold antibody-AIHA usually have a negative or cases of ES should also be considered, using, when available,
C3d+ DAT, however, a negative DAT should not exclude the both traditional DNA assays or more modern techniques,
diagnosis of WA-AIHA, as about 10% of patients (Petz & such as next generation sequencing or whole exome sequenc-
Garratty, 2004) may have an undetectably low number of Ab ing.
molecules on the RBC surface thus requiring a more sophis- Tables II and III show my suggested diagnostic work-up
ticated laboratory work-up. DAT can also be negative when and differential diagnosis for patients with ES and AIHA.

526 ª 2015 John Wiley & Sons Ltd


British Journal of Haematology, 2016, 172, 524–534
Review

Table II. Diagnostic work up for Evans syndrome and autoimmune Table III. Differential diagnosis of autoimmune haemolytic anaemia
haemolytic anemia. and Evans syndrome.

Haematological evaluation: Autoimmune Non immune hereditary red-cell membrane


haemolytic disorders
• Full Blood Count* anaemia Erythrocyte enzyme deficiency
• Reticulocytes* Dyserythropoietic anaemia
• Blood smear*,† Haemoglobinopathies
• Blood group* Wilson disease
• Red cell antigen typing including: C, c, D, E, e, K, Jka, Jkb, Fya, Paroxysmal nocturnal haemoglobinuria
Fyb, S, s Monoclonal lymphoproliferation
• Haemolysis indexes (LDH, haptoglobin, bilirubin)* Evans Acquired thrombotic thrombocyotpenic
• Direct and Indirect Antiglobulin Tests* syndrome purpura
• BUN, creatinine ALT, AST, cGT, C-reactive protein* Inherited ADAMTS13 deficiency
• PTT, Fibrinogen, d-Dimer Haemolytic uraemic syndrome
• Bone marrow aspirate (if reticulocytopenia and/or ES) Kasabach-Merrit syndrome
Infective screening: Disseminated intravascular coagulation
Monoclonal lymphoproliferation
• HIV, HCV, HBV serological evaluation†
• EBV, CMV, Parvovirus B19, serological † and molecular
evaluation Mathew et al, 1997; Meyer et al, 1997; Naithani et al, 2007;
• Helicobacter Pylori
Ware, 2009). This approach induces remission in 80–85% of
Immunological work-up: patients with most children responding to 1–2 mg/kg/d. I
start with 1–2 mg/kg prednisolone and give higher doses
• Lymphocytes subsets: CD3*, CD4*, CD19*, CD16*, CD56*, B (4–6 mg/kg/d for the first 72 h) in the case of severe clinical
naive (CD19+IgD+CD27 ), B memory (CD19+CD27+) symptoms in patients with more aggressive forms. Although
• Immunoglobulin serum levels*,†
remission is obtained in most cases, the risk of relapse is
• IgG subclasses (patients aged >2 years)†
• C3, C4, CH50 high, especially during the tapering off. This is why a full
• Autoantibodies: anti-nuclear, anti-extractable nuclear, dose of steroid should be given for at least 3–4 weeks.
anti-DNA, anti-phospholipid, anti-Sm, anti-TG, anti-TPO† Patients should be evaluated after 3 weeks to determine
• ALPS screening: double-negative T-cells, Vitamin B12, whether to continue full dosage for another week in respond-
IL10, IL18, circulating FAS, Fas-apoptosis functional test ing subjects and then slowly taper off over not less than
Radiological evaluation: 6 months. In patients who show a partial response after the
first 3 weeks, the full dosage might be given for 2 further
• Chest X-ray weeks, however, children who do not show any response
• Abdominal sonography (spleen, liver, lymph nodes) after 3 weeks probably require a different strategy and should
be shifted to a second-line treatment. Another reported
ES, Evans syndrome; LDH, lactate dehydrogenase; BUN, blood urea scheme includes mega-doses of methylprednisolone: 30 mg/
nitrogen; ALT, alanine aminotransferase; AST, aspartate aminotrans- kg/d for 3 d, 20 mg/kg/d for the following 4 d and then
ferase; cGT, gamma glutamyl transpeptidase; PTT, partial thrombo- tapering by half of the dose each week (Meytes et al, 1985;
plastin time; HIV, Human Immunodeficiency virus; HCV, Hepatitis
Nanan et al, 1995; Ozsoylu, 2004).
C virus; HBV, Hepatitis B virus; EBV, Epstein–Barr virus; CMV,
Packed red blood cell (PRBC) transfusions should be
Cytomegalovirus; anti-TG, anti-thyroglobulin; anti-TPO, anti-thyro-
peroxidase; ALPS; Autoimmune lymphoproliferative syndrome; IVIG,
reserved for very symptomatic patients, who may suffer from
intravenous immunoglobulin; IL, interleukin. life-threatening events. In fact, transfused PRBC may be
*Mandatory first-line evaluation. destroyed by the auto-antibody, resulting in a further hyper-
†Before transfusion/IVIG administration. activation of the abnormal immunological process. The deci-
sion to transfuse should be driven by the aim to correct the
clinical symptoms, rather than the haemoglobin level. In fact,
First-line treatment
the usual wide range of auto-antibody reactivity makes it dif-
The aim of the treatment is to control cytopenias, of which ficult to find a truly compatible donor, and the choice of the
AIHA is by far the most frequent expression, and to mini- ‘best available’ matched unit should be carefully made on the
mize toxicity. basis of extensive RBC phenotyping. Small (3 ml/kg)
Steroids are the first-choice treatment, although the first amounts of leucodepleted PRBC should be given slowly
successful experiences (Dameshek et al, 1951; Pui et al, 1980; under careful supervision. In the case of life-threatening
Wang, 1988) have never been validated by any clinical trial. events not responding to transfusions, plasma-exchange may
Patients are usually treated with prednisolone at a dose of be an option to remove circulating antibodies that, however,
1–6 mg/kg/d (Allgood & Chaplin, 1967; Pui et al, 1980; do not look fully established. In this respect, because little

