Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 1.
Published in final edited form as: Leuk Res. 2013 May 18;37(8):894–899. doi: 10.1016/j.leukres.2013.04.007

Myelodysplastic Syndromes and Autoimmune Diseases - Case Series and Review of Literature

Omar Al Ustwani 1, Laurie A Ford 1, Sheila JN Sait 2, AnneMarie W Block 2, Maurice Barcos 3, Carlos E Vigil 1, Elizabeth A Griffiths 1, James E Thompson 1, Eunice S Wang 1, Julian Ambrus Jr 4, Meir Wetzler 1
PMCID: PMC3699612  NIHMSID: NIHMS467924  PMID: 23692654

Abstract

Our objective was to recognize the association of autoimmune disease (AD) in patients with myelodysplastic syndromes (MDS) and understand how this association could affect prognosis and management of both diseases. We describe our cohort of 10 patients and 34 patients reported in the English literature in addition to ten cohort studies. Interestingly, four cases showed improvement in AD after 5-azacitidine treatment. The mechanism(s) of the association between AD and MDS are discussed. Treatment could be targeted against AD, MDS or both, though based on recent reports, treating MDS with hypomethylating agents alone could improve the associated AD.

Keywords: myelodysplastic syndrome, autoimmune diseases, outcome, hypomethylating agents, treatment

Introduction

Approximately 10–20% of patients with myelodysplastic syndrome (MDS) present with autoimmune diseases (AD) which can be challenging to recognize. The autoimmunity is believed to be triggered by the increased apoptosis in the dysplastic bone marrow. Recent evidence suggests that both diseases are characterized by dendritic (1) and T-cell (2) abnormalities. AD presentation varies from clinical syndromes such as vasculitis, lupus and rheumatoid arthritis to laboratory abnormalities such as thrombocytopenia, hemolytic anemia and autoantibodies (3). We encountered ten cases of adults with MDS and AD at Roswell Park Cancer Institute (RPCI) from 2007 to 2010 out of 123 (8.1%) cases diagnosed during the same time period. To gain more insight into the association of MDS and AD, we also examined the English literature on all MDS and AD cases between the years 1991 to 2011 with reported clinical features.

Patients and Methods

Data on ten MDS patients with AD diagnosed at RPCI, Buffalo were reviewed on IRB-approved institute protocol. We attempted to compare these patients with a similar group of patients with MDS without AD but could not identify young female patients with MDS without AD. In addition, 34 MDS and AD cases described in the literature were also reviewed. Cases with incomplete clinical features were excluded. We recorded patients' age, gender, whether the presentation was synchronous or metachronous, MDS pathology, karyotype, AD characteristics, erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), anti-mitochondrial antibody (AMA), antinuclear antibody (ANA), complement (C3) 3, complement 4 (C4), anti-double strand DNA antibodies (anti-DsDNA), rheumatoid factor (RF), cold agglutinins, treatment of MDS/AD, outcome of MDS/AD and overall survival. Finally, we performed a Medline search and identified ten large series describing MDS and AD patients.

Results

Patient Characteristics

As shown in Tables 1 and 2, ten large series (312) encompassing a total of 2,466 patients demonstrated that 15% (range, 7%–25%) of MDS patients had AD. Five of the ten cohorts listed the laboratory characteristics of patients with both MDS and AD (Table 3). The median age of the patients was 62 years and 60% of the patients were males.

Table 1.

Clinical presentations of AD associated with MDS from ten series

Presentation Ref# 7 Ref# 8 Ref# 11 Ref# 5 Ref# 6 Ref# 4 Ref# 3 Ref# 10 Ref# 9 Ref# 12 Median (%)
AD (% of MDS) 20/284 (7) 16/162 (10) 8/82 (10) 16/153 (10) 10/83 (12) 30/221 (14) 13/70 (19) 16/80 (20) 21/84 (25) 574/2471 (23) 15
Vasculitis (% of AD) 5/20 (25) 7/16 (43) 2/8 (25) - 6/10 (60) 20/30 (66) 7/13 (53) 8/16 (50) 6/21 (28) 21/574 (4) 40
Seronegative arthritis (% of AD) - 7/16 (43) 5/8 (62) - - 1/30 (3) - - - 5/574(1) 27
Neuro (% of AD) - 8/16 (50) - - - 7/30 (23) 3/13 (23) - - 5/574(1) 24
BOOP and IPF (% of AD) - - - - - 9/30 (30) 2/13 (15) 1/16 (6) - - 15
GN (% of AD) - 2/16 (12) - - - 5/30 (16) 1/13 (7) - - - 12
RA (% of AD) 4/20 (20) 1/16 (6) 2/8 (25) - - 1/30 (3) - 1/16 (6) - 150/574 (26) 12
Sweet's (% of AD) 2/20 (10) - - 5/16 (31) 1/10 (10) - - 2/16 (12) 2/21 (10) - 10

Only AD manifestations >5% are depicted in the table

Data are not shown for: SLE and Raynaud's (4.6% each of all AD); RP, Sjogren's, photosensitivity, myositis, PMR, Panniculitis (4% each of all AD); IBD and PBC combined 3.3%; Thyroditis 2.6%; Vitiligo and Erythema Nodosum (1.3% each of all AD). Anderson had 55/574 MDS with PMR (not shown).

