Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

YBLRE-00467; No of Pages 14

Blood Reviews xxx (2017) xxx–xxx

Contents lists available at ScienceDirect

Blood Reviews

journal homepage: www.elsevier.com/locate/blre

REVIEW

The role of splenectomy in autoimmune hematological disorders:


Outdated or still worth considering?
Judith Sys a, Drew Provan b, Alexander Schauwvlieghe c, Steven Vanderschueren d,1, Daan Dierickx e,⁎,1
a
KU Leuven – University of Leuven, Department of General Internal Medicine, University Hospitals Leuven, B-3000, Leuven, Belgium
b
Department of Hematology, Barts and The London School of Medicine and Dentistry, Whitechapel, London, UK
c
Department of Hematology, University Hospital Ghent, B-9000, Ghent, Belgium
d
KU Leuven – University of Leuven, Department of Microbiology and Immunology, Laboratory for Clinical Infectious and Inflammatory Disorders, University Hospitals Leuven, Department of Gen-
eral Internal Medicine, B-3000, Leuven, Belgium
e
KU Leuven – University of Leuven, Department of Oncology, Laboratory for Experimental Hematology, University Hospitals Leuven, Department of Hematology, B-3000, Leuven, Belgium

a r t i c l e i n f o a b s t r a c t

Available online xxxx We discuss the role of splenectomy in the autoimmune hematological disorders immune thrombocytopenia
Keywords: (ITP), autoimmune hemolytic anemia (AIHA) and thrombotic thrombocytopenic purpura (TTP). Management
Immune thrombocytopenia of these disorders has dramatically changed the past decade as increasing knowledge of the
Autoimmune hemolytic anemia immunopathogenesis has led to the introduction of new therapies. Until 10 years ago, splenectomy was the
Thrombotic thrombocytopenic purpura
established second-line treatment for ITP, considered when corticosteroids failed to induce a sustained response.
Splenectomy
Second-line treatment
Concurrently, novel treatments, including anti-CD20 antibodies (rituximab) and thrombopoietin receptor ago-
nists are increasingly used. This has led to uncertainty as to when splenectomy is advisable as the next step.
The lack of comparative studies of second-line treatment options further fuels this uncertainty. Splenectomy con-
tinues to provide the highest cure rate, but it is an invasive, irreversible treatment option with a downside of post-
operative complications and a largely unpredictable outcome. Careful selection of patients, widespread adoption of a
laparoscopic approach, perioperative thromboprophylaxis, and better approaches to prevent and mitigate sepsis
have however reduced morbidity and mortality. As in ITP, splenectomy is considered the standard second-line treat-
ment in warm AIHA as well, although its position is nowadays less robust as rituximab tends to reach approximately
the same success rate in second-line treatment. The role of splenectomy in TTP has never been clarified. Although
rituximab is forwarded as best second-line therapy in recent guidelines, there are some case series suggesting
that splenectomy is a safe and effective option in refractory or relapsing disease as well.
© 2017 Published by Elsevier Ltd.

1. Introduction increasingly viewed as the last resort. This is evidenced by a decrease


in the rate of splenectomy for ITP of 50–60% in older cohorts to 15–
Immune thrombocytopenia (ITP), autoimmune hemolytic anemia 25% in more recent studies [1,2]. Nevertheless, the evolution of new
(AIHA) and thrombotic thrombocytopenic purpura (TTP) are well- therapies was accompanied by an evolution in the approach to splenec-
defined, predominantly immune-mediated conditions with significant tomy as well, with the adoption of laparoscopy instead of open surgery
morbidity and mortality. Although first-line treatment induces a re- and new measures to reduce the thrombotic and infectious risk. We
sponse in a substantial proportion of patients, refractory disease and re- highlight new insights regarding the pathogenesis of ITP, TTP and
lapse are frequently encountered. Splenectomy is a possible second-line AIHA, we explain novel treatment options and we investigate the pres-
treatment in each of these diseases, but its role has changed dramatical- ent position of splenectomy in each of the three disorders while focus-
ly the past years, with the introduction of new promising therapies. Be- ing on the durability of response. Finally, we discuss the current risks
side new therapeutic options, also the fear of important postoperative and preventable measures that should be undertaken.
complications and the unpredictable response to splenectomy, have re-
sulted in physicians tending to avoid or defer splenectomy, which is 2. Immune thrombocytopenia

⁎ Corresponding author. 2.1. Definition, classification and epidemiology


E-mail addresses: judith.sys@student.kuleuven.be (J. Sys), a.b.provan@qmul.ac.uk
(D. Provan), alexander.schauwvlieghe@ugent.be (A. Schauwvlieghe),
steven.vanderschueren@uzleuven.be (S. Vanderschueren), daan.dierickx@uzleuven.be
Immune thrombocytopenia is an acquired autoimmune disorder
(D. Dierickx). characterized by a peripheral blood platelet count b100 × 109/l, without
1
Shared senior authorship. abnormalities in the erythroid and myeloid/lymphoid lineages [3]. ITP

http://dx.doi.org/10.1016/j.blre.2017.01.001
0268-960X/© 2017 Published by Elsevier Ltd.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
2 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

may be found in the absence of any obvious initiating or underlying In 1951, Harrington and Hollingsworth demonstrated that the pas-
cause and is then called primary ITP. If associated with other disorders, sive transfer of plasma from ITP patients induced a transient thrombo-
including autoimmune conditions (e.g. antiphospholipid antibody syn- cytopenia in healthy recipients [7]. Further studies demonstrated the
drome), infections (e.g. hepatitis C virus, HIV, H. pylori) and certain presence of platelet-reactive antibodies (Abs), mainly of the IgG1 sub-
drugs, it is defined as secondary ITP. ITP can also be classified based on class. The most commonly identified antigenic targets of these IgG auto-
disease duration by the following definitions: newly diagnosed (from di- antibodies are platelet glycoproteins (GP) IIb/IIIa and Ib/IX, with a
agnosis until 3 months), persistent (3–12 months duration), and chronic number of ITP patients having antibodies directed to multiple platelet
(N12 months duration) [4]. The incidence of ITP in adults is estimated at antigens [8].
approximately 2.2–3.9 per 105 persons per year [5,6]. There is no straightforward explanation as to why patients develop
autoantibodies to several structurally unrelated platelet surface pro-
2.2. Immunopathogenesis of ITP teins. It has been theorized that proteosomal degradation of antibody-
coated platelets in antigen presenting cells (APCs) may generate novel
ITP is a complex immune process in which cellular and humoral im- immunogenic epitopes from normal platelet proteins, leading to epi-
munity are involved in the destruction of platelets as well as impaired tope spread. Alternative explanations, including somatic mutation of
platelet production. Several theories have emerged in the last decade autoantibodies and crossreactivity to unrecognized sharing of structural
to explain this autoimmune process. The main mechanisms are summa- motifs, have not been excluded [9].
rized in Fig. 1. Antibodies are only detectable in 40 to 50% of patients. This may be
explained because brisk clearance of some types of antibody-platelet
2.2.1. Role of B-cells complexes may reduce circulating antibody titers to below the thresh-
old of detection. Also tightly bound antiplatelet Abs may be difficult to
dissociate for study. Secondly, there may be some undetected antigens,
1. IgG autoantibodies against GP IIb/IIIa and Ib/IX
such as minor or cryptic antigens on platelets or antigens that reside pri-
2. Elevated expression of BAFF and APRIL marily on megakaryocytes. Finally, there may simply be a subset of pa-
3. Impaired regulatory B-cells tients in which there are other mechanisms of platelet loss [10].

Fig. 1. Schematic representation of the pathophysiology of ITP. Platelets opsonized by autoantibodies are destroyed by macrophages in the spleen and peptide fragments expressed with
MHC class II stimulate helper T-cells, that in turn activate autoreactive B-cells. Impaired regulatory T-cells (Tregs) fail to suppress this vicious cycle. Regulatory B-cells (Bregs), which induce
the recruitment or differentiation of regulatory T-cells, are also impaired. Autoantibodies furthermore suppress megakaryocytopoiesis. Platelet autoantibodies may fix complement,
enhancing opsonization or facilitating direct platelet lysis. They can also cause platelet destruction via induction reactive oxygen species (ROS). Autoreactive cytotoxic T-cells may play
a role in the destruction of platelets and megakaryocytes. Finally, decreased levels of TPO (thromopoietin) impair an effective megakaryocytosis.
This figure is modified with permission from a figure originally published in International Journal of Hematology. Kashiwagi H, Tomiyama Y. Pathophysiology and management of primary
immune thrombocytopenia. Int J Hematol. 2013;98(1):24–33. The original publication is available at www.springerlink.com.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 3

Several features of the platelet-reactive autoantibodies indicate that based activation motifs (ITAM) or inhibitory motifs (ITIM), respectively.
they arise as part of an antigen-driven clonal expansion. The destruction FcγRIIA and FcγRIIIA have ITAM domains and activate phagocytosis.
of B-cell equilibrium in ITP could be partially mediated by the elevated FcγRIIB contains an ITIM domain and inhibit phagocytosis and pro-
expression of B-cell activating factor (BAFF) and a proliferation inducing inflammatory cytokine release [24]. Evidence has accumulated to indi-
ligand (APRIL), which has been shown to support the survival and dif- cate that FcγRIIA and FcγRIIIA are primarily responsible for removal of
ferentiation of B-cells [11,12]. opsonized platelets [25]. Shifting of the monocyte FcγR balance toward
In addition to the regulatory role undertaken by regulatory T-cells the inhibitory FcγRIIB has been demonstrated in ITP patients who
(Tregs) in immune tolerance, there is recent evidence for a new subset responded to H. pylori eradication [26] or high-dose dexamethasone
of peripheral immune regulatory cells, known as regulatory B-cells treatment [27].
(Bregs), associated with chronic ITP. Bregs promote peripheral toler- In addition to Fc-mediated phagocytosis, platelet autoantibodies
ance by the production of IL-10, thereby inducing the recruitment may fix complement, enhancing opsonization or facilitating direct
and/or differentiation of Tregs, and reducing the function of CD4+ T- platelet lysis [10,28,29]. Moreover, Nardi et al. [30] showed that anti-
cells. It was recently suggested that non-splenectomized patients with GPIIIa antibodies found in an HIV-associated ITP patient caused platelet
chronic ITP exhibit functionally impaired Bregs, concomitant with destruction via induction of reactive oxygen species (ROS) and that
lowered IL-10 levels in all B-cells [13] these mechanisms may also contribute to primary ITP. Some patients
with ITP lack detectable anti-platelet antibodies, and platelet clearance
2.2.2. Role of T-cells in these cases may result from CD8+ T-cell mediated cytotoxicity [16,
31].