ª 2015 John Wiley & Sons Ltd 527


British Journal of Haematology, 2016, 172, 524–534
Review

data is available (McCarthy et al, 1999; Roy-Burman & In the absence of prospective trials and based on the
Glader, 2002; Beretta et al, 2009; Lucchini et al, 2009), the above data, at Istituto Giannina Gaslini, rituximab is pre-
American Society for Apheresis (ASFA) states that its optimal ferred to splenectomy as second line treatment for children
role is not established for WA-AHIA (category III) even if it with AHIA due to the higher risk of infective complications
may have some efficacy in cold-AHIA (Category II) following this practice in younger children. The time to
(Schwartz et al, 2013). Therefore, the cost/benefit ratio of response to the treatment, described as a median of
this procedure should also be considered, especially for small 3–6 weeks but also after up to 12 weeks, must be considered
weighted children. Platelet transfusions should be reserved for the eventual choice to temporarily overlap the steroid
only for cases with life-threatening haemorrhages. In cases of treatment according to the severity of the disease. In patients
severe neutropenia, granulocyte colony-stimulating factor with ES, however, the use of rituximab as a second-line
(5 lg/kg/dose) might be used ‘on demand’ if severe infec- option should be carefully weighed, taking into consideration
tions coexist, but only for the duration of the infection a potential underlying diagnosis of ALPS. Indeed, ALPS
(Fioredda et al, 2012). patients were reported to have lower response rates, higher
risk of infection and prolonged hypogammaglobulinaemia
(Teachey et al, 2009; Rao & Oliveira, 2011) after this treat-
Second and further-line treatment
ment, which should therefore be limited to severe forms
Most children with ES experience a relapsing/resistant requiring a strong approach (Rao & Oliveira, 2011).
cytopenia, or a chronic disease, often requiring prolonged Nonetheless, careful monitoring of B-cells and immunoglob-
treatment with steroids, which may lead to severe side effects ulin levels after treatment should be performed in all patients
on the bone and the endocrine systems. Before the introduc- receiving intravenous immunoglobulin (IVIG) until their
tion of new therapeutic options, splenectomy represented the recovery. Apart from ALPS patients, severe B-cell depletion
only choice for these patients. Due to its risks, this option and the consequent need for long-term IVIG replacement
should be now considered only after the failure of other mainly occurs in patients undergoing repeated doses and/or
alternative drugs available for refractory patients and should in the setting of other underlying immune-disorders.
not be performed in patients with ALPS (Rao, 2015). The (Cooper et al, 2009).
choice of the most appropriate second/further line treatment The most important side-effect of rituximab is multifocal
should take into consideration the immunological back- leucoencephalopathy, which has often been described in rela-
ground of the disease and the severity of clinical symptoms. tion with John Cunningham virus (JCV) infection in
Another crucial point is the time required to achieve efficacy, immunocompromised patients (Bellizzi et al, 2013). How-
which has to be short in severe and life-threatening events, ever, this complication is very rare in children, perhaps due
but could be more delayed in the case of steroid-dependency to the lack of exposure to the virus, and has been mostly
or when a maintenance treatment is indicated. Monoclonal reported after chemotherapy and SCT or in patients with
antibodies, multi-agent approaches including chemotherapy systemic lupus erythematosus (Carson et al, 2009).
and splenectomy can be used more as ‘attack’ therapy to
obtain a fast response. Immunosuppressive agents, such as
Mycophenolate mofetil
mycophenolate mofetil (MMF), sirolimus and ciclosporin,
can be more suitable for response maintenance or as steroid- MMF is an inhibitor of inosine monophosphate dehydroge-
sparing tools. nase in purine synthesis that targets B, T and NK cells. It has
been shown to be effective in AIHA and ITP in some retro-
spective studies on adult patients that occasionally included
Rituximab
children (Howard et al, 2002; Hou et al, 2003; Zhang et al,
Rituximab is a monoclonal antibody against the CD20 mole- 2005; Provan et al, 2006). In those studies, response rates
cule that causes B-cell depletion and is usually given at a ranged between 62% and 82%. Other anecdotal studies
dose of 375 mg/m2 on days 1, 8, 15 and 22. Most available reported efficacy in children with ES (Guirat-Dhouib et al,
data comes from retrospective studies of patients with AHIA 2010) and ALPS-associated cytopenia (Rao et al, 2005). In a
(Quartier et al, 2001; Zecca et al, 2003; Garvey, 2008; Bus- recent study of primary and secondary AC (Miano et al,
sone et al, 2009; Svahn et al, 2009; Pe~
nalver et al, 2010; Bar- 2015), MMF was safe and effective, mainly as a second line
cellini & Zanella, 2011) and immune thrombocytopenic treatment, in all 11 patients with ES associated with an
purpura (ITP) (Bennett et al, 2006; Patel et al, 2012) with underlying diagnosis of ALPS (n = 9) or ‘ALPS-related’ dis-
overall response rates between 77–93% and 39–68%, respec- order (n = 2), defined as the presence of at least one absolute
tively. Experience in ES patients is limited to case reports or primary additional diagnostic criterion for ALPS (Oliveira
and to a series of adults (Michel et al, 2009) and children et al, 2010), suggesting a potential role for this drug in
(Bader-Meunier et al, 2007) where rituximab, alone or in patients with ES who may suffer from a broader underlying
combination with steroids, showed a response rate of 82% immunological disorder than ALPS. I use MMF at a dose of
and 75%, respectively. 600 mg/m2 twice a day (maintaining serum levels between

528 ª 2015 John Wiley & Sons Ltd


British Journal of Haematology, 2016, 172, 524–534
Review

1–35 lg/ml) and evaluate the response after 3 months. Due and in those who need a steroid-sparing/maintenance treat-
to the risk of relapse after discontinuation and the absence of ment.
data on MMF long-term toxicity, the treatment is continued
in responding children, with a full dose for a total of 2 years,
Ciclosporin
then the drug is tapered off over 6 months before stopping.
Although further studies are required to understand its Ciclosporin has been used at a dose of 5 mg/kg as an alter-
real efficacy and safety, current results seem to suggest that native option in adults and children with refractory isolated
MMF may play a role as second/third-line treatment. Given AIHA (D€ undar et al, 1991) or ES (Emilia et al, 1996; Ucßar
that MMF needs rather a long time to generate a response, et al, 1999; Williams & Boxer, 2003). Ciclosporin has also
ideally it should be overlapped early with more aggressive been used as an alternative treatment in the context of a
therapy, such as high-dose steroids or rituximab, to enable multi-agent approach including danazol (Scaradavou & Bus-
the achievement of therapeutic plasma levels, and given as a sel, 1995) or in protocols including weekly vincristine and
‘maintenance treatment’. This scheme seems to be applicable methylprednisone.
to relapsing patients, steroid-dependent children requiring Due to the side effects and the need for frequent clinical
drug sparing or those who experience flare-up episodes dur- and serum level monitoring, ciclosporin alone might be an
ing tapering. In general, given the good results in patients option as a steroid-sparing/maintenance treatment only after
with ALPS (Rao et al, 2005; Teachey et al, 2009; Rao & Oli- the failure of newer and more tolerable agents, such as MMF
veira, 2011; Miano et al, 2015) and the risk of rituximab- and sirolimus. Its use in the context of a multi-agent inten-
related complications, I administer MMF to children with ES sive protocol could be considered in severe refractory cases
secondary to ALPS in a more up-front approach soon after only, after failure of a more targeted option, such as
the failure of first-line treatment. rituximab.