Abbreviations: AD, Autoimmune disease; BOOP, Bronchiolitis obliterans organizing pneumonia; GN, Glomerulonephritis; IBD, Inflammatory bowel disease; IPF, Idiopathic pulmonary fibrosis; MDS, Myelodysplastic syndrome; PBC, Primary biliary cirrhosis; PMR, Polymyalgia rheumatic; RA, Rheumatoid arthritis; RP, Relapsing polychondritis; SLE, Systemic lupus erythematous

Table 2.

MDS characteristics in patients with MDS and AD from six series

Presentation Ref# 8 Ref# 5 Ref# 6 Ref# 4 Ref# 3 Ref# 9 Median (%)
AD (% of MDS) 16/162(10) 16/153(10) 10/83(12) 30/221(14) 13/70(19) 21/84 (25) 12
Median age (years) 67 48 66 63 67 NA 62
Male (%) 12/16(75) 4/16(50) 7/10(70) 16/30(53) 7/13(53) NA 60
RA (%) 3/16(18) 9/16(56) 3/10(33) 15/30(50) 6/13(46) 5/21(24) 39
RARS (%) 2/16(12) 0 0 3/30(10) 1/13(7) 3/21(14) 7
RAEB (%) 2/16(12) 2/16(12) 3/10(33) 7/30(23) 2/13 (14) 4/21 (19) 18
RAEB-t (%) 1/16(6) 5/16(31) 1/10(10) 2/30(6) 2/30(6) 5/21(24) 15
CMML (%) 1/16 (6) 0 0 1/30 (3) 2/30(6) 4/21(19) 7
Cytogenetics* Normal Normal Complex Del(5q) NA NA
*

Only most common cytogenetic finding is listed.

Abbreviations: AD, autoimmune diseases; CMML, chronic myelomonocytic leukemia; MDS, myelodysplastic syndrome; NA, not available; RA, refractory anemia; RAEB, refractory anemia with excess blasts; RAEB-t, refractory anemia with excess blasts in transformation; RARS, Refractory anemia with ringed sideroblasts;

Table 3.

Laboratory abnormalities in patients with associated MDS and AD from five series

Presentation Ref# 3 Ref# 9 Ref# 4 Ref# 5 Ref# 7 Median (%)
AD (% of patients) 13/70 (19) 21/84 (25) 40/221 (18) N/A N/A 19
Hypergammaglobulinemia (% of AD) 10/13 (76) Present 9/40 (22) 50/128 (39) 90/284 (31) 35
ANA (% of AD) 4/13 (30) 6/21 (28) 14/40 (35) 35/115 (30) 67/284* (23) 30
RF (% of AD) 3/13 (23) 18/21 (85) 2/40 (5) 16/115 (14) * 18.5
Hypocomplementemia (% of AD) - - 7/40 (17) - - 17
DAT (% of AD) 3/13 (23) 6/21 (28) 6/40 (15) 13/108 (12) 14/2 (5) 15
Cryoglobulins (% of AD) - - 5/40 (12) - - 12
Antiphospholipids antibodies (% of AD) - - 4/40 (10) - - 10
Hypogammaglobulinemia (% of AD) 1/13 (7) - 2/40 (5) 11/128 (8) 42/284 (14) 7.5
Anti DsDNA (% of AD) - - - 8/110 (7) - 7
AMA (% of AD) 1/13 (7) - 1/40 (2.5) - * 4.75
Antiplatelets antibodies (% of AD) 1/13 (7) - - 2/153 (1) - 4
*

Combined ANA, RF and AMA reported values

Only mean gammaglobulin value is available

Higher than weak positive result

ASMA was reported in one study in 1/40 patients (2.5%)

Abbreviations: AD, autoimmune disease; AMA, anti-mitochondrial antibodies; ANA, antinuclear antibodies; ASMA, anti-smooth muscle antibodies; DAT, direct antiglobulin test; RF, rheumatoid factor;

Ten patients from RPCI were identified to have MDS and AD between 2007 and 2010. Patients' basic characteristics, AD manifestations and outcomes were analyzed along with 34 cases from our MEDLINE search [Tables 4, 5 (8, 1337)]. As seen in Table 4, most of these patients were males (70%) and the median age at diagnosis of MDS was 59 years. The presentation of MDS and AD was synchronous in 56% of the patients. In patients with asynchronous presentations, the preceding diagnosis distributed equally whether it was MDS or AD first.