1. Auto-reactive T-cells against cryptic GPIIB/IIIa epitopes 2.2.4. Impaired platelet production
2. Increase of oligoclonal T-cells
3. Cytotoxic T-cells
4. Imbalanced Th1/Th2 ratio
5. Increase of Th17 cells and Th22 cells 1. Antibodies against megakaryocytes
6. Reduced regulatory T-cells 2. Lowered serum thrombopoietin (TPO) concentration
7. Altered gene expression of CD3+ T-lymphocytes
Because megakaryocytes express also GPIIb/IIIa and GPIb/IX on their
Recent evidence also links T-cells to the pathogenic process in ITP. surfaces, these cells may also be affected by autoantibodies, leading to
Platelet-reactive T-cells have been found in the blood of patients with impaired megakaryocytopoiesis and/or platelet production and release
ITP, with the major target antigen being GPIIb/IIIa [14]. In these patients, from the bone marrow. Two in vitro studies using megakaryocyte cul-
T-cells stimulate the synthesis of antibody after exposure to fragments ture systems from cord blood derived CD34+ cells showed that autoan-
of GPIIb/IIIa, but not after exposure to native proteins [15]. Other studies tibodies against anti-GPIb/IX and GPIIb/IIIa in ITP patients reduce
have shown an expansion of oligoclonal T-cells and the presence of cy- megakaryocyte production and maturation [32,33].
totoxic T-cells with a direct lytic effect on autologous platelets and/or Electron microscopy studies of bone marrow megakaryocytes from
megakaryocytes [16]. patients with ITP have shown extensive cellular abnormalities (e.g. mi-
The emergence of anti-platelet autoantibodies and anti-platelet cy- tochondrial swelling, distension of demarcation membranes and con-
totoxic T-cells is a consequence of the loss of immunological tolerance densation of nuclear chromatin) in a significant percentage of all
for self-antigens. As in the majority of autoimmune diseases, this is stages of the ITP megakaryocyte. This para-apoptotic phenomenon sug-
caused by an imbalance in pro-inflammatory and anti-inflammatory gests that autoantibodies may initiate the cascade of programmed cell
mechanisms [17]. For example, chronic ITP patients exhibit an imbal- death [34].
anced Th1/Th2 ratio (ratio between type 1 T helper and type 2 T helper Thrombopoietin (TPO) is the major physiologic regulator of platelet
cells) favoring autoreactive B-cell development [18]. Furthermore, IL-17 production. Serum TPO levels in patients with ITP were within the nor-
produced by pro-inflammatory CD4+ T helper 17 cells is increased in mal range or only slightly elevated in ITP patients compared with other
patients with ITP [19]. Likewise, Th22 cells - another newly identified thrombocytopenic disorders such as aplastic anemia with TPO levels up
potent proinflammatory T-cell subset - were significantly elevated in to 10 times higher [35,36]. Taken together with the moderately im-
active ITP patients [20] and glucocorticoid therapy could downregulate paired platelet production in ITP, the finding of relatively low levels of
expression of Th22 as well as plasma IL-22 [21]. TPO paved the way for treatment with TPO receptor agonists.
On the other hand, it has been shown that CD4+ CD25+ immunoreg-
ulatory T-cells (Tregs), which suppress cell-mediated and antibody- 2.3. Treatment
mediated immune responses and protect a host against autoimmunity,
have an impaired suppressive activity when compared to healthy sub- Treatment is generally confined to patients who are bleeding or are
jects [22]. Also, CD3+ lymphocytes from patients with active ITP present perceived to be at significant risk of bleeding. Although a platelet count
an altered expression of genes associated with apoptosis and are signif- of b30 × 109/l is often used as a surrogate marker, lower (10–20 × 109/l)
icantly more resistant to dexamethasone-induced suppression com- or higher (50 × 109/l) thresholds are pursued depending on the bleed-
pared to normal lymphocytes [16,23]. ing risk of the patient, which needs to be weighed against the probable
benefits and the risk of treatment [37].
2.2.3. Mechanisms of accelerated platelet clearance
2.3.1. First-line treatment

1. Destruction by macrophages in spleen via Fc receptors 2.3.1.1. Corticosteroids. Corticosteroids are the main first line therapy.
2. Complement-mediated platelet lysis The mechanism by which steroids act is believed to be through their im-
3. Induction of reactive oxygen species munosuppressant effect on T-cells. Oral prednisone is usually given at
4. Clearance by CD8+ T-cell mediated cytotoxicity
0.5 to 2 mg/kg/day in tapering doses until the platelet count increases
Platelets opsonized by IgG antibodies are destroyed mainly by mac- (which requires on average 1 to 6 weeks). An alternative is high dose
rophages in spleen via low-affinity Fc receptors. Functionally, there are dexamethasone (HDD), 40 mg daily for 4 days per month for several cy-
two different classes of Fc receptors: the activation and the inhibitory cles. Approximately two-thirds of patients achieve a complete or partial
receptors, which transmit their signals via immunoreceptor tyrosine- response with corticosteroids at these standard doses [3]. However,

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
4 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

only 5% to 30% of patients have sustained remission, depending on age series over 58 years and across 29 countries, found that 1731 (66%) of
and type of ITP (primary versus secondary), and further depending on 2623 patients had a complete response (defined as a platelet count of
the criteria used to define a response, and the duration of follow-up N100 × 109/l) and 1853 (88%) of 2116 had a complete or partial re-
[38]. sponse (defined as a platelet count of N 50 × 109/l) following splenecto-
my. Similar results were found in a smaller study of Vianelli et al., that
2.3.1.2. Intravenous immunoglobulin. Intravenous immunoglobulin followed 402 splenectomized patients until 5 years after surgery, with
(IVIg) is recommended for patients who are unresponsive to corticoste- a complete response in 66% of patients and a complete or partial re-
roids and is often used in pregnancy. It is thought to act by blocking Fc sponse in 86% of patients [43].
receptors in the reticulo-endothelial system [39]. Intravenous immuno- The rate of relapse in Vianelli et al. was estimated 23% with a median
globulin rapidly increases platelet counts in 65% to 80% of patients but time to relapse of 8 months, whereas in Kojouri et al. relapses occurred
the effect is transient, lasting no longer than 3 to 4 weeks, and the in a median 15% of patients, with a median follow-up of 33 months
drug requires frequent administration [3]. (range 3–153 months). The relapse rate appeared to increase with dura-
tion of follow-up, but the correlation did not reach statistical signifi-
2.3.2. Second-line treatment cance. Two case series with follow-up durations of N7 years (longer
Second-line treatment is defined as treatment of patients who are than most case series that were reviewed) reported however a continu-
unresponsive to, have intolerable adverse effects of, or relapse after ing occurrence of relapses with long-term follow-up [44,45]. Therefore,
first-line therapy. As there have been no comparative trials of treatment it is possible that publication of more case series with longer follow-up
options in this setting, second-line treatment is challenging. Available will demonstrate a decreasing frequency of complete remissions over
treatment modalities have quite different mechanisms of action. In time. We consider however that these results have been created by
this review we will discuss the three main therapies used in second- adding several retrospective series and smaller cohorts, which brings
line treatment: splenectomy, rituximab and TPO receptor agonists, about a risk of publication bias.
with a focus on splenectomy. The choice among these three options 2.3.2.1.2. Pre-operative factors to predict outcome. A lot of factors have
for second-line treatment of adults with ITP has been addressed in been studied to better predict the response of patients undergoing sple-
two recent guidelines, the International Consensus Report (2010) [40] nectomy. The large review of Kojouri et al. reviewed all articles pub-
and the American Society of Hematology Practice guideline (2011) lished before 2004 in which data were presented to support the
[41]. Table 1 compares the grade of recommendation of both guidelines conclusion that a variable did or did not predict a response to splenecto-
to the three main therapies used in second-line treatment. In Table 2 we my [42]. Predictors that were studied include patient age, previous re-
summarize their efficacy, advantages, side effects and contra- sponse to corticosteroid and IVIg therapies, preoperative and
indications. postoperative platelet count. In the review of Kojouri et al., none of
these factors was consistently found to be a statistically significant inde-
2.3.2.1. Splenectomy. Splenectomy was the first effective treatment in ITP pendent predictor of a good response to splenectomy. Later studies
and still has the highest curative potential now. By removing the spleen, [46–49] also reported conflicting results.
the clearance of autoantibody-coated platelets by Fcγ receptors An interesting method to demonstrate the site of platelet destruc-
expressed on macrophages is impaired. Besides, splenectomy works tion is by radionuclide labeling. Indium-labeled autologous platelet
by impairing the interactions between T-cells and B-cells that are in- scanning (ILAPS) is a nuclear imaging modality in which autologous
volved in the synthesis of antibodies. platelets are reinfused into the patient after ex vivo labeling with
111
In the Consensus guideline, splenectomy was given a recommenda- In. Subsequent scintigraphy demonstrates the site(s) of platelet se-
tion grade of C, the lowest level of recommendation and evidence. The questration and clearance. It has been proposed that patients with pre-
statement with this recommendation was to ‘wait at least 6 months dominantly splenic sequestration determined by ILAPS may be more
from diagnosis before performing splenectomy due to the chance of likely to respond to splenectomy than those exhibiting hepatic, mixed,
spontaneous remission’. The ASH guideline gave a strong recommenda- or diffuse patterns [50]. Kojouri et al. mentioned 6 studies showing a
tion (Grade 1) for splenectomy as the treatment for patients who have correlation of predominant splenic sequestration with response, 8
failed initial corticosteroid therapy supported by an intermediate level other studies not reporting a correlation and one study that was not in-
of evidence (level B). terpretable. In a pooled analysis of six 111In sequestration studies from
This discussion focuses mainly on primary ITP, as splenectomy has a 1986 to 2010, of which 5 studies were not included in the review of
lesser role in secondary ITP. Kojouri et al., Cuker and Cines [51] found that patients with a predomi-
2.3.2.1.1. Durability of complete responses. It is estimated that two- nantly splenic pattern of platelet sequestration demonstrated a higher
thirds of patients achieve durable complete responses with splenecto- response rate (91.4%) following splenectomy than those with a hepatic,
my. A large systematic review of Kojouri et al. [42] that included case diffuse, or mixed pattern (40.9%). All of these studies included patients
with different clinical characteristics, used different techniques and dif-
Table 1 ferent definitions of response. It is clear that additional prospective
Second-line treatments for ITP: Recommendations of the International Consensus Report studies utilizing well-defined patient populations, definitions of re-
(ICR) and the American Society of Hematology Practice Guideline [40,41].
sponse and standardized methods for ILAPS are needed to confirm the
Treatments Strength of recommendation value of this procedure. The ASH guideline does not recommend the
Consensus reporta ASH guidelinesb
routine use of ILAPS for predicting response to splenectomy in centers
without extensive experience with this modality (Grade 2C).
Splenectomy C 1B
2.3.2.1.3. Timing of splenectomy. Chronic ITP in adults has a variable
Rituximab B 2C
TPO receptor agonists A 2C and unpredictable course, with very wide inter-patient heterogeneity.
a In many institutions splenectomy is recommended if the patient still
For the ICR, the level of evidence and recommendations were linked together: A:
strong recommendation based on evidence from RCT's; B: intermediate recommendation has severe steroid-dependent thrombocytopenia after a trial with ste-
based on evidence from well-designed studies; C: weak recommendation based on expert roid or high-dose immunoglobulins after 6 weeks to 6 months from di-
opinion and clinical experience. agnosis [52], because remission is regarded as unlikely thereafter.
2b
For the ASH Practice Guideline, the levels of evidence and recommendations were However, there is increasing evidence that remissions can occur up to
assessed according to the GRADE system and were not linked: 1: strong recommendation;
2: weak recommendation; A: RCT or exceptionally strong observational studies; B: RCT
1 or 2 years after onset, suggesting that surgery may be deferred for
with important limitations or strong observational studies; C: RCT with serious flaws or prolonged periods. For instance, Sailer et al. [53] found that in 114 pa-
weaker observational study. tients with platelet counts b 20 × 109/l at diagnosis who were treated

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 5

Table 2
Comparison of the three main second-line treatment options for ITP in terms of efficacy, advantages, adverse effects and contra-indications.