Sirolimus Other options


Sirolimus is an inhibitor of the mammalian (mechanistic) IVIG. Although the role of IVIG for patients with ITP has
target of rapamycin (mTOR), which is known to increase been clearly established, their use in the setting of AIHA and
T-regulatory cells (T-regs) and to induce apoptosis in abnor- ES is still controversial and little data is available for these
mal lymphocytes (Zhan et al, 2013). The drug has been used diseases. Their potential efficacy seems to be related to the
for over 20 years (Hartford & Ratain, 2007) in the setting of saturation of Fc phagocyte receptors in the reticuloendothe-
autoimmune diseases (Brusko et al, 2008) and solid organ lial system. A few reports have been published on their use
transplantation (Karim & Giles, 2008) and has a safe profile. in AIHA at the dose of 04–05 g/kg for 4–5 d, concomi-
Very limited data is available on sirolimus use in ES and tantly with steroids or after their failure (Bussel & Hilgartner,
most information comes from AIHA and ALPS settings. Its 1984; Oda et al, 1985; Otheo et al, 1997). The largest retro-
mechanism of action accounts for successful results in post- spective report on 73 patients with AIHA showed around
transplant AIHA (Valentini et al, 2006; Acquazzino et al, 40% of responses, which reached 55% in children (Flores
2013; Loar et al, 2013) and in some paediatric patients with et al, 1993). The recently published Canadian guidelines do
primary refractory AIHA (Miano et al, 2014a,b). Sirolimus not recommend the routine use of IVIG in patients with
has also been described as useful in ALPS patients with or AIHA and suggest their use only in patients with severe dis-
without cytopenia (Teachey et al, 2009) in which it also ease, however, in cases of symptomatic thrombocytopenia,
reduced the count of double-negative T-cells. In a recent IVIG are a recommended option (Anderson et al, 2007).
study from my institution (Miano et al, 2015), sirolimus was Based on this evidence and on the low risk of severe side-
effective in all five children with refractory AIHA, in 6/10 effects, IVIG may be a complementary tool that can be added
with ITP, and also in the only patient affected with ES. All to treatment in severe forms where thrombocytopenia domi-
these children previously failed MMF, suggesting a potential nates the clinical scenario.
role for sirolimus in patients with primary refractory cytope-
nias or with underlying defects other than ALPS. Splenectomy. Splenectomy has been considered the second-
At the Istituto Giannina Gaslini, sirolimus is given at the line option for the treatment of ES for many years, due to
dose of 2–3 mg/m2 once a day, maintaining serum levels the good response and short-term efficacy. No data is avail-
between 4–12 ng/ml, with an optimal target of 9 ng/ml, for able on its efficacy compared to newer approaches, such as
at least 3 months before evaluating its efficacy. As with rituximab. In the largest published cohort on AIHA in chil-
MMF, I continue the treatment in responders for a total of dren (Aladjidi et al, 2011), splenectomy was performed in
2 years followed by another 6 months of tapering off before 139% of cases. Although it is difficult to evaluate its real
finally stopping. On the basis of the available data so far, efficacy, children seem to have a better response than adults
sirolimus may be used as a rescue treatment after MMF mainly because of a reduced need for steroid treatment.
failure in patients with chronic/refractory ES and AIHA, Splenectomy-related complications are still a crucial consid-

ª 2015 John Wiley & Sons Ltd 529


British Journal of Haematology, 2016, 172, 524–534
Review

eration for the choice of the right treatment, although in 1–2 mg/kg for 2–3 months (Oda et al, 1985; Wang, 1988;
recent years they have been significantly reduced thanks to Gombakis et al, 1999) have proved to be effective. Higher i.v.
the introduction of both laparoscopy, which has diminished doses (200 mg/kg in 4 d) without stem-cell rescue were also
the surgical risk compared to traditional surgery (05–16% given in patients with AIHA, ITP and ES (Brodsky et al, 1998;
vs. 6%) (Casaccia et al, 2006), and pre-operative vaccination Moyo et al, 2002) with some partial remissions reported.
(American Academy of Pediatrics, 2000), which has damp-
ened the incidence of overwhelming infections (Pilishvili Alemtuzumab. Alemtuzumab is a humanized anti-CD52
et al, 2010) previously reported in 33–5% of cases (Bisharat monoclonal antibody that targets T and B lymphocytes,
et al, 2001; Davidson & Wall, 2001). Mortality due to post- monocytes and eosinophils. It has been successfully used in
splenectomy sepsis remains an important problem, however, some reported adult and paediatric patients with AIHA or
and can be as high as 50% (Rubin & Schaffner, 2014). For AC (Willis et al, 2001; Ru & Liebman, 2003; Cheung et al,
this reason, the use of prophylactic antimicrobial therapy is 2006; Miano et al, 2014a) at a dose of 10 mg/m2/d for 10 d,
recommended, especially in younger children. Moreover, or of 02 mg/kg for 5 d. After the initial response, most
despite the quick response, splenectomy has proven to patients experience relapse. For this reason, I see some space
induce a rather short term (few months) remission (Coon, for alemtuzumab in severe forms of ES after failure of ritux-
1985) in patients with ES (Mathew et al, 1997) and it is not imab, who, however, might need for further maintenance
recommended in children under 6 years of age, due to treatment.
immaturity of the immune system and incomplete vaccina-
tion programmes. Overall, based on the data described
New drugs
above, splenectomy should be considered after the failure of
other treatments in patients with severe forms who require a Thrombopoietin receptor agonists. In the last few years, romi-
prompt response. plostim and eltrombopag have been increasingly used in
some haematological disorders. Reports of sustained remis-
Cyclophosphamide. Little data is available on the use of sion after thrombopoietin receptor agonist discontinuation
cyclophosphamide in ES. First experiences, at an oral dose of 
in adults (Cervinek et al, 2015) and children (K€ uhne, 2015)

Fig 1. Therapeutic approach to patients with Evans syndrome at the Istituto Giannina Gaslini. CR, complete remission; PR, partial remission;
NR, no repsonse; IVIG, intravenous immunoglobulin; MMF, mycophenolate mofetil; ALPS, Autoimmune Lymphoproliferative Syndrome.