Therapy Efficacy Advantages Adverse effects Contra-indications

Splenectomy - Short term response 80% - Over 60 years of experience - 0.2% mortality and 10% post-- - Patients with comorbid
- Long term response 60–70% at - Most effective treatment for ITP operative complications conditions that increase
5–10 years (bleeding, infections, thrombo- the risk of complications
- Response hard to predict sis) - Relative: elderly patients
- Increased risk of infection with over 60–70 years
rare occurrence of OPSI - Patients with immunode-
- Increased risk for arterial or ve- ficiency and secondary ITP
nous thrombosis
- Increased risk for pulmonary
hypertension
Rituximab - Short term response 50–60% - Non-surgical treatment - Infusion-related side effects - Active hepatitis B virus
- Long term response 20% at - Extensive experience since first reported use (chills, fever, dyspnea) - Known clinical allergy in-
3–5 years for ITP in 1999 - Neutropenia cluding past serum sick-
- Increased risk of infection and ness
viral reactivation - Pregnancy and lactation
- Hypogammaglobulinemia
- Serum sickness
- Increased risk of PML
TPO RAs - Platelet activation in 79–95% - Non-surgical treatment - Bone marrow reticulin fibrosis - Pregnancy and lactation
- Sustained response in 70–90% - Daily oral agent (eltrombopag) or weekly (1.4%) - Relative: past history of
in long-treatment studies subcutaneous injection (romiplostim) with - Arterial and venous thrombosis venous or arterial
- Maintenance treatment, but the potential for self-administration - Progression to AML if MDS thrombosis
recent evidence that it may - Rebound thrombocytopenia
modify clinical course of ITP
Romiplostim - Upper respiratory infection
- Nasofaryngitis
- Fatigue
- Headache
Eltrombopag - Elevated liver enzymes - Child-Pugh ≥ 5
- Nausea, vomiting
- Cataract

Abbreviations: OPSI (overwhelming post-splenectomy infection), PML (progressive multifocal leukoencephalopathy), AML (acute myeloid leukemia), MDS (myelodysplasia).

2.3.2.3. TPO receptor agonists. Because recent evidence suggests that sub-
with first-line therapies (corticosteroids and/or IVIg), complete or par- optimal platelet production is an underlying cause of ITP, increasing
tial remission was most likely to occur within the first 6 months (28 pa- platelet production has become a potential treatment option. The TPO
tients and 15 patients respectively), but another 17 complete and 8 receptor agonists now used are the TPO peptide mimetic romiplostim
partial responses occurred between 6 months and 3 years. Thus, ap- (Nplate®) and the nonpeptide mimetic eltrombopag (Revolade® or
proximately 60% of patients eventually achieved remission, supporting Promacta®).
the argument for delaying definitive decisions as splenectomy. Recent phase II and III trials confirmed the efficacy and safety of
these agents in splenectomized and non-splenectomized patients. In
patients with previously treated chronic ITP and platelets b 30 × 109/l,
2.3.2.2. Rituximab. Rituximab is a chimeric anti-CD20+ monoclonal anti- a response (defined as a platelet count N 50 × 109/l at least once during
body that targets B-cells responsible for antiplatelet antibody produc- the study) was observed in 79 to 95% of patients during a follow-up pe-
tion by Fc-mediated cell lysis. It also has an influence on the different riod of 6 months to 5 years [59–63]. In the 5-year study of Kuter et al.
T-cell subsets (including elevation of Tregs and restoration of the Th1/ [63], platelet response which happened in 95% of patients, was main-
Th2 balance) and it may lead to macrophage Fc-receptor blockade tained over 92% of their time on study.
[54]. In the largest systematic review of published reports of rituximab The expectation is that these responses are maintained while the
use in ITP, Arnold et al. [55] reported an overall response (platelet drugs are taken, but will drop to baseline levels when the drug is
count N50 × 109/l) in 62.5% of patients and a complete response (plate- stopped. However, published reports show that 25–30% of patients
let count N 150 × 109/l) in 44% after four once-weekly intravenous infu- may expect a sustained response tot the TRAs even after the drugs
sions of 375 mg/m2. The median duration of response was 10.5 months have been stopped. The mechanism for this sustained response is not
(range 3–20). The complete response rate falls to approximately 20% by clear. It is suggested that TRAs may have an effect on regulatory T-
2–5 years [56,57]. cells [64].
A recent multicenter, randomized, double masked, placebo- The Consensus guideline gave a recommendation Grade of A for
controlled trial enrolled 112 corticosteroid unresponsive adult patients treatment with TPO RAs for patients, either before or after splenectomy,
with primary ITP. They were randomly assigned (1:1) to four weekly in- based on randomized clinical trials [61,62,65] evaluating romiplostim
fusions of 375 mg/m2 rituximab or placebo. Treatment failure was seen and eltrombopag compared to placebo in patients with or without pre-
in 32 (58%) of the 55 patients in the rituximab group and 37 (69%) of 54 vious splenectomy. The ASH guideline distinguished between patients
patients in the placebo group. There was no reduction in long-term who have not had a splenectomy and patients who relapse after a sple-
treatment failure with rituximab, but results suggested apparently lon- nectomy or in whom a splenectomy is contraindicated. The ASH guide-
ger duration of response [58]. line gave a weak recommendation (Grade 2) supported by a weak level
The ASH guideline gave a weak recommendation for rituximab of evidence (level C) for patients who have not had a splenectomy. The
(Grade 2) supported by a weak level of evidence (level C), based on basis for the weak recommendation was infrequent sustained remis-
the relatively poor durable response rate and the relatively high fre- sions when treatment with TPO receptor agonists is discontinued and
quency of adverse effects. The Consensus guideline gave a recommen- the lack of long-term follow-up for adverse events. For patients who re-
dation grade of B for treatment with rituximab. lapse after a splenectomy or in whom a splenectomy is contraindicated,

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
6 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

TPO receptor agonists were given a strong recommendation (Grade which are listed and briefly explained underneath. They are derived
1) supported by an intermediate level of evidence (level B). from both experimental animal models as well as from human studies.

2.3.3. Suggested treatment algorithm for ITP 1. Autoantibodies


Fig. 2 presents a flowchart in which overall factors that direct physi- 2. Complement mediated clearance
cians toward a certain treatment option are presented. 3. Loss of self-tolerance
4. Functional abnormalities of B- and T-cells
5. Polyclonal lymphocyte activation
3. Autoimmune hemolytic anemia 6. Th1/Th2 imbalance
7. Immunoregulatory T-cells
3.1. Definition and classification
AIHA is mediated by immunoglobulins (most frequently IgG) direct-
Autoimmune hemolytic anemia (AIHA) is a group of diseases de- ed against self-RBC antigens, with Rhesus (Rh) polypeptides as the
fined by autoantibody mediated red blood cell (RBC) destruction with commonest target [69,70]. The mechanisms that have been proposed
inadequate compensation. It has an estimated incidence of 1–3 per to explain the cause of autoantigens are cross-reactivity or molecular
105 persons per year [66,67] mimicry between environmental antigens and autoantigens [71–73].
Classification divides AIHA into warm (wAIHA) or cold-reactive Control of complement activation may be critical in erythrocytes to
(cAIHA) subtypes based on the optimal RBC-autoantibody reactivity resist spontaneous complement attack. This is done by membrane-
temperatures. Warm AIHA can be further classified into primary (idio- anchored proteins including complement receptor (CR1 or CD35),
pathic) and secondary in nature. The classification of all types of AIHA decay-accelerating factor (DAF or CD55) and also CD59. Recent studies
and their prevalence is summarized in Table 3. We will confine our dis- in mice indicate that mouse RBCs deficient in CR1 or DAF are cleared
cussion to idiopathic wAIHA, being the only subtype in which splenec- spontaneously by the alternative pathway of complement [74]. Like-
tomy is a possible treatment. wise, a deficiency of CD59 expression has been reported in patients
with autoimmune cytopenias, including warm AIHA [75,76].
3.2. Immunopathogenesis of warm AIHA Another proposed mechanism is the loss of tolerance of
autoantigens. Normally, antigen-presenting cells (APCs) capture self-
The pathogenesis of AIHA is a complex process in which many fac- antigens from other cells and present them to autoreactive T-cells to in-
tors play an essential role. Barros et al. [68] report 7 mechanisms, duce T-cell tolerance by either deletion or anergy [77]. There are several

Fig. 2. Evidence based treatment algorithm for ITP. *The size of the arrows reflect the current strength of evidence. Based on recent FDA and EMEA decisions, TPO RAs are considered second
line treatment in both adult and pediatric populations.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 7

Table 3 in the generation and maintenance of peripheral tolerance. It has been


Classification of autoimmune hemolytic anemia. shown that older CD25 knockout mice develop autoimmune disease, in-
Subtype of AIHA (% of all AIHA cases) cluding AIHA [84]. Furthermore, the peripheral blood of patients with
I Warm reactive AIHA: optimal reactivity of autoantibodies at 37 °C (75%)
AIHA has been found to contain Tregs capable of inhibiting the Th1 ef-
fector responses in vitro by secretion of IL-10 [82].
A. Primary or idiopathic (37.5%)
B. Secondary (37.5%)
3.3. Treatment

Treatment of AIHA is still not evidence-based, but essentially


i. Associated with various lymphoproliferative disorders (e.g. non-Hodgkin
lymphoma, chronic lymphatic leukemia) experience-based. There are no randomized studies and only a few pro-
ii. Associated with rheumatic disorders (e.g. systemic lupus erythematosus) spective phase 2 trials. Otherwise, only retrospective studies, small se-
iii. Associated with non-lymphoid malignancies (e.g. ovarian cancer) ries of patients or single cases have been reported (evidence level V).
iv. Associated with chronic inflammatory disorders (e.g. ulcerative colitis) There is no formal consensus on the definition of complete (CR) or par-
v. Drug induced autoimmune hemolytic anemia
II Cold reactive AIHA: optimal reactivity of autoantibodies at b37 °C (17–25%)
tial (PR) hematologic remission and refractoriness. Therefore, all state-
ments on treatment recommendations in the literature have to be
regarded with caution [85].
A. Cold agglutinin mediated AIHA (CAS) (15%)

3.3.1. First line treatment


i. Primary cold agglutinin disease (CAD)
Glucocorticoids are the mainstay of treatment of newly diagnosed
ii. Secondary cold agglutinin syndrome (CAS)
wAIHA. For most cases prednisone at a dose of 1 mg/kg/day is the first
line treatment of choice. After three weeks of treatment with predni-
a. Post-infectious (e.g. mycoplasma or infectious mononucleosis)
sone, about 80–90% of all patients have a clear clinical response (i.e. he-
b. Associated with aggressive lymphoma
moglobin N 10.0 g/dl). Patients who have no clinical response at that
time are unlikely to improve from continuation of steroids, and should
B. Paroxysmal cold hemoglobinuria (Donath-Landsteiner syndrome)
thus be switched to alternative treatments [86].
(2–10%)
III Mixed type AIHA: presence of both warm and cold type antibodies (b5%) Nevertheless, only about 20% of patients will remain in remission
after withdrawal of steroids and are possibly cured. Subsequently,
A. Primary of idiopathic
most responders require maintenance steroids to maintain an accept-
B. Secondary: often associated with rheumatic disorders able hemoglobin value (N 9–10 g/dl). Approximately 40% to 50% of pa-
IV Drug induced AIHA: associated with an estimated 150 drugs (1/106) tients need 15 mg/day or less prednisone (which is generally regarded
as the highest tolerable dose for long-term treatment), but 15 to 20%
A. Drug dependent AIHA need higher maintenance prednisone doses [86–88].

i. Hapten or drug absorption 3.3.2. Second-line treatment


ii. Immune (ternary) complex Patients who are refractory to initial steroids or who need N15 mg
prednisone per day or who relapse after initial response are absolute
B. Drug independent AIHA candidates for a second-line therapy. Splenectomy and rituximab are
the only second-line treatments with a proven short-term efficacy [85].

hypotheses to explain the lack of effective presentation of autoantigens. 3.3.2.1. Splenectomy. Because the spleen is a major organ for antibody
One hypothesis is that the APCs remain relatively immature and there- synthesis and destruction of immunoprotein-coated erythrocytes, sple-
fore tolerate rather than activate self-reactive cells [77]. A second expla- nectomy has classically been proposed as the logical second-line ap-
nation is that many self-epitopes may be inefficiently processed and proach. In their guidelines, Lechner et al. [85] recommend
presented by APCs. This would result in minimal cross-presentation splenectomy to all patients without contraindications as the best
and as a result, the self-reactive naïve T-cells would remain a part of second-line therapy because of a high short-term efficacy and a good
the peripheral T-cell repertoire by escaping thymic deletion [78]. evidence of a long-term response.
In several murine models of AIHA, functional abnormalities of B- and 3.3.2.1.1. Durability of response. After splenectomy short-term com-
T-cells have been investigated, particularly their lymphocyte activation, plete response or partial response can be expected in two-thirds of pa-
cytokine production and their role in the onset of their autoimmunity. tients [89] with a range of 38% to 82% [86,88,90–94]. Although no high-
Polyclonal activation of B-cells by activation of superantigens or mi- quality data on long-term success are available, there is good evidence
togens, that are associated with a failure to control lymphoproliferation, that a substantial number of patients will remain in remission without
has been suggested as a possible mechanism by which viruses trigger need of medical intervention for years. The best data on follow-up
autoimmune disease [79,80]. were provided by Coon [94]. In 52 patients who had undergone splenec-
As in ITP, there is also an imbalance in Th1/Th2 cells. T-cells from tomy they found that 63% of patients had a hematocrit level N30% with-
murine models of AIHA produce high levels of the Th1 cytokine out steroids after a mean follow-up of 33 months, and 21% had a
interferon-γ (IFN-γ) but little or no of the Th2 cytokines IL-4, IL-5 or hematocrit level N30% with a prednisone requirement of 15 mg/d or
IL-10 [81]. Interleukin-12, a cytokine produced primarily by APCs, pro- less after a mean follow-up of 73 months. In a study of Allgood and
motes the development of Th1 cells and suppresses Th2-cell expansion, Chaplin [88], 44% of patients were in complete remission after N1 year
promoting B-cell maturation and pathologic antibody generation. In after splenectomy. It is also the experience of many hematologists that
support of the hypothesis that Th2 cytokines protect against autoim- patients with recurrence after splenectomy require lower doses of ste-
mune disease and Th1 cytokines promote autoimmune disease, it has roids to maintain acceptable hemoglobin levels [92].
been shown that high IL-10 responses in AIHA patients were associated 3.3.2.1.2. Prediction of response. As in ITP, it would be desirable to
with less severe anemia [82] and that IL-4 treatment ameliorates the identify patient characteristics that could predict the response to sple-
anemia in AIHA mice models [83]. nectomy. Studies about this are not to the same extent available in liter-
As in ITP, AIHA is also associated with abnormalities in the number ature as they were for ITP and are mostly of older age. Chertkow and
or function of regulatory T-cells (Tregs), which play an important role Dacie [91] suggested that people under the age of forty, those with a

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
8 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

small spleen, low platelet counts and severe anemia, tended to have by a severe deficiency of the protease ADAMTS13. This deficiency is ei-
poorer results from splenectomy than people over the age of forty or ther the result of an autoantibody which binds to ADAMTS13 and im-
those with large spleen, normal platelet counts and mild to moderate pairs its function (so called ‘acquired TTP’, approximately 95% of
degrees of anemia. Allgood et al. [88] found no correlation between cases), or a congenital deficiency in ADAMTS13 due to mutations pres-
the effectiveness of splenectomy and the patient's age, race, sex, initial ent in the ADAMTS13 gene (also called Upshaw-Shulman syndrome). In
hemoglobin level, reticulocyte count, white blood cell count or platelet the majority of patients with acquired TTP, there is no associated clini-
count. Yet it appeared that patients with a prior good response to corti- cally overt immune dysregulation and the condition is termed ‘primary’
costeroid therapy had a good response to splenectomy. The number of or ‘idiopathic’. In the smaller proportion of patients in which an overt
patients in each group was nonetheless too small to make this correla- cause of immune dysregulation is identifiable (e.g. systemic lupus ery-
tion statistically significant. thematosus, HIV infection), the term ‘secondary’ acquired TTP is used
Most of research, however, went to the role of radioisotopic 51Cr- [107].
RBC sequestration studies. Beside the use of spleen to liver ratios in sev-
eral studies, Jandl et al. [95] suggested to rely on an index of sequestra- 4.2. Pathophysiology
tion calculated by a complex formula. McCurdy and Roth [96]
introduced still another formula, which they termed the spleen localiza- The pathophysiology of TTP can be largely explained by a deficiency
tion index (SLI). In all these studies, irrespective of the index used, the of the von Willebrand Factor (VWF)-cleaving protease ADAMTS13,
finding of significant splenic sequestration seemed to predict a favor- which is a metalloproteinase produced by the liver in response to in-
able response to splenectomy, but the lack of evidence for significant creased vascular shear stress. Under normal conditions, ultra-large
splenic sequestration did not preclude a good response to splenectomy. (UL), hyperreactive VWF multimers are secreted from endothelial
As to give an example, Allgood et al. [88] showed in their study that 11 cells. Due to shear stress in flowing blood, they unfold and are processed
of 13 patients with a spleen to liver ratio of N2.3 benefited of splenecto- by ADAMTS13 into normally sized, quiescent multimers. This process
my, but also 4 of 7 patients with a spleen to liver ratio of b2.3 responded prevents spontaneous interaction between UL-VWF and the glycopro-
to splenectomy. tein (GP) Ib/IX/V receptor on circulating platelets. In the absence of
Since no factor consistently or significantly predicted response on ADAMTS13 activity, UL-VWF multimers can accumulate, leading to
splenectomy, the decision remains a difficult one and is better made formation of disseminated thrombi that are rich in platelets and VWF
on the basis of individual considerations, taking into account operative (Fig. 3). These microthrombi can block arterioles and capillaries
risk, probability of thrombosis, availability of rituximab, and patient resulting in organ failure and death when left untreated. Thrombocyto-
preferences. penia observed in TTP is explained by consumption of platelets in this
thrombotic process, and microangiopathic hemolytic anemia is as-
3.3.2.2. Rituximab. Rituximab is used for relapsed or refractory AIHA as a sumed to be caused by mechanical rupture of red blood cells in the
second-line alternative instead of splenectomy, or after its failure. Sev- obstructed microvasculature [108,109].
eral retrospective studies and case series regarding its use in AIHA
have shown variable response rates ranging from 40 to 100% with a me- 4.3. Treatment
dian of 60% and disease-free survival of 50% in 2 years [97–103]. Wheth-
er some patients can be considered cured after rituximab 4.3.1. First line treatment
administration is not clear, given the lack of long-term follow-up in
most published series [104]. Although it is unknown if refractoriness 4.3.1.1. Plasma exchange therapy (PEX). PEX has reduced mortality from
to its administration will develop, retreatment with rituximab has N90% without treatment to about 10–20% [110–112]. However, about
been reported to be effective for relapsing cases, and extended rituxi- 35–50% of patients surviving an acute bout of TTP will relapse after ini-
mab therapy has also been used [98]. As to reduce the side effects of ri- tial response to PEX [113,114], and some are plasma-refractory or re-
tuximab (listed in Table 2), recently, a prospective phase-II study was main PEX-dependent for long periods [115–117]. About 10% of
performed using low-dose rituximab in AIHA. In this study, overall re- patients with TTP have multiple relapses and develop a chronic relaps-
sponse rate and relapse-free survival at 12 months were both 100% in ing form of TTP [117].
the warm type, with an estimated relapse-free survival of 81% at
2 years. These results suggests that lower doses of rituximab may be suf- 4.3.1.2. Corticosteroids. In conjunction to PEX, steroids should be used in
ficient to produce long-term responses in most patients, with a lower the initial treatment of TTP to achieve relatively rapid immunosuppres-
incidence of side effects [105]. Guidelines recommend rituximab as sion [107]. There is no firm evidence to guide the choice and dose of ste-
the best option for patients who are not eligible for or who refuse sple- roid; however, there is some prospective evidence that higher doses of
nectomy [85]. methylprednisolone (10 mg/kg/day) are associated with an improved
Birgens et al. [106] published the first randomized trial of 64 patients patient outcome [118].
who received prednisolone and rituximab combined (N = 32) or pred-
nisolone monotherapy (N = 32) as first-line treatment for warm AIHA. 4.3.1.3. Caplacizumab. Caplacizumab, an anti-von Willebrand factor hu-
After 36 months, about 70% of the patients were still in remission in the manized immunoglobulin, inhibits the interaction between ultralarge
rituximab-prednisolone group, whereas only about 45% were still in von Willebrand factor multimers and platelets. In a recent phase 2 con-
complete or partial remission in the prednisolone group. These data trolled study, caplacizumab or placebo were given during plasma ex-
suggest that rituximab combined with prednisolone for the first-line change and for 30 days afterwards. Caplacizumab induced a faster
treatment of warm AIHA can increase both the rate and the duration resolution of the acute TTP episode than did placebo [119]. If this is con-
of response. firmed in further studies, Caplacizumab has the potential to become a
new treatment of TTP in addition to PEX and immunosuppressive
4. Thrombotic thrombocytopenic purpura therapy.

4.1. Definition and classification 4.3.2. Second-line treatment

Thrombotic thrombocytopenic purpura (TTP) is classically described 4.3.2.1. Rituximab. In patients refractory to daily PEX and in relapsed
as a pentad of severe thrombocytopenia, hemolytic anemia with acute idiopathic TTP where rituximab was administred together with
schistocytosis, fever and neurologic and renal abnormalities. It is caused reinitiation of PEX, remission rates of N85% were reached in some

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 9

Fig. 3. ADAMTS13 and its role in the pathophysiology of TTP. (A) UL-VWF (blue) is secreted from endothelial cells, after which it unfolds shear forces in the flowing blood thereby exposing
the ADAMTS13 cleavage site. ADAMTS13 (scissors) proteolysis trims the UL-VWF multimers into smaller multimers that do not spontaneously react with platelets (red). (B) When
ADAMTS13 is absent or inactive, UL-VWF is not cleaved and accumulates. Spontaneous interaction of UL-VWF with platelets results in the formation of microthrombi that block
capillaries and arterioles.
Figure adopted from Vanhoorelbeke K, De Meyer SF. Animal models form thrombotic thrombocytopenic purpura. J Thromb Haemost. 2013;11 Suppl 1:2–10. Used with permission from
John Wiley and Sons.