530 ª 2015 John Wiley & Sons Ltd


British Journal of Haematology, 2016, 172, 524–534
Review

suffering from ITP have been reported, and encouraging overall transplant-related mortality of 15%. SCT should be
results have been published in some ES children, (Pasquet taken into consideration for patients with relapsing/resistant
et al, 2014; Seidel et al, 2014). Its use in ES remains anecdo- disease who experience life-threatening events.
tal, however, and might be clarified by future studies.
Conclusion
Bortezomib. Bortezomib is an inhibitor of 26S proteasome
that has been successfully used in AC following SCT (Waespe The management of ES is challenging due to the lack of evi-
et al, 2014) and in a case of IgM-driven AIHA secondary to dence-based data on treatment. An accurate immunological
a monoclonal gammopathy (Carson et al, 2010). Seven chil- work-up is necessary in all newly diagnosed children in order
dren with primary, secondary or post-transplant AC, includ- to promptly identify the underlying disorders that may
ing one with ES, treated with bortezomib, have also been require specific treatment. After the failure of up-front treat-
recently reported (Khandelwal et al, 2014) and a clinical trial ment with steroids, I give rituximab to patients with ES,
is on-going in the setting of post-transplant AC (ClinicalTri- apart from children with an underlying diagnosis of ALPS
als.gov identifier: NCT01930253). The idea to target plasma with no severe phenotype for whom I would rather suggest
cells in a context where an antibody-mediated attack cannot the use of MMF and sirolimus. In my opinion, these drugs
be controlled by anti-CD20 may be based on the speculation may also be useful as a maintenance treatment in patients
that a cluster of plasma cells not targetable by previous treat- responding to rituximab or in the case of steroid-dependent
ments might be a cause of the persistence of the disease. disease.
Further alternative treatments should be chosen on a case-
by-case basis, taking into consideration the possible underly-
Stem cell transplantation
ing disorder and the response to the previous therapies. In
There is little data, and mainly from small series/case reports, fact, a partial/short response to anti-CD20 would prompt the
on both autologous and allogeneic haematopoietic SCT in concomitant involvement of a T-cell component in the
patients with ES and AIHA. Overall, these studies reported pathogenesis and then I would suggest a T-cell targeting
complete remission of around 50% (Petz & Garratty, 2004; drug. Figure 1 shows the therapeutic approach adopted at
Urban et al, 2006). Passweg and Rabusin (2008) published a my institution for ES, which is inevitably based on data from
study on a cohort of patients with AC from the European retrospective studies and therefore should be regarded with
Society of Blood and Marrow Transplantation (EBMT) data- caution. The need to obtain evidence-based treatment data
base, which included 7 patients with ES and 7 with AIHA. urgently requires the design of international clinical trials
Four out of 14 achieved a continuous remission with an that would enable stronger indications to be derived.

References coccal infections, including the use of cephalopathy. Clinical and Developmental
pneumococcal conjugate vaccine (Prevnar), Immunology, 2013, 839719. Epub 2013 Apr 17.
Acquazzino, M.A., Fischer, R.T., Langnas, A. & pneumococcal polysaccharide vaccine, and Bennett, C.M., Rogers, Z.R., Kinnamon, D.D., Bus-
Coulter, D.W. (2013) Refractory autoimmune antibiotic prophylaxis. Pediatrics, 106(2 Pt 1), sel, J.B., Mahoney, D.H., Abshire, T.C., Sawaf,
hemolytic anemia after intestinal transplant 362–366. H., Moore, T.B., Loh, M.L., Glader, B.E.,
responding to conversion from a calcineurin to Anderson, D., Ali, K., Blanchette, V., Brouwers, McCarthy, M.C., Mueller, B.U., Olson, T.A.,
mTOR inhibitor. Pediatric Transplantation, 17, M., Couban, S., Radmoor, P., Huebsch, L., Lorenzana, A.N., Mentzer, W.C., Buchanan,
466–471. Hume, H., McLeod, A., Meyer, R., Moltzan, C., G.R., Feldman, H.A. & Neufeld, E.J. (2006)
Aladjidi, N., Leverger, G., Leblanc, T., Picat, M.Q., Nahirniak, S., Nantel, S., Pineo, G. & Rock, G. Prospective phase 1/2 study of rituximab in
Michel, G., Bertrand, Y., Bader-Meunier, B., (2007) Guidelines on the use of intravenous childhood and adolescent chronic immune
Robert, A., Nelken, B., Gandemer, V., Savel, H., immunoglobulin for hematologic conditions. thrombocytopenic purpura. Blood, 107, 2639–
Stephan, J.L., Fouyssac, F., Jeanpetit, J., Thomas, Transfusion Medicine Reviews, 21, S9–S56. 2642.
C., Rohrlich, P., Baruchel, A., Fischer, A., Bader-Meunier, B., Aladjidi, N., Bellmann, F., Beretta, C., Leoni, V., Rossi, M.R., Jankovic, M.,
Ch^ene, G., Perel, Y.; Centre de Reference Monpoux, F., Nelken, B., Robert, A., Armari- Patronati, N., Foti, G. & Biagi, E. (2009) Pro-
National des Cytopenies Auto-immunes de Alla, C., Picard, C., Ledeist, F., Munzer, M., longed extracorporeal membrane oxygenation
l’Enfant (CEREVANCE). (2011) New insights Yacouben, K., Bertrand, Y., Pariente, A., therapy for severe acute respiratory distress syn-
into childhood autoimmune hemolytic anemia: Chausse, A., Perel, Y. & Leverger, G. (2007) drome in a child affected by rituximab-resistant
a French national observational study of 265 Rituximab therapy for childhood Evans syn- autoimmune hemolytic anemia: a case report.
children. Haematologica, 96, 655–663. drome. Haematologica, 92, 1691–1694. Journal of Medical Case Reports, 3, 6443.
Allgood, J.W., Chaplin, H. Jr. (1967) Idiopathic Barcellini, W. & Zanella, A. (2011) Rituximab Bisharat, N., Omari, H., Lavi, I. & Raz, R. (2001)
acquired autoimmune hemolytic anemia. A therapy for autoimmune haematological dis- Risk of infection and death among post-sple-
review of forty-seven cases treated from 1955 eases. European Journal of Internal Medicine, 22, nectomy patients. Journal of Infection, 43, 182–
through 1965. The American Journal of Medicine, 220–229. 186.
43, 254–257. Bellizzi, A., Anzivino, E., Rodio, D.M., Palamara, Bleesing, J.J., Brown, M.R., Novicio, C., Guarraia,
American Academy of Pediatrics. Committee on A.T., Nencioni, L. & Pietropaolo, V. (2013) New D., Dale, J.K., Straus, S.E. & Fleisher, T.A.
Infectious Diseases. (2000) Policy statement: rec- insights on human polyomavirus JC and patho- (2002) A composite picture of TcR alpha/beta
ommendations for the prevention of pneumo- genesis of progressive multifocal leukoen- (+) CD4( )CD8( ) T Cells (alpha/beta-