studies [120,121]. Typically, 375 mg/m2 has been used weekly for
4 weeks. Although in most reported cases no relapses were observed, splenectomy for relapsing TTP with a mean follow-up of 53 months.
length of follow-up was limited. Chemnitz et al. [122] published long- The authors conclude that the relapse rate following splenectomy was
term follow-up data for 12 patients, all achieving an initial remission lower for patients with refractory disease than for patients with relaps-
after application of rituximab. However, after a mean follow-up of ing TTP (8% vs 17%). The reviewed studies have several limitations. First,
49.6 months, 3 patients relapsed. The investigators conclude that ex- available data are from retrospectively performed small case series lack-
tended follow-up remains necessary. ing control groups. Secondly, follow-up duration in most studies was
Furthermore, a prospective multicenter trial has shown benefit in b3 years, which likely results in an underestimation of the true relapse
using rituximab as a first-line therapy [123]. In this study examining rate. Thirdly, other therapies used in these studies also varied, with
the use of rituximab in newly diagnosed or relapsed patients with TTP, some patients undergoing splenectomy alone and others taking immu-
rituximab has been shown to decrease the number of PEX required to nosuppressant medication before splenectomy [130,131].
achieve remission, to decrease the hospital duration and to reduce the The evidence by these retrospective trials is not as convincing as
risk of relapse by over 80% when compared to historical controls. some prospective studies performed with rituximab. Blombery and
British guidelines recommend rituximab in acute idiopathic TTP Scully state that rituximab should be considered the mainstay of treat-
with neurological/cardiac pathology, which are associated with a high ment for relapsing disease, as there is little good evidence regarding
mortality, in conjunction with PEX and steroids (Grade 1B) and in pa- the efficacy and safety of splenectomy [107]. British guidelines state
tients with refractory or relapsing immune-mediated TTP (Grade 1B) that splenectomy may rarely be considered in the non-acute period of
[124]. immune-mediated TTP but has limited proven benefit (Grade 2C) [124].
4.3.2.2.2. Prediction of response. In the search for parameters that can
4.3.2.2. Splenectomy. In TTP, splenectomy has been controversial since be used to tailor patient treatment, it is important to know whether an-
employed. The first splenectomy for the treatment of TTP was in 1927. tibodies against ADAMTS13 are useful as a surrogate predictor for dis-
In 1957, steroids and splenectomy were first combined in the treatment ease outcome. Some small studies indicate that response to plasma
of TTP with response rates of about 50% [125–127]. Introduction of plas- exchange may occur independent of the level of ADAMTS13 or inhibito-
ma therapy in the 1970s significantly improved prognosis with remis- ry activity [110,132]. This still remains to be established in larger
sion rates of over 70–80% [112,114]. Since then, splenectomy was studies.
predominantly performed in patients primarily refractory or with pro-
gressive disease despite plasma exchange. In these unfavorable situa- 5. Point of focus: splenectomy: risks and preventive measures
tions, splenectomy was associated with a high fatality rate of up to
40% [116,125,128]. 5.1. Perioperative complications and mortality
The mechanism by which splenectomy induces remissions in TTP is
unclear. It is speculated that removal of the spleen may alter either In the past, removal of the spleen had been performed by open sur-
ADAMTS13-antibody complex clearance and/or decrease antibody pro- gery with the surgical approach made through a midline abdominal in-
duction. In plasmapheresis-refractory TTP, splenectomy produces re- cision. The surgical wound has been responsible for much of the
missions within days. This suggests that in this situation, splenectomy morbidity leading to wound infection or herniation, difficulty in mobili-
does not work by decreasing antibody production, but rather by de- zation after surgery, increased risk of thromboembolic events and ob-
creasing clearance of the antibody-antigen complexes [129]. struction due to adhesions. Modern surgical approaches to
4.3.2.2.1. Durability of response. Dubois et al. [130] reviewed a total of splenectomy for ITP have sought to minimize these complications and
74 reported cases who underwent splenectomy for refractory disease maximize response to treatment by utilizing minimally invasive (lapa-
with a mean follow-up of 39 months and 87 cases who underwent roscopic) techniques [133].

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
10 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

In the review of Kojouri et al. [42], the complication rate in the im- The Advisory Committee on Immunization Practices (ACIP) proposes a
mediate postoperative period was 9.6% for laparoscopic splenectomy dose of the conjugated vaccine first, followed by a dose of the polysac-
and 12.9% for open splenectomy, while associated mortality rates charide vaccine at least 8 weeks later [145]. Vaccination against H.
were very low, 48 deaths on 4955 laparatomies were reported (1%), influenzae type b with a conjugated vaccine is also recommended.
and 3 deaths on 1301 laparoscopies (0.2%). Severe hemorrhage was re- The vaccine mainly protects against the risk of meningitis caused by
sponsible for 22.9% of deaths; other causes included infection, thrombo- H. influenzae type b. Furthermore, the conjugated tetravalent
embolic and cardiovascular events. These relatively high rates of ACDYW135 vaccine against N. meningitidis is recommended before
complications and death, despite advances in anesthesia and surgical splenectomy and has to be repeated when traveling in endemic areas.
care, may be due to the increasing recognition of ITP among older per- Finally, vaccination against influenza is recommended in winter. A ret-
sons [134,135], the greater risk of surgical complications in older pa- rospective case-control study showed a reduction in mortality and the
tients [136], and the willingness of surgeons to perform surgery in risk of OPSI in splenectomized patients vaccinated against influenza,
older patients. prompting strongly to follow these recommendations [146]. We note
that, in patients treated with rituximab in the previous 6 months, vacci-
5.2. Infectious risk nation may not be effective. Vaccination for these patients should be
readdressed once B-cell recovery has occurred [40].
The risk of severe sepsis throughout life, led some authors to propose
a course of antibiotics of indefinite duration. Patient adherence to such
1. Epidemiology of infections after splenectomy recommendations however, has not been studied which led some phy-
2. Characteristics of ‘OPSI’ sicians to limit the duration of this antibiotic to a period of 2 years for
3. Vaccinations adults and 5 years for children. These recommendations are not based
4. Preventive antibiotic therapy on controlled trials. In fact, continuation of antibiotics should be
5. Patient education
discussed in consideration of persistent risk factors for infection, severe
Splenectomized patients are exposed to an increased risk of severe immunosuppression or history of invasive pneumococcal infection
infections, particularly with encapsulated bacteria. This is due to the re- [147]. Some authors propose to develop a better predictive approach
moval of a large pool of B-lymphocytes, especially in the marginal zone, based on genetic profiling of patients with different susceptibility to in-
as well as to a global insufficiency in the phagocytosis of certain patho- fectious pathogens, host-pathogens interactions as well as to identify
gens [137]. The meta-analysis of Kojouri et al. estimated the prevalence the impact of factors such as age on immunological competence [148].
of infectious events to be 3.2% [42]. Pneumococcal infections are the The best prophylaxis to infection, however, is patient education. Pa-
most common infections, but splenectomy also exposes to other bacte- tients should be advised to go to the emergency department if fever
rial pathogens (Haemophilus influenzae type b, Neisseria meningitidis, higher than 38 °C occurs. Each splenectomized patient should have a
Escherichia coli, Capnocytophaga canimorsus), but also to fungal infec- card, reminding the medical corps of his statute and the measures that
tions and intra-erythrocytic pathogens (Babesia, Plasmodium should be taken in case of fever. If patients have fever greater than or
falciparum, Ehrlichia). A cohort study in Denmark on 3812 splenecto- equal to 38.5 °C, an antibiotic active against pneumococcal fever should
mies performed for various indications between 1966 and 2005 showed be immediately started. A practical policy for splenectomized patients is
a significant increase in the risk of infection within 90 days after sple- to have a home supply of broad spectrum antibiotics for use in case of a
nectomy (10.2% vs 0.6% in the general population) [138]. After one febrile illness [40,147].
year, the risk was much lower especially in the subgroup
splenectomized for ITP (relative risk 1.2), probably because of the effec-
tiveness of preventive measures (antibiotics, vaccination, patient infor- 5.3. Thrombotic risk
mation). Nevertheless, this study does not allow to assess the late
infection risk. The long-term risk of infection in ITP patients still has to
be evaluated.
The one major and feared complication associated with splenectomy 1. Venous thrombosis in the splenic and portal venous system
is the spectrum of overwhelming post-splenectomy infection (OPSI). 2. Venous thrombosis at distance of the spleen
The term OPSI defines fulminating sepsis, meningitis or pneumonia, 3. Pulmonary arterial hypertension
mainly caused by encapsulated bacteria, such as pneumococci, menin-
gococci and Haemophilus influenzae type b (Hib) [139–141]. OPSI has The risk of thrombosis after splenectomy is not well evaluated. The
a high mortality risk of approximately 40% to as much as 70% real incidence is not known, the pathophysiology is still debated and it
[141–143]. The risk is estimated at 0.89% per person per year [143]. is impossible to propose preventive measures based on solid evidence
Patients are usually given prophylactic polyvalent pneumococcal, [147]. One of the explanations for the increase of thrombotic risk after
meningococcal C conjugate and Hib vaccines at least 4 weeks before splenectomy is the loss of the role as filter (carried out by the red
(preferably) or 2 weeks after splenectomy and are revaccinated accord- pulp), suggested by studies that showed an increase in the amount of
ing to the country-specific recommendations [40]. These vaccines may circulating microparticles of platelet and red cell origin, with pro-
not be effective in patients who have received rituximab in the thrombotic properties [149,150].
6 months prior to splenectomy. Immediately after surgery, there is a risk of thrombosis in the splenic
Antigenic diversity of S. pneumoniae is significant (N 90 known an- and portal venous (SPV) system. The number of portal vein thrombosis
tigens) and is responsible for the difficulties in obtaining effective cover- varies from 5 to 37% [151–154].
age. Two types of vaccines are available, the unconjugated T-cell Occurrence of deep vein thrombosis (DVT) outside the SPV system
independent polysaccharide vaccine covering 23 serotypes or pulmonary embolism (PE) seems to be very low (0.63% and 0.73% re-
(Pneumo23®), and the conjugated vaccine covering 13 serotypes spectively) in the first 90 days after splenectomy according to data from
(Prévenar13®). The latter was originally developed to protect young the Danish National Patient registry. Nevertheless, the overall adjusted
children whose immature immune system and in particular the absence odds ratio for venous thromboembolism in splenectomized patients
of marginal zone in the spleen makes the polysaccharide vaccine ineffi- was 32.6 versus the general population and 3.2 versus appendectomy
cient before the age of 2 years. Response to polysaccharide vaccine is patients in the first 90 days after surgery, falling to 7.1 and 2.8 respec-
also reduced in splenectomized patients with more than a quarter of tively at 91–365 days, and 3.4 and 3.2 respectively, at N1 year [155].
poor antibody response in a cohort of splenectomized patients [144]. These data suggest a pro-thrombogenic effect of splenectomy.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 11

Prescribing prophylactic anticoagulation in patients splenectomized in • Treatment of ITP is confined to patients who are bleeding or are at
thrombogenic situation should be discussed on a case by case basis. significant risk of bleeding.
Splenectomy is also a newly identified risk factor for the develop- • First-line therapy for ITP consists of corticosteroids.
ment of post-embolic pulmonary arterial hypertension (PAH). In a • Second-line therapies for ITP are splenectomy, rituximab and TPO
study conducted by Jaïs et al. [156] involving N250 patients with post- receptor agonists (comparison: Table 2).
embolic pulmonary hypertension, the prevalence of splenectomy was • International guidelines fail to offer a consistent approach about
8.6% while this prevalence was significantly lower in patients with second-line therapy. Treatment should be personalized (proposed
other chronic lung diseases. Post-embolic PAH occurred on average treatment algorithm: Fig. 2).
16 ± 9 years after splenectomy and almost exclusively in patients • There are no clinical relevant variables that predict the success of
with a hemolytic anemia or who had been splenectomized in the after- splenectomy.
math of trauma, while none of the patients in this study had ITP. The risk • The best timing for splenectomy is debatable.
of post-embolic pulmonary hypertension in ITP remains to be deter-
mined and certainly does not warrant a systematic cardiac ultrasound 2. Autoimmune hemolytic anemia
monitoring as proposed in other diseases at risk of PAH. • Treatment is essentially experience-based.
• Corticosteroids are the mainstay of first-line treatment.
6. Conclusion • Splenectomy or rituximab are the second-line treatment options.
• There are no clinical factors significantly predicting response on
The autoimmune disorders ITP, AIHA and TTP are three rare but in- splenectomy.
teresting conditions with a complex pathogenesis and a difficult man- • Rituximab combined with corticosteroids can increase both the rate
agement. In recent years, major advances in understanding the and the duration of response. Further research is warranted.
immunopathogenesis has led to the introduction of new therapies, stag-
gering the role of splenectomy as standard second-line treatment. Ri- 3. Thrombotic thrombocytopenic purpura
tuximab and TPO receptor agonists could be valid alternatives to • Pathophysiology is largely explained by a deficiency of ADAMTS13.
splenectomy but, unfortunately, the international guidelines fail to • First-line treatment is plasma exchange therapy combined with
offer a consistent approach. Whereas the European guidelines consider corticosteroids.
splenectomy at the same level of rituximab and TPO RAs, the ASH guide- • The preferred second-line treatment is rituximab.
line definitely recommends reserving TPO RAs and rituximab to pa- • Splenectomy seems to play a minor role in the treatment of TTP, al-
tients failing or with a contra-indication to splenectomy. though in small studies the efficacy is demonstrated.
In warm AIHA, guidelines still propose splenectomy as best second- • Caplacizumab, an anti-von Willebrand factor humanized immuno-
line therapy. However, in some studies with low-dose rituximab in globulin, induces a faster resolution of the acute TTP episode. It
combination with steroids, success rates similar to splenectomy were has the potential to become a new treatment of TTP in addition to
attained in steroid-refractory patients. In TTP, rituximab conquered PEX en steroids.
the throne of standard second-line therapy. Splenectomy seems to
play a minor role in the treatment of TTP, although in small studies 4. Splenectomy
the efficacy and safety of splenectomy is demonstrated. The great vari- • Splenectomy is irreversible and has 3 major categories of important
ability of opinion and interpretation of current data, shows that treat- complications: postoperative, infectious and thrombotic.
ment should be personalized and adapted to the patient's condition • Mortality rate has decreased with the laparoscopic approach.
and opinion. Unfortunately, of all factors proposed as predictors of re- • The infectious risk can be reduced but not erased by preventive
sponse to splenectomy in ITP and AIHA, none was found to be statistical- measures (e.g. vaccinations) and the occurrence of OPSI remains a
ly significant. Therefore, selection of patients remains to be based on big fear. The best prophylaxis is patient education.
their surgical risk, their possible contra-indications and the personal • Splenectomy causes an increased risk of thrombosis in the splenic
preferences of patient and physician. and portal venous (SPV) system and at distance of the spleen.
While the complication rate of splenectomy is still relatively high,
the mortality rate has considerably decreased with the introduction of
Research agenda
a laparoscopic approach. However, the occurrence of infections, espe-
cially OPSI, remains a big fear of many physicians. Several preventive
• Evidence in our review came mainly from small retrospective studies.
measures, about which no consensus is reached, can be undertaken,
Future research must be focused on carefully designed randomized
but the risk of infection persists a whole lifetime and education of pa-
trials and multicentre prospective cohort studies.
tients about this is indispensable.
• There is a need of standardization of terminology such as partial and
It is reasonable to expect that, the more the pathogenesis is clarified,
complete response, refractoriness and relapse.
the more specific targets for therapy will arise. It needs to be seen in the
• Better comprehension of pathogenesis will reveal more specific tar-
future if new therapies will surpass splenectomy or if on the other hand,
gets for therapy in the future.
possible complications of splenectomy will become better prevented,
making this old treatment option more popular. In the meanwhile, sple-
Conflict of interest
nectomy certainly stays worth considering.
It is notable that evidence in our review came mainly from small ret-
None of the authors declare any conflicts of interest.
rospective studies. Consequently, future research must be focused on
carefully designed randomized trials and multicentre prospective co-
Acknowledgement
hort studies. Furthermore, there is a need of standardization of termi-
nology such as partial and complete response, refractoriness and
The authors wish to thank Dr. Emilie Beke and Esther Sys for their
relapse, so that study results can be better compared.
help with the figures.

Practice points References


[1] Rodeghiero F, Ruggeri M. Is splenectomy still the gold standard for the treatment of
1. Immune thrombocytopenia chronic ITP? Am J Hematol 2008;83(2):91.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
12 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

[2] Palandri F, Polverelli N, Sollazzo D, et al. Have splenectomy rate and main outcomes hyperdestructive and hypoplastic thrombocytopenia. Am J Clin Pathol 2001;
of ITP changed after the introduction of new treatments? A monocentric study in 115(5):656–64.
the outpatient setting during 35 years. Am J Hematol 2016;91(4):E267–72. [36] Porcelijn L, Folman CC, Bossers B, et al. The diagnostic value of thrombopoietin level
[3] Stasi R, Provan D. Management of immune thrombocytopenic purpura in adults. measurements in thrombocytopenia. Thromb Haemost 1998;79(6):1101–5.
Mayo Clin Proc 2004;79(4):504–22. [37] George JN. Definition, diagnosis and treatment of immune thrombocytopenic pur-
[4] Rodeghiero F, Stasi R, Gernsheimer T, et al. Standardization of terminology, defini- pura. Haematologica 2009;94(6):759–62.
tions and outcome criteria in immune thrombocytopenic purpura of adults and [38] Cheng Y, Wong RS, Soo YO, et al. Initial treatment of immune thrombocytopenic
children: report from an international working group. Blood 2009;113(11): purpura with high-dose dexamethasone. N Engl J Med 2003;349(9):831–6.
2386–93. [39] Thota S, Kistangari G, Daw H, et al. Immune thrombocytopenia in adults: an update.
[5] Schoonen WM, Kucera G, Coalson J, et al. Epidemiology of immune thrombocyto- Cleve Clin J Med 2012;79(9):641–50.
penic purpura in the general practice research database. Br J Haematol 2009; [40] Provan D, Stasi R, Newland AC, et al. International consensus report on the investi-
145(2):235–44. gation and management of primary immune thrombocytopenia. Blood 2010;
[6] Kurata Y, Fujimura K, Kuwana M, et al. Epidemiology of primary immune thrombo- 115(2):168–86.
cytopenia in children and adults in Japan: a population-based study and literature [41] Neunert C, Lim W, Crowther M, et al. The American Society of Hematology 2011
review. Int J Hematol 2011;93(3):329–35. evidence-based practice guideline for immune thrombocytopenia. Blood 2011;
[7] Harrington WJ, Minnich V, Hollingsworth JW, et al. Demonstration of a thrombocy- 117(16):4190–207.
topenic factor in the blood of patients with thrombocytopenic purpura. J Lab Clin [42] Kojouri K, Vesely SK, Terrell DR, et al. Splenectomy for adult patients with idiopath-
Med 1951;38(1):1–10. ic thrombocytopenic purpura: a systematic review to assess long-term platelet
[8] McMillan R. The pathogenesis of chronic immune thrombocytopenic purpura. count responses, prediction of response, and surgical complications. Blood 2004;
Semin Hematol 2007;44(4 Suppl. 5):S3–S11. 104(9):2623–34.
[9] Cines DB, Cuker A, Semple JW. Pathogenesis of immune thrombocytopenia. Presse [43] Vianelli N, Galli M, de Vivo A, et al. Efficacy and safety of splenectomy in immune
Med 2014;43(4 Pt 2):e49–59. thrombocytopenic purpura: long-term results of 402 cases. Haematologica 2005;
[10] Najaoui A, Bakchoul T, Stoy J, et al. Autoantibody-mediated complement activation 90(1):72–7.
on platelets is a common finding in patients with immune thrombocytopenic pur- [44] Fabris F, Tassan T, Ramon R, et al. Age as the major predictive factor of long-term
pura (ITP). Eur J Haematol 2012;88(2):167–74. response to splenectomy in immune thrombocytopenic purpura. Br J Haematol
[11] Gu D, Ge J, Du W, et al. Raised expression of APRIL in Chinese patients with immune 2001;112(3):637–40.
thrombocytopenia and its clinical implications. Autoimmunity 2009;42(8):692–8. [45] Schwartz J, Leber MD, Gillis S, et al. Long term follow-up after splenectomy per-
[12] Zhu XJ, Shi Y, Peng J, et al. The effects of BAFF and BAFF-R-Fc fusion protein in im- formed for immune thrombocytopenic purpura (ITP). Am J Hematol 2003;72(2):
mune thrombocytopenia. Blood 2009;114(26):5362–7. 94–8.
[13] Li X, Zhong H, Bao W, et al. Defective regulatory B-cell compartment in patients [46] Balagué C, Vela S, Targarona EM, et al. Predictive factors for successful laparoscopic
with immune thrombocytopenia. Blood 2012;120(16):3318–25. splenectomy in immune thrombocytopenic purpura: study of clinical and laborato-
[14] Kuwana M, Kaburaki J, Ikeda Y. Autoreactive T cells to platelet GPIIb-IIIa in immune ry data. Surg Endosc 2006;20(8):1208–13.
thrombocytopenic purpura. Role in production of anti-platelet autoantibody. J Clin [47] Ojima H, Kato T, Araki K, et al. Factors predicting long-term responses to splenec-
Invest 1998;102(7):1393–402. tomy in patients with idiopathic thrombocytopenic purpura. World J Surg 2006;
[15] Kuwana M, Kaburaki J, Kitasato H, et al. Immunodominant epitopes on glycopro- 30(4):553–9.
tein IIb-IIIa recognized by autoreactive T cells in patients with immune thrombocy- [48] Syed NN, Adil SN, Sajid R, et al. Chronic ITP: analysis of various factors at presenta-
topenic purpura. Blood 2001;98(1):130–9. tion which predict failure to first line treatment and their response to second line
[16] Olsson B, Andersson PO, Jernås M, et al. T-cell-mediated cytotoxicity toward plate- therapy. J Pak Med Assoc 2007;57(3):126–9.
lets in chronic idiopathic thrombocytopenic purpura. Nat Med 2003;9(9):1123–4. [49] Shojaiefard A, Mousavi SA, Faghihi SH, et al. Prediction of response to splenectomy
[17] Stasi R. Pathophysiology and therapeutic options in primary immune thrombocy- in patients with idiopathic thrombocytopenic purpura. World J Surg 2008;32(3):
topenia. Blood Transfus 2011;9(3):262–73. 488–93.
[18] Panitsas FP, Theodoropoulou M, Kouraklis A, et al. Adult chronic idiopathic throm- [50] Peters AM, Saverymuttu SH, Wonke B, et al. The interpretation of platelet kinetic
bocytopenic purpura (ITP) is the manifestation of a type-1 polarized immune re- studies for the identification of sites of abnormal platelet destruction. Br J Haematol
sponse. Blood 2004;103(7):2645–7. 1984;57(4):637–49.
[19] Hu Y, Ma DX, Shan NN, et al. Increased number of Tc17 and correlation with Th17 [51] Cuker A, Cines DB. Evidence-based mini-review: is indium-labeled autologous
cells in patients with immune thrombocytopenia. PLoS One 2011;6(10):e26522. platelet scanning predictive of response to splenectomy in patients with chronic
[20] Hu Y, Li H, Zhang L, et al. Elevated profiles of Th22 cells and correlations with Th17 immune thrombocytopenia? Hematology Am Soc Hematol Educ Program 2010;
cells in patients with immune thrombocytopenia. Hum Immunol 2012;73(6): 2010:385–6.
629–35. [52] Stasi R, Stipa E, Masi M, et al. Long-term observation of 208 adults with chronic id-
[21] Cao J, Chen C, Li L, et al. Effects of high-dose dexamethasone on regulating iopathic thrombocytopenic purpura. Am J Med 1995;98(5):436–42.
interleukin-22 production and correcting Th1 and Th22 polarization in immune [53] Sailer T, Lechner K, Panzer S, et al. The course of severe autoimmune thrombocyto-
thrombocytopenia. J Clin Immunol 2012;32(3):523–9. penia in patients not undergoing splenectomy. Haematologica 2006;91(8):1041–5.
[22] Liu B, Zhao H, Poon MC, et al. Abnormality of CD4(+)CD25(+) regulatory T cells in [54] Dierickx D, Beke E, Devos T, et al. The use of monoclonal antibodies in immune-
idiopathic thrombocytopenic purpura. Eur J Haematol 2007;78(2):139–43. mediated hematologic disorders. Med Clin North Am 2012;96(3):583–619 [xi].
[23] Olsson B, Andersson PO, Jacobsson S, et al. Disturbed apoptosis of T-cells in patients [55] Arnold DM, Dentali F, Crowther MA, et al. Systematic review: efficacy and safety of
with active idiopathic thrombocytopenic purpura. Thromb Haemost 2005;93(1): rituximab for adults with idiopathic thrombocytopenic purpura. Ann Intern Med
139–44. 2007;146(1):25–33.
[24] Kashiwagi H, Tomiyama Y. Pathophysiology and management of primary immune [56] Godeau B, Porcher R, Fain O, et al. Rituximab efficacy and safety in adult splenecto-
thrombocytopenia. Int J Hematol 2013;98(1):24–33. my candidates with chronic immune thrombocytopenic purpura: results of a pro-
[25] Crow AR, Song S, Siragam V, et al. Mechanisms of action of intravenous immuno- spective multicenter phase 2 study. Blood 2008;112(4):999–1004.
globulin in the treatment of immune thrombocytopenia. Pediatr Blood Cancer [57] Medeot M, Zaja F, Vianelli N, et al. Rituximab therapy in adult patients with re-
2006;47(Suppl. 5):710–3. lapsed or refractory immune thrombocytopenic purpura: long-term follow-up re-
[26] Asahi A, Nishimoto T, Okazaki Y, et al. Helicobacter pylori eradication shifts mono- sults. Eur J Haematol 2008;81(3):165–9.
cyte Fcgamma receptor balance toward inhibitory FcgammaRIIB in immune [58] Ghanima WKA, Waage A, et al. Rituximab as second-line treatment for adult im-
thrombocytopenic purpura patients. J Clin Invest 2008;118(8):2939–49. mune thrombocytopenia (the RITP trial): a multicentre, randomised, double-
[27] Liu XG, Ma SH, Sun JZ, et al. High-dose dexamethasone shifts the balance of stimu- blind, placebo-controlled trial. Lancet 2015;385(9978):1653–61.
latory and inhibitory Fcgamma receptors on monocytes in patients with primary [59] Cheng G, Saleh MN, Marcher C, et al. Eltrombopag for management of chronic im-
immune thrombocytopenia. Blood 2011;117(6):2061–9. mune thrombocytopenia (RAISE): a 6-month, randomised, phase 3 study. Lancet
[28] Tsubakio T, Tani P, Curd JG, et al. Complement activation in vitro by antiplatelet an- 2011;377(9763):393–402.
tibodies in chronic immune thrombocytopenic purpura. Br J Haematol 1986;63(2): [60] Saleh MN, Bussel JB, Cheng G, et al. Safety and efficacy of eltrombopag for treat-
293–300. ment of chronic immune thrombocytopenia: results of the long-term, open-label
[29] Peerschke EI, Andemariam B, Yin W, et al. Complement activation on platelets cor- EXTEND study. Blood 2013;121(3):537–45.
relates with a decrease in circulating immature platelets in patients with immune [61] Kuter DJ, Bussel JB, Lyons RM, et al. Efficacy of romiplostim in patients with chronic
thrombocytopenic purpura. Br J Haematol 2010;148(4):638–45. immune thrombocytopenic purpura: a double-blind randomised controlled trial.
[30] Nardi M, Tomlinson S, Greco MA, et al. Complement-independent, peroxide- Lancet 2008;371(9610):395–403.
induced antibody lysis of platelets in HIV-1-related immune thrombocytopenia. [62] Bussel JB, Kuter DJ, Pullarkat V, et al. Safety and efficacy of long-term treatment
Cell 2001;106(5):551–61. with romiplostim in thrombocytopenic patients with chronic ITP. Blood 2009;
[31] Chow L, Aslam R, Speck ER, et al. A murine model of severe immune thrombocyto- 113(10):2161–71.
penia is induced by antibody- and CD8+ T cell-mediated responses that are differ- [63] Kuter DJ, Bussel JB, Newland A, et al. Long-term treatment with romiplostim in pa-
entially sensitive to therapy. Blood 2010;115(6):1247–53. tients with chronic immune thrombocytopenia: safety and efficacy. Br J Haematol
[32] Chang M, Nakagawa PA, Williams SA, et al. Immune thrombocytopenic 2013;161(3):411–23.
purpura (ITP) plasma and purified ITP monoclonal autoantibodies inhibit [64] Provan D, Newland AC. Current management of primary immune thrombocytope-
megakaryocytopoiesis in vitro. Blood 2003;102(3):887–95. nia. Adv Ther 2015;32(10):875–87.
[33] McMillan R, Wang L, Tomer A, et al. Suppression of in vitro megakaryocyte produc- [65] Bussel JB, Provan D, Shamsi T, et al. Effect of eltrombopag on platelet counts and
tion by antiplatelet autoantibodies from adult patients with chronic ITP. Blood bleeding during treatment of chronic idiopathic thrombocytopenic purpura: a
2004;103(4):1364–9. randomised, double-blind, placebo-controlled trial. Lancet 2009;373(9664):641–8.
[34] Houwerzijl EJ, Blom NR, van der Want JJ, et al. Ultrastructural study shows morpho- [66] Chaplin H, Avioli LV. Grand rounds: autoimmune hemolytic anemia. Arch Intern
logic features of apoptosis and para-apoptosis in megakaryocytes from patients Med 1977;137(3):346–51.
with idiopathic thrombocytopenic purpura. Blood 2004;103(2):500–6. [67] Sokol RJ, Hewitt S, Stamps BK. Autoimmune haemolysis: an 18-year study of 865
[35] Kurata Y, Hayashi S, Kiyoi T, et al. Diagnostic value of tests for reticulated platelets, cases referred to a regional transfusion centre. Br Med J (Clin Res Ed) 1981;
plasma glycocalicin, and thrombopoietin levels for discriminating between 282(6281):2023–7.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx 13