ª 2015 John Wiley & Sons Ltd 531


British Journal of Haematology, 2016, 172, 524–534
Review

DNTCs) in humans with autoimmune lympho- Coon, W.W. (1985) Splenectomy in the treatment Garvey, B. (2008) Rituximab in the treatment of
proliferative syndrome. Clinical Immunology, of hemolytic anemia. Archives of Surgery, 120, autoimmune haematological disorders. British
104, 21–30. 625–628. Journal of Haematology, 141, 149–169.
Brodsky, R.A., Petri, M., Smith, B.D., Seifter, E.J., Cooper, N., Davies, E.G. & Thrasher, A.J. (2009) Gombakis, N., Trahana, M., Athanassiou, M. &
Spivak, J.L., Styler, M., Dang, C.V., Brodsky, I. Repeated courses of rituximab for autoimmune Kanakoudi-Tsakalidou, F. (1999) Evans syn-
& Jones, R.J. (1998) Immunoablative high-dose cytopenias may precipitate profound hypogam- drome: successful management with multi-agent
cyclophosphamide without stem-cell rescue for maglobulinaemia requiring replacement intra- treatment including intermediate-dose intra-
refractory, severe autoimmune disease. Annals of venous immunoglobulin. British Journal of venous cyclophosphamide. Journal of Pediatric
Internal Medicine, 129, 1031–1035. Haematology, 146, 120–122. Hematology/Oncology, 21, 248–249.
Brusko, T.M., Putnam, A.L. & Bluestone, J.A. Dameshek, W., Rosenthal, M.C. & Schwartz, L.I. Guirat-Dhouib, N., Mellouli, F., Kouki, R. &
(2008) Human regulatory T cells: role in (1951) The treatment of acquired hemolytic Bejaoui, M. (2010) Successful treatment of
autoimmune disease and therapeutic opportuni- anemia with adrenocorticotrophic hormone mycophenolate mofetil in a child with refractory
ties. Immunological Reviews, 223, 371–390. (ACTH). The New England Journal of Medicine, Evans syndrome. Journal of Pediatric Hematol-
Bussel, J.B. & Hilgartner, M.W. (1984) The use 244, 117–127. ogy/Oncology, 32, e244.
and mechanism of action of intravenous Davidson, R.N. & Wall, R.A. (2001) Prevention Hartford, C.M. & Ratain, M.J. (2007) Rapamycin:
immunoglobulin in the treatment of immune and management of infections in patients with- something old, something new, sometimes bor-
haematologic disease. British Journal of Haema- out a spleen. Clinical Microbiology and Infection, rowed and now renewed. Clinical Pharmacology
tology, 56, 1–7. 7, 657–660. and Therapeutics, 82, 381–388.
Bussone, G., Ribeiro, E., Dechartres, A., Viallard, D€undar, S., Ozdemir, O. & Ozcebe, O. (1991) Hou, M., Peng, J., Shi, Y., Zhang, C., Qin, P.,
J.F., Bonnotte, B., Fain, O., Godeau, B. & Michel, Cyclosporin in steroid-resistant auto-immune Zhao, C., Ji, X., Wang, X. & Zhang, M. (2003)
M. (2009) Efficacy and safety of rituximab in haemolytic anaemia. Acta Haematologica, 86, Mycophenolate mofetil (MMF) for the treat-
adults’ warm antibody autoimmune haemolytic 200–202. ment of steroid-resistant idiopathic thrombocy-
anemia: retrospective analysis of 27 cases. Ameri- Emilia, G., Messora, C., Longo, G. & Bertesi, M. topenic purpura. European Journal of
can Journal of Hematology, 84, 153–157. (1996) Long-term salvage treatment by cyclos- Haematology, 70, 353–357.
Carson, K.R., Evens, A.M., Richey, E.A., Haber- porin in refractory autoimmune haematological Howard, J., Hoffbrand, A.V., Prentice, H.G. &
mann, T.M., Focosi, D., Seymour, J.F., Laubach, disorders. British Journal of Haematology, 93, Mehta, A. (2002) Mycophenolate mofetil for the
J., Bawn, S.D., Gordon, L.I., Winter, J.N., Fur- 341–344. treatment of refractory auto-immune haemolytic
man, R.R., Vose, J.M., Zelenetz, A.D., Mamtani, Evans, R.S., Takahashi, K., Duane, R.T., Payne, R. anaemia and auto-immune thrombocytopenia
R., Raisch, D.W., Dorshimer, G.W., Rosen, S.T., & Liu, C. (1951) Primary thrombocytopenic purpura. British Journal of Haematology, 117,
Muro, K., Gottardi-Littell, N.R., Talley, R.L., purpura and acquired hemolytic anemia; evi- 712–715.
Sartor, O., Green, D., Major, E.O. & Bennett, dence for a common etiology. AMA Archives of Jastaniah, W.A., Pritchard, S.L., Wu, J.K. & Wads-
C.L. (2009) Progressive multifocal leukoen- Internal Medicine, 87, 48–65. worth, L.D. (2004) Hyperuricemia and reticulo-
cephalopathy after rituximab therapy in HIV- Faraci, M., Zecca, M., Pillon, M., Rovelli, A., Men- cytopenia in association with autoimmune
negative patients: a report of 57 cases from the coni, M.C., Ripaldi, M., Fagioli, F., Rabusin, M., hemolyticanemia in two children. American
Research on Adverse Drug Events and Reports Ziino, O., Lanino, E., Locatelli, F., Daikeler, T. Journal of Clinical Pathology, 122, 849–854.
project. Blood, 113, 4834–4840. & Prete, A. (2014) Autoimmune hematological Karakantza, M., Moukaki, A., Theodoropoulou,
Carson, K.R., Beckwith, L.G. & Mehta, J. (2010) diseases after allogeneic hematopoietic stem cell M., Bussel, J.B. & Maniatis, A. (2000) Th1 and
Successful treatment of IgM-mediated autoim- transplantation in children: an italian multicen- Th2 cytokines in a patient with Evans’ syndrome
mune hemolytic anemia with bortezomib. Blood, ter experience. Biology of Blood and Marrow and profound lymphopenia. British Journal of
115, 915. Transplantation, 20, 272–278. Haematology, 110, 968–970.
Casaccia, M., Torelli, P., Squarcia, S., Sormani, Farruggia, P. & Dufour, C. (2015) Diagnosis and Karim, I.A. & Giles, F.J. (2008) Mammalian target
M.P., Savelli, A., Troilo, B., Santori, G. & management of primary autoimmune neutrope- of rapamycin as a target in hematological malig-
Valente, U. (2006) Laparoscopic splenectomy nia in children: insights for clinicians. Therapeu- nancies. Current Problems in Cancer, 32, 161–
for hematologic diseases: a preliminary analysis tic Advances in Hematology, 6, 15–24. 177.
performed on the Italian Registry of Laparo- Fioredda, F., Calvillo, M., Bonanomi, S., Coliva, Khandelwal, P., Davies, S.M., Grimley, M.S., Jor-
scopic Surgery of the Spleen (IRLSS). Surgical T., Tucci, F., Farruggia, P., Pillon, M., Martire, dan, M.B., Curtis, B.R., Jodele, S., Marsh, R. &
Endoscopy, 20, 1214–1220. B., Ghilardi, R., Ramenghi, U., Renga, D., Filipovich, A.J. (2014) Bortezomib for refrac-