[68] Barros MM, Blajchman MA, Bordin JO. Warm autoimmune hemolytic anemia: re- [102] Quartier P, Brethon B, Philippet P, et al. Treatment of childhood autoimmune
cent progress in understanding the immunobiology and the treatment. Transfus haemolytic anaemia with rituximab. Lancet 2001;358(9292):1511–3.
Med Rev 2010;24(3):195–210. [103] Zecca M, Nobili B, Ramenghi U, et al. Rituximab for the treatment of refractory au-
[69] Barker RN, Casswell KM, Reid ME, et al. Identification of autoantigens in autoim- toimmune hemolytic anemia in children. Blood 2003;101(10):3857–61.
mune haemolytic anaemia by a non-radioisotope immunoprecipitation method. J [104] Dierickx D, Kentos A, Delannoy A. The role of rituximab in adults with warm anti-
Haematol]–>Br J Haematol 1992;82(1):126–32. body autoimmune hemolytic anemia. Blood 2015;125(21):3223–9.
[70] Leddy JP, Falany JL, Kissel GE, et al. Erythrocyte membrane proteins reactive with [105] Barcellini W, Zaja F, Zaninoni A, et al. Low-dose rituximab in adult patients with id-
human (warm-reacting) anti-red cell autoantibodies. J Clin Invest 1993;91(4): iopathic autoimmune hemolytic anemia: clinical efficacy and biologic studies.
1672–80. Blood 2012;119(16):3691–7.
[71] Barker RN, Elson CJ. Multiple self epitopes on the Rhesus polypeptides stimulate [106] Birgens H, Frederiksen H, Hasselbalch HC, et al. A phase III randomized trial com-
immunologically ignorant human T cells in vitro. Eur J Immunol 1994;24(7): paring glucocorticoid monotherapy versus glucocorticoid and rituximab in patients
1578–82. with autoimmune haemolytic anaemia. Br J Haematol 2013;163(3):393–9.
[72] Barker RN, Hall AM, Standen GR, et al. Identification of T-cell epitopes on the [107] Blombery P, Scully M. Management of thrombotic thrombocytopenic purpura: cur-
Rhesus polypeptides in autoimmune hemolytic anemia. Blood 1997;90(7): rent perspectives. J Blood Med 2014;5:15–23.
2701–15. [108] Tsai HM. von Willebrand factor, shear stress, and ADAMTS13 in hemostasis and
[73] Elson CJ, Barker RN, Thompson SJ, et al. Immunologically ignorant autoreactive T thrombosis. ASAIO J 2012;58(2):163–9.
cells, epitope spreading and repertoire limitation. Immunol Today 1995;16(2): [109] Kremer Hovinga JA, Lämmle B. Role of ADAMTS13 in the pathogenesis, diagnosis,
71–6. and treatment of thrombotic thrombocytopenic purpura. Hematology Am Soc
[74] Molina H, Miwa T, Zhou L, et al. Complement-mediated clearance of erythrocytes: Hematol Educ Program 2012;2012:610–6.
mechanism and delineation of the regulatory roles of Crry and DAF. Decay- [110] Zheng XL, Kaufman RM, Goodnough LT, et al. Effect of plasma exchange on plasma
accelerating factor. Blood 2002;100(13):4544–9. ADAMTS13 metalloprotease activity, inhibitor level, and clinical outcome in pa-
[75] Barros MM, Yamamoto M, Figueiredo MS, et al. Expression levels of CD47, CD35, tients with idiopathic and nonidiopathic thrombotic thrombocytopenic purpura.
CD55, and CD59 on red blood cells and signal-regulatory protein-alpha,beta on Blood 2004;103(11):4043–9.
monocytes from patients with warm autoimmune hemolytic anemia. Transfusion [111] Török TJ, Holman RC, Chorba TL. Increasing mortality from thrombotic thrombocy-
2009;49(1):154–60. topenic purpura in the United States—analysis of national mortality data,
[76] Richaud-Patin Y, Pérez-Romano B, Carrillo-Maravilla E, et al. Deficiency of red cell 1968–1991. Am J Hematol 1995;50(2):84–90.
bound CD55 and CD59 in patients with systemic lupus erythematosus. Immunol [112] Rock GA, Shumak KH, Buskard NA, et al. Comparison of plasma exchange with plas-
Lett 2003;88(2):95–9. ma infusion in the treatment of thrombotic thrombocytopenic purpura. Canadian
[77] Ohashi PS, DeFranco AL. Making and breaking tolerance. Curr Opin Immunol 2002; Apheresis Study Group. N Engl J Med 1991;325(6):393–7.
14(6):744–59. [113] Hayward CP, Sutton DM, Carter WH, et al. Treatment outcomes in patients with
[78] Barker RN, Vickers MA, Ward FJ. Controlling autoimmunity—lessons from the study adult thrombotic thrombocytopenic purpura-hemolytic uremic syndrome. Arch
of red blood cells as model antigens. Immunol Lett 2007;108(1):20–6. Intern Med 1994;154(9):982–7.
[79] Musaji A, Meite M, Detalle L, et al. Enhancement of autoantibody pathogenicity by [114] Bell WR, Braine HG, Ness PM, et al. Improved survival in thrombotic thrombocyto-
viral infections in mouse models of anemia and thrombocytopenia. Autoimmun penic purpura-hemolytic uremic syndrome. Clinical experience in 108 patients. J
Rev 2005;4(4):247–52. Med]–>N Engl J Med 1991;325(6):398–403.
[80] Bordin JO, Kerbauy J, Souza-Pinto JC, et al. Quantitation of red cell-bound IgG by an [115] Allan DS, Kovacs MJ, Clark WF. Frequently relapsing thrombotic thrombocytopenic
enzyme-linked antiglobulin test in human immunodeficiency virus-infected per- purpura treated with cytotoxic immunosuppressive therapy. Haematologica 2001;
sons. Transfusion 1992;32(5):426–9. 86(8):844–50.
[81] Shen CR, Mazza G, Perry FE, et al. T-helper 1 dominated responses to erythrocyte [116] Cuttner J. Thrombotic thrombocytopenic purpura: a ten-year experience. Blood
Band 3 in NZB mice. Immunology 1996;89(2):195–9. 1980;56(2):302–6.
[82] Hall AM, Ward FJ, Vickers MA, et al. Interleukin-10-mediated regulatory T-cell re- [117] Bell WR. Thrombotic thrombocytopenic purpura/hemolytic uremic syndrome re-
sponses to epitopes on a human red blood cell autoantigen. Blood 2002;100(13): lapse: frequency, pathogenesis, and meaning. Semin Hematol 1997;34(2):134–9.
4529–36. [118] Balduini CL, Gugliotta L, Luppi M, et al. High versus standard dose methylprednis-
[83] Youssef AR, Shen CR, Lin CL, et al. IL-4 and IL-10 modulate autoimmune haemolytic olone in the acute phase of idiopathic thrombotic thrombocytopenic purpura: a
anaemia in NZB mice. Clin Exp Immunol 2005;139(1):84–9. randomized study. Ann Hematol 2010;89(6):591–6.
[84] Willerford DM, Chen J, Ferry JA, et al. Interleukin-2 receptor alpha chain regulates [119] Peyvandi F, Scully M, Kremer Hovinga JA, et al. Caplacizumab for acquired throm-
the size and content of the peripheral lymphoid compartment. Immunity 1995; botic thrombocytopenic purpura. N Engl J Med 2016;374(6):511–22.
3(4):521–30. [120] Dierickx D, Delannoy A, Saja K, et al. Anti-CD20 monoclonal antibodies and their
[85] Lechner K, Jäger U. How I treat autoimmune hemolytic anemias in adults. Blood use in adult autoimmune hematological disorders. Am J Hematol 2011;86(3):
2010;116(11):1831–8. 278–91.
[86] Murphy S, LoBuglio AF. Drug therapy of autoimmune hemolytic anemia. Semin [121] Caramazza D, Quintini G, Abbene I, et al. Relapsing or refractory idiopathic throm-
Hematol 1976;13(4):323–34. botic thrombocytopenic purpura-hemolytic uremic syndrome: the role of rituxi-
[87] Gehrs BC, Friedberg RC. Autoimmune hemolytic anemia. Am J Hematol 2002; mab. Transfusion 2010;50(12):2753–60.
69(4):258–71. [122] Chemnitz JM, Uener J, Hallek M, et al. Long-term follow-up of idiopathic thrombot-
[88] Allgood JW, Chaplin H. Idiopathic acquired autoimmune hemolytic anemia. ic thrombocytopenic purpura treated with rituximab. Ann Hematol 2010;89(10):
A review of forty-seven cases treated from 1955 through 1965. Am J Med 1967; 1029–33.
43(2):254–73. [123] Scully M, McDonald V, Cavenagh J, et al. A phase 2 study of the safety and efficacy
[89] Packman CH. Hemolytic anemia due to warm autoantibodies. Blood Rev 2008; of rituximab with plasma exchange in acute acquired thrombotic thrombocytope-
22(1):17–31. nic purpura. Blood 2011;118(7):1746–53.
[90] Serrano J. Autoimmune hemolytic anemia. Review of 200 cases studied in a period [124] Scully M, Hunt BJ, Benjamin S, et al. Guidelines on the diagnosis and management
of 20 years (1970–1989). Sangre (Barc) 1992;37(4):265–74. of thrombotic thrombocytopenic purpura and other thrombotic microangiopa-
[91] Chertkow G, Dacie JV. Results of splenectomy in auto-immune haemolytic anae- thies. Br J Haematol 2012;158(3):323–35.
mia. Br J Haematol 1956;2(3):237–49. [125] Bernard RP, Bauman AW, Schwartz SI. Splenectomy for thrombotic thrombocyto-
[92] Genty I, Michel M, Hermine O, et al. Characteristics of autoimmune hemolytic ane- penic purpura. Ann Surg 1969;169(4):616–24.
mia in adults: retrospective analysis of 83 cases. Rev Med Interne 2002;23(11): [126] Bukowski RM, Hewlett JS, Reimer RR, et al. Therapy of thrombotic thrombocytope-
901–9. nic purpura: an overview. Semin Thromb Hemost 1981;7(1):1–8.
[93] Akpek G, McAneny D, Weintraub L. Comparative response to splenectomy in [127] Schneider PA, Rayner AA, Linker CA, et al. The role of splenectomy in multimodality
Coombs-positive autoimmune hemolytic anemia with or without associated dis- treatment of thrombotic thrombocytopenic purpura. Ann Surg 1985;202(3):
ease. Am J Hematol 1999;61(2):98–102. 318–22.
[94] Coon WW. Splenectomy in the treatment of hemolytic anemia. Arch Surg 1985; [128] Rutkow IM. Thrombotic thrombocytopenic purpura (TTP) and splenectomy: a cur-
120(5):625–8. rent appraisal. Ann Surg 1978;188(5):701–5.
[95] Jandl JH, Greenberg MS, Yonemoto RH, et al. Clinical determination of the sites [129] Aqui NA, Stein SH, Konkle BA, et al. Role of splenectomy in patients with refractory
of red cell sequestration in hemolytic anemias. J Clin Invest 1956;35(8): or relapsed thrombotic thrombocytopenic purpura. J Clin Apher 2003;18(2):51–4.
842–67. [130] Dubois L, Gray DK. Case series: splenectomy: does it still play a role in the manage-
[96] McCurdy PR, Rath CE. Splenectomy in hemolytic anemia; results predicted by body ment of thrombotic thrombocytopenic purpura? Can J Surg 2010;53(5):349–55.
scanning after injection of Cr51-tagged red cells. N Engl J Med 1958;259(10): [131] Kappers-Klunne MC, Wijermans P, Fijnheer R, et al. Splenectomy for the treatment
459–63. of thrombotic thrombocytopenic purpura. Br J Haematol 2005;130(5):768–76.
[97] Bussone G, Ribeiro E, Dechartres A, et al. Efficacy and safety of rituximab in adults' [132] Böhm M, Betz C, Miesbach W, et al. The course of ADAMTS-13 activity and inhibitor
warm antibody autoimmune haemolytic anemia: retrospective analysis of 27 titre in the treatment of thrombotic thrombocytopenic purpura with plasma ex-
cases. Am J Hematol 2009;84(3):153–7. change and vincristine. Br J Haematol 2005;129(5):644–52.
[98] D'Arena G, Laurenti L, Capalbo S, et al. Rituximab therapy for chronic lymphocytic [133] Dolan JP, Sheppard BC, DeLoughery TG. Splenectomy for immune thrombocytope-
leukemia-associated autoimmune hemolytic anemia. Am J Hematol 2006;81(8): nic purpura: surgery for the 21st century. Am J Hematol 2008;83(2):93–6.
598–602. [134] Neylon AJ, Saunders PW, Howard MR, et al. Clinically significant newly presenting
[99] D'Arena G, Califano C, Annunziata M, et al. Rituximab for warm-type idiopathic au- autoimmune thrombocytopenic purpura in adults: a prospective study of a
toimmune hemolytic anemia: a retrospective study of 11 adult patients. Eur J population-based cohort of 245 patients. Br J Haematol 2003;122(6):966–74.
Haematol 2007;79(1):53–8. [135] Frederiksen H, Schmidt K. The incidence of idiopathic thrombocytopenic purpura
[100] Narat S, Gandla J, Hoffbrand AV, et al. Rituximab in the treatment of refractory au- in adults increases with age. Blood 1999;94(3):909–13.
toimmune cytopenias in adults. Haematologica 2005;90(9):1273–4. [136] Portielje JE, Westendorp RG, Kluin-Nelemans HC, et al. Morbidity and mortality in
[101] Peñalver FJ, Alvarez-Larrán A, Díez-Martin JL, et al. Rituximab is an effective and adults with idiopathic thrombocytopenic purpura. Blood 2001;97(9):2549–54.
safe therapeutic alternative in adults with refractory and severe autoimmune he- [137] Wasserstrom H, Bussel J, Lim LC, et al. Memory B cells and pneumococcal antibody
molytic anemia. Ann Hematol 2010;89(11):1073–80. after splenectomy. J Immunol 2008;181(5):3684–9.