Cervinek, L., Mayer, J. & Doubek, M. (2015) Sus- Menna, G., Pusiol, A., Barone, A., Gambineri, tory autoimmunity in pediatrics. Biology in
tained remission of chronic immune thrombo- E., Palazzi, G., Casazza, G., Lanciotti, M. & Blood and Marrow Transplantation, 20, 1654–
cytopenia after discontinuation of treatment Dufour, C. (2012) Neutropenia Committee of 1659.
with thrombopoietin-receptor agonists in adults. the Marrow Failure Syndrome Group of the K€uhne, T. (2015) Treatment of pediatric primary
International Journal of Hematology, 102, 7–11. AIEOP (Associazione Italiana Emato-Oncologia immune thrombocytopenia with thrombopoietin
Cheung, W.W., Hwang, G.Y., Tse, E. & Kwong, Pediatrica) Congenital and acquired neutrope- receptor agonists. Seminars in Hematology, 52,
Y.L. (2006) Alemtuzumab induced complete nias consensus guidelines on therapy and fol- 25–30.
remission of autoimmune hemolytic anemia low-up in childhood from the Neutropenia Liesveld, J.L., Rowe, J.M. & Lichtman, M.A. (1987)
refractory to corticosteroids, splenectomy and Committee of the Marrow Failure Syndrome Variability of the erythropoietic response in
rituximab. Haematologica, 91(5 Suppl.), ECR13. Group of the AIEOP (Associazione Italiana autoimmune haemolytic anemia: analysis of 109
Cines, D.B. & Blanchette, V.S. (2002) Immune Emato-Oncologia Pediatrica). American Journal cases. Blood, 69, 820–826.
thrombocytopenic purpura. N Engl J Med., 346 of Hematology, 87, 238–243. Loar, R.W., Driscoll, D.J., Kushwaha, S.S., Cramer,
(13), 995-1008Coon WW. Splenectomy in the Flores, G., Cunningham-Rundles, C., Newland, C.H., O’Leary, P.W., Daly, R.C., Mauriello, D.A.
treatment of hemolytic anemia (1985). Archives A.C. & Bussel, J.B. (1993) Efficacy of intra- & Johnson, J.N. (2013) Empiric switch from cal-
of Surgery, 120, 625–628. venous immunoglobulin in the treatment of cineurin inhibitor to sirolimus-based immuno-
Clipstone, N.A. & Crabtree, G.R. (1992) Identifica- autoimmune hemolytic anemia: results in 73 suppression in pediatric heart transplantation
tion of calcineurin as a key signalling enzyme in patients. American Journal of Hematology, 44, recipients. Pediatric Transplantation, 17, 794–
T-lymphocyte activation. Nature, 357, 695–697. 237–242. 799.

532 ª 2015 John Wiley & Sons Ltd


British Journal of Haematology, 2016, 172, 524–534
Review

Lucchini, G., Masera, N., Foti, G., Assali, G., Perse- disease responsive to steroid pulse therapy. Provan, D. & Newland, A. (2002) Fifty years of
ghin, P. & Biagi, E. (2009) A life threatening paedi- Annals of Hematology, 71, 101–102. idiopathic thrombocytopenic purpura (ITP):
atric case of acute autoimmune haemolytic anemia Oda, H., Honda, A., Sugita, K., Nakamura, A. & management of refractory ITP in adults. British
(AIHA) successfully cured by plasma exchange Nakajima, H. (1985) High dose intravenous IgG Journal of Haematology, 118, 933–944.
and combined immunosoppressive treatment. infusion in refractory autoimmune hemolytic Provan, D., Moss, A.J., Newland, A.C. & Bussel,
Transfusion and Apheresis Science, 40, 115–118. anemia (Evans syndrome). Journal of Pediatrics, J.B. (2006) Efficacy of mycophenolate mofetil as
Mathew, P., Chen, G. & Wang, W. (1997) Evans 107, 744–746. single-agent therapy for refractory immune
syndrome: results of a national survey. Journal Oliveira, J.B., Bleesing, J.J., Dianzani, U., Fleisher, thrombocytopenic purpura. American Journal of
of Pediatric Hematology/Oncology, 19, 433–437. T.A., Jaffe, E.S., Lenardo, M.J., Rieux-Laucat, F., Hematology, 81, 19–25.
McCarthy, I.J., Danielson, C.F.M., Fernandez, C., Siegel, R.M., Su, H.C., Teachey, D.T. & Rao, Pui, C.H., Wilimas, J. & Wang, W. (1980) Evans
Skipworth, E., Limiac, C.A., Prahlow, T. & V.K. (2010) Revised diagnostic criteria and clas- syndrome in childhood. Journal of Pediatrics, 97,
Goldman, J. (1999) Intensive plasma-exchange sification for the autoimmune lymphoprolifera- 754–758.
for severe autoimmune hemolytic anemia in a tive syndrome (ALPS): report from the 2009 Quartier, P., Brethon, B., Philippet, P., Landman-
four months-old infant. Journal of Clinical NIH International Workshop. Blood, 116, e35– Parker, J., Le Deist, F. & Fischer, A. (2001)
Apheresis, 14, 190–192. e40. Treatment of childhood autoimmune haemolytic
Meyer, O., Stahl, D., Beckhove, P., Huhn, D. & Otheo, E., Maldonado, M.S., Mu~ noz, A. & anaemia with rituximab. Lancet, 358, 1511–1513.
Salama, A. (1997) Pulsed high-dose dexametha- Hernandez-Jodra, M. (1997) High-dose intra- Rao, V.K. (2015) Approaches to managing autoim-
sone in chronic autoimmune haemolytic anae- venous immunoglobulin as single therapy in a mune cytopenias in novel immunological disor-
mia of warm type. British Journal of child with autoimmune hemolytic anemia. Pedi- ders with genetic underpinnings like
Haematology, 98, 860–862. atric Hematology and Oncology, 4, 487–490. autoimmune lymphoproliferative syndrome.
Meytes, D., Adler, M., Viraq, I., Feigl, D. & Ozsoylu, F. (2004) Megadose methylprednisolone Frontiers in Pediatrics, 3, 65.
Levene, C. (1985) High-dose methylpred- for the treatment of patients with Evans syn- Rao, V.K. & Oliveira, J.B. (2011) How I treat
nisolone in acute immune cold hemolysis. New drome. Pediatric Hematology and Oncology, 21, autoimmune lymphoproliferative syndrome.
England Journal of Medicine, 312, 318. 739–740. Blood, 118, 5741–5751.
Miano, M., Poggi, V., Banov, L., Fioredda, F., Pasquet, M., Aladjidi, N., Guiton, C., Courcoux, Rao, V.K., Dugan, F., Dale, J.K., Davis, J., Tretler, J.,
Micalizzi, C., Svahn, J., Montobbio, G., Galli- M.F., Munzer, M., Auvrignon, A., Lutz, P., Hurley, J.K., Fleisher, T., Puck, J. & Straus, S.E.
cola, F., Molinari, A.C., Parasole, R., Petruz- Ducassou, S., Leroy, G., Munzer, C. & Leverger, (2005) Use of mycophenolate mofetil for chronic,
ziello, F., Fischer, A., Calvillo, M. & Dufour, C. G.; Centre de Reference National des Cytopenies refractory immune cytopenias in children with
(2014a) Sirolimus as maintenance treatment in Auto-immunes de l’Enfant (CEREVANCE). autoimmune lymphoproliferative syndrome. Bri-
an infant with life-threatening multiresistant (2014) Romiplostim in children with chronic tish Journal of Haematology, 129, 534–538.
pure red cell anemia/autoimmune hemolytic immune thrombocytopenia (ITP): the French Roy-Burman, R.A. & Glader, B.E. (2002) Resolu-
anemia. Journal of Pediatric Hematology/Oncol- experience. British Journal of Haematology, 164, tion of severe Donath Landsteiner autoimmune
ogy, 36, e145–e148. 266–271. haemolytic anemia temporally associated with
Miano, M., Calvillo, M., Palmisani, E., Fioredda, Passweg, J.R. & Rabusin, M. (2008) Hematopoetic institution of plasmapheresis. Critical Care Med-
F., Micalizzi, C., Svahn, J., Banov, L., Russo, G., stem cell transplantation for immune thrombo- icine, 30, 931–934.
Lanza, T. & Dufour, C. (2014b) Sirolimus for cytopenia and other refractory autoimmune Ru, X. & Liebman, H.A. (2003) Successful treat-
the treatment of multi-resistant autoimmune cytopenias. Autoimmunity, 41, 660–665. ment of refractory pure red cell aplasia associ-
haemolytic anaemia in children. British Journal Patel, V.L., Mahevas, M., Lee, S.Y., Stasi, R., Cun- ated with lymphoproliferative disorders with the
of Haematology, 167, 571–574. ningham-Rundles, S., Godeau, B., Kanter, J., anti-CD52 monoclonal antibody alemtuzumab
Miano, M., Scalzone, M., Perri, K., Palmisani, E., Neufeld, E., Taube, T., Ramenghi, U., Shenoy, (Campath-1H). British Journal of Haematology,
Caviglia, I., Micalizzi, C., Svhan, J., Calvillo, M., S., Ward, M.J., Mihatov, N., Patel, V.L., Bierling, 123, 278–281.
Banov, L., Terranova, P., Lanza, T., Dufour, C. P., Lesser, M., Cooper, N. & Bussel, J.B. (2012) Rubin, L.G. & Schaffner, W. (2014) Clinical prac-
& Fioredda, F. (2015) Mychofenolate Mofetile Outcomes 5 years after response to rituximab tice. Care of the asplenic patient. New England
and Sirolimus as second or further line treat- therapy in children and adults with immune Journal of Medicine, 371, 349–356.
ment in children with chronic refractory Primi- thrombocytopenia. Blood, 119, 5989–5995. Saif, M.W. (2001) HIV-associated autoimmune
tive or Secondary Autoimmune Cytopenias: a Pe~nalver, F.J., Alvarez-Larran, A., Dıez-Martin, hemolytic anemia: an update. AIDS Patient Care
single centre experience. British Journal of Hae- J.L., Gallur, L., Jarque, I., Caballero, D., Dıaz- and STDS, 15:217–224. Review.
matology, 171, 247–253. Mediavilla, J., Bustelos, R., Fernandez-Ace~ nero, Savasßan, S., Warrier, I. & Ravindranath, Y. (1997)
Michel, M., Chanet, V., Dechartres, A., Morin, M.J. & Cabrera, J.R. (2010) Multi-institutional The spectrum of Evans’ syndrome. Archives of
A.S., Piette, J.C., Cirasino, L., Emilia, G., Zaja, Retrospective Study on the use of rituximab in Disease in Childhood, 77, 245–248.
F., Ruggeri, M., Andres, E., Bierling, P., Godeau, refractory AIHA. Rituximab is an effective and Scaradavou, A. & Bussel, J. (1995) Evans syn-
B. & Rodeghiero, F. (2009) The spectrum of safe therapeutic alternative in adults with refrac- drome. Results of a pilot study utilizing a multi-
Evans syndrome in adults: new insight into the tory and severe autoimmune hemolytic anemia. agent treatment protocol. Journal of Pediatric
disease based on the analysis of 68 cases. Blood, Annals of Hematology, 89, 1073–1080. Hematology. Oncology, 17, 290–295.
114, 3167–3172. Petz, L.D. & Garratty, G. (2004) Immune Hemoly- Schwartz, J., Winters, J.L., Padmanabhan, A., Balo-
Moyo, V.M., Smith, D., Brodsky, I., Crilley, P., tic Anemias. Churchill Livingstone, New York. gun, R.A., Delaney, M., Linenberger, M.L.,
Jones, R.J. & Brodsky, R.A. (2002) High-dose Pilishvili, T., Lexau, C., Farley, M.M., Hadler, J., Szczepiorkowski, Z.M., Williams, M.E., Wu, Y.
cyclophosphamide for refractory autoimmune Harrison, L.H., Bennett, N.M., Reingold, A., & Shaz, B.H. (2013) Guidelines on the use of
hemolytic anemia. Blood, 100, 704–706. Thomas, A, Schaffner, W., Craig, A.S., Smith, therapeutic apheresis in clinical practice-evi-
Naithani, R., Agrawal, N., Mahapatra, M., Kumar, P.J., Beall, B.W., Whitney, C.G. & Moore, M.R. dence-based approach from the Writing Com-
R., Pati, H.P. & Choudhry, V.P. (2007) Autoim- (2010) Sustained reductions in invasive pneu- mittee of the American Society for Apheresis:
mune hemolytic anemia in children. Pediatric mococcal disease in the era of conjugate vaccine. the sixth special issue. Journal of Clinical Aphere-
Hematology and Oncology, 24, 309–315. Active Bacterial Core Surveillance/Emerging sis, 28, 145–284.
Nanan, R., Scheurlen, W., Gerlich, M. & Huppertz, Infections Program Network. The Journal of Seidel, M.G. (2014) Autoimmune and other
H.I. (1995) Severe low-titer cold-hemagglutinin Infectious Diseases Jan 1;201:32–41. cytopenias in primary immunodeficiencies:

ª 2015 John Wiley & Sons Ltd 533


British Journal of Haematology, 2016, 172, 524–534
Review

pathomechanisms, novel differential diagnoses, Toy, P.T., Rid, M.E. & Bums, M. (1985) Positive Ware, R.E. (2009) Autoimmune hemolytic anemia.
and treatment. Blood, 124, 2337–2344. direct antiglobulin test associated with hyper- In: Nathan and Oski’s Hematology of Infancy
Seidel, M.G., Urban, C., Sipurzynski, J., Beham- globulinemia in acquired immunodeficiency and Childhood (eds by S.H. Orkin, D.G.
Schmid, C., Lackner, H. & Benesch, M. (2014) syndrome (AIDS). American Journal of Hematol- Nathan, D. Ginsburg, A.T. Look, D.E. Fisher &
High response rate but short-term effect of ogy, 19, 145–150. S.E. Lux IV), pp. 613–658. 7th edn. Saunders
romiplostim in paediatric refractory chronic Ucßar, B., Akg€ un, N., Aydogdu, S.D., Kirel, B. & Elsevier, Philadelphia.
immune thrombocytopenia. British Journal of Idem, S. (1999) Treatment of refractory Evans’ Williams, J.A. & Boxer, L.A. (2003) Combination
Haematology, 165, 419–421. syndrome with cyclosporine and prednisone. therapy for refractory idiopathic thrombocy-
Seif, A.E., Manno, C.S., Sheen, C., Grupp, S.A. & Pediatrics International, 41, 104–107. topenic purpura in adolescents. Journal of Pedi-
Teachey, D.T. (2010) Identifying autoimmune Urban, C., Lackner, H., Sovinz, P., Benesch, M., atric Hematology Oncology, 25, 232–235.
lymphoproliferative syndrome in children with Schwinger, W., Dornbusch, H.J. & Moser, A. Willis, F., Marsh, J.C., Bevan, D.H., Killick, S.B.,
Evans syndrome: a multi-institutional study. (2006) Successful unrelated cord blood trans- Lucas, G., Griffiths, R., Ouwehand, W., Hale,
Blood, 115, 2142–2145. plantation in a 7-year-old boy with Evans syn- G., Waldmann, H. & Gordon-Smith, E.C.
Svahn, J., Fioredda, F., Calvillo, M., Molinari, drome refractory to immunosuppression and (2001) The effect of treatment with
A.C., Micalizzi, C., Banov, L., Schmidt, M., double autologous stem cell transplantation. Campath-1H in patients with autoimmune
Caprino, D., Marinelli, D., Gallisai, D. & European Journal of Haematology, 76, 526–530. cytopenias. British Journal of Haematology, 114,
Dufour, C. (2009) Rituximab-based immuno- Valentini, R.P., Imam, A., Warrier, I., Ellis, D., 891–898.
suppression for autoimmune haemolytic anae- Ritchey, A.K., Ravindranath, Y., Shapiro, R. & Zecca, M., Nobili, B., Ramenghi, U., Perrotta, S.,
mia in infants. British Journal of Haematology, Moritz, M.L. (2006) Sirolimus rescue for tacroli- Amendola, G., Rosito, P., Jankovic, M., Pierani,
145, 96–100. mus-associated post-transplant autoimmune P., De Stefano, P., Bonora, M.R. & Locatelli, F.
Teachey, D.T. & Lambert, M.P. (2013) Diagnosis hemolytic anemia. Pediatric Transplantation, 10, (2003) Rituximab for the treatment of refractory
and management of autoimmune cytopenias in 358–361. autoimmune hemolytic anemia in children.
childhood. Pediatric Clinics of North America, Waespe, N., Zeilhofer, U. & G€ ung€
or, T. (2014) Blood, 101, 3857–3861.
60, 1489–1511. Treatment-refractory multi-lineage autoimmune Zhan, P., Tey, S.K., Koyama, M., Kuns, R.D.,
Teachey, D.T., Manno, C.S., Axsom, K.M., cytopenia after unrelated cord blood transplan- Olver, S.D., Lineburg, K.E., Lor, M., Teal, B.E.,
Andrews, T., Choi, J.K., Greenbaum, B.H., tation: remission after combined bortezomib Raffelt, N.C., Raju, J., Levegue, L., Markey, K.A.,
McMann, J.M., Sullivan, K.E., Travis, S.F. & and vincristine treatment. Pediatric Blood and Varelias, A., Clouston, A.D., Lane, S.W., Mac-
Grupp, S.A. (2005) Unmasking Evans syndrome: Cancer, 61, 2112–2114. DOnald, K.P. & Hill, G.R. (2013) Induced regu-
T-cell phenotype and apoptotic response reveal Wang, W. (1988) Evans syndrome in childhood: latory T cells promote tolerance when stabilized
autoimmune lymphoproliferative syndrome pathophysiology, clinical course, and treatment. by Rapamycin and IL-2 in vivo. The Journal of
(ALPS). Blood, 105, 2443–2448. Journal of Pediatric Hematology/Oncology, 10, Immunology, 191, 5291–5303.
Teachey, D.T., Seif, A.E. & Grupp, S.A. (2009) 330–338. Zhang, W.G., Ji, L., Cao, X.M., Chen, Y.X., He,
Advances in the management and understanding Wang, W., Herrod, H., Pui, C.H., Presbury, G. & A.L., Liu, J., Zhao, W.H. & Zou, S.P. (2005)
of autoimmune lymphoproliferative syndrome Wilimas, J. (1983) Immunoregulatory abnormal- Mycophenolate mofetil as a treatment for refrac-
(ALPS). British Journal of Haematology, 148, ities in Evans syndrome. American Journal of tory idiopathic thrombocytopenic purpura. Acta
205–216. Hematology, 15, 381–390. Pharmacologica Sinica, 26, 598–602.

534 ª 2015 John Wiley & Sons Ltd


British Journal of Haematology, 2016, 172, 524–534

Anda mungkin juga menyukai