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001
14 J. Sys et al. / Blood Reviews xxx (2017) xxx–xxx

[138] Thomsen RW, Schoonen WM, Farkas DK, et al. Risk for hospital contact with infec- [148] Kaplinsky C, Spirer Z. Post-splenectomy antibiotic prophylaxis—unfinished story:
tion in patients with splenectomy: a population-based cohort study. Ann Intern to treat or not to treat? Pediatr Blood Cancer 2006;47(Suppl. 5):740–1.
Med 2009;151(8):546–55. [149] Fontana V, Jy W, Ahn ER, et al. Increased procoagulant cell-derived microparticles
[139] Schwartz PE, Sterioff S, Mucha P, et al. Postsplenectomy sepsis and mortality in (C-MP) in splenectomized patients with ITP. Thromb Res 2008;122(5):599–603.
adults. JAMA 1982;248(18):2279–83. [150] Sewify EM, Sayed D, Abdel Aal RF, et al. Increased circulating red cell microparticles
[140] Ejstrud P, Kristensen B, Hansen JB, et al. Risk and patterns of bacteraemia after sple- (RMP) and platelet microparticles (PMP) in immune thrombocytopenic purpura.
nectomy: a population-based study. Scand J Infect Dis 2000;32(5):521–5. Thromb Res 2013;131(2):e59–63.
[141] Bisharat N, Omari H, Lavi I, et al. Risk of infection and death among post- [151] Ikeda M, Sekimoto M, Takiguchi S, et al. High incidence of thrombosis of the portal
splenectomy patients. J Infect 2001;43(3):182–6. venous system after laparoscopic splenectomy: a prospective study with contrast-
[142] Holdsworth RJ, Irving AD, Cuschieri A. Postsplenectomy sepsis and its mortality enhanced CT scan. Ann Surg 2005;241(2):208–16.
rate: actual versus perceived risks. Br J Surg 1991;78(9):1031–8. [152] Ikeda M, Sekimoto M, Takiguchi S, et al. Total splenic vein thrombosis after laparo-
[143] Kyaw MH, Holmes EM, Toolis F, et al. Evaluation of severe infection and survival scopic splenectomy: a possible candidate for treatment. Am J Surg 2007;193(1):
after splenectomy. Am J Med 2006;119(3):276.e1–7. 21–5.
[144] Cherif H, Landgren O, Konradsen HB, et al. Poor antibody response to pneumococ- [153] Hassn AM, Al-Fallouji MA, Ouf TI, et al. Portal vein thrombosis following splenecto-
cal polysaccharide vaccination suggests increased susceptibility to pneumococcal my. Br J Surg 2000;87(3):362–73.
infection in splenectomized patients with hematological diseases. Vaccine 2006; [154] Stamou KM, Toutouzas KG, Kekis PB, et al. Prospective study of the incidence and
24(1):75–81. risk factors of postsplenectomy thrombosis of the portal, mesenteric, and splenic
[145] (CDC) CfDCaP. Use of 13-valent pneumococcal conjugate vaccine and 23-valent veins. Arch Surg 2006;141(7):663–9.
pneumococcal polysaccharide vaccine for adults with immunocompromising con- [155] Thomsen RW, Schoonen WM, Farkas DK, et al. Risk of venous thromboembolism in
ditions: recommendations of the Advisory Committee on Immunization Practices splenectomized patients compared with the general population and
(ACIP). MMWR Morb Mortal Wkly Rep 2012;61(40):816–9. appendectomized patients: a 10-year nationwide cohort study. J Thromb Haemost
[146] Langley JM, Dodds L, Fell D, et al. Pneumococcal and influenza immunization in 2010;8(6):1413–6.
asplenic persons: a retrospective population-based cohort study 1990–2002. [156] Jaïs X, Ioos V, Jardim C, et al. Splenectomy and chronic thromboembolic pulmonary
BMC Infect Dis 2010;10:219. hypertension. Thorax 2005;60(12):1031–4.
[147] Mahévas M, Coignard-Biehler H, Michel M, et al. Post-splenectomy complications
in primary immune thrombocytopenia. Literature review and preventive mea-
sures. Rev Med Interne 2014;35(6):382–7 [in French].

Please cite this article as: Sys J, et al, The role of splenectomy in autoimmune hematological disorders: Outdated or still worth considering?, Blood
Rev (2017), http://dx.doi.org/10.1016/j.blre.2017.01.001

You might also like