Table 4.

Patient characteristics: N=44 (10 from RPCI and 34 from the literature)

Characteristics
Age at diagnosis of MDS, median (range) 59.2 (16–82)
Age at diagnosis of AD, median (range) 58.8 (16–82)
Male gender, n (%) 31 (70)
Synchronous, n (%) 25 (56)
MDS pathology
 RA, n (%) 16 (36)
 RAEB-1 and 2, n (%) 15 (34)
 RARS, n (%) 6 (14)
 Other (MDS/MPN), n (%) 4 (9)
 CMML, n (%) 3 (6)
Karyotype
 Normal, n (%) 21 (44)
 ≥2 abnormalities, n (%) 6 (13)
 del(5q) (sole), n (%) 5 (11)
 del(20q), n (%) 4 (9)
 Trisomy 8, n (%) 3 (6)
 N/A, n (%) 9 (20)
AD Characteristics
Vasculitis, n (%) 12 (27)
Seronegative arthritis, n (%) 8 (18)
Skin lesions, (Sweet, pyoderma and ictyosis) n (%) 6 (13)
Relapsing polychondritis, n (%)
Pulmonary, n (%) 4 (9)
GI (Colitis and pancreatitis), n (%) 4 (9)
Thyroiditis, n(%)
Hemolytic anemia with DAT, n (%) 3 (7)
Othera n (%) 3 (7)
3 (7)
15 (31)
Treatment of MDS
 Hypomethylating agents, n (%) 13 (29)
 Other, n (%) 9 (20)
 None, n (%) 8 (18)
 N/A, n (%) 6 (13)
 Low-dose cytarabine, n (%) 4 (9)
 Transfusion only, n (%) 4 (9)
Treatment of AD
 Prednisone alone, n (%) 21 (47)
 Prednisone and additional drugs, n (%) 11 (25)
 Other, n (%) 6 (13)
 None, n (%) 4 (9)
 N/A, n (%) 2 (4)
Outcome of AD
 Improvement of AD, n (%)b 31 (70)
 Worsening, n (%) 8 (18)
 N/A, n (%) 5 (11)
Outcome of MDS
 Persistent, n (%) 17 (38)
 Transformation to AML, n (%) 11 (25)
 Improvement of MDS, n (%) 8 (18)
 N/A, n (%) 8 (18)
a

Only AD manifestations >5% are presented, less frequent diseases included:

Two cases of rheumatoid arthritis, ITP, nephritis, Sjogren's and myositis each.

One case of PMR, SLE, Uveitis, positive rheumatoid factor and ANA associated with athralgias each.

b

Only described in patients who received treatment for AD

Abbreviations: AD, autoimmune diseases; AML, acute myeloid leukemia; ANA, antinuclear antibodies; CMML, chronic myelomonocytic leukemia; DAT, direct antiglobulin test; del, deletion; Gl, gastrointestinal; ITP, idiopathic thrombocytopenic purpura; MDS, myelodysplastic syndromes; MPN, myeloproliferative neoplasm; n, number; N/A, not available; PMR, polymyalgia rheumatica; RA, refractory anemia; RAEB, refractory anemia with excess blasts; RARS, refractory anemia with ringed sideroblasts; SLE, systemic lupus erythematous;

Table 5.

List of the frequencies of abnormal laboratory autoimmune findings from 44 cases reported (including 10 from our institute):

Test resula Number of cases with abnormal results/Number of case tested (%)
Elevated ESR 15/17 (88%)
Elevated CRP 11/14 (78%)
DAT 5/15 (33%)
RF 4/15 (26%)
ANA 5/23 (21%)
Anti DsDNA 1/7 (14%)
a

Only presentations >5% are presented.

Less frequent presentations include:

Two cases with anti-platelets antibodies.

Abbreviations: ANA, antinuclear antibodies; Anti DsDNA, double stranded DNA antibodies; CRP, C reactive protein; DAT, direct antiglobulin test; ESR, erythrocyte sedimentation rate; RF, rheumatoid factor;

Characteristics of AD

The most prevalent manifestations in the ten series (Table 1) were vasculitis syndromes (40%) followed by seronegative arthritis (27%) and neuropathy (24%). Vasculitis varied in presentation from small vessel disease such as leucocytoclastic vasculitis, skin vasculitis and microscopic polyangiitis to medium vessel diseases such as anti-neutrophil cytoplasmic antibody (ANCA)-positive vasculitis and finally large vessel disease such as temporal arteritis and Takayasu's vasculitis. Rare cases of isolated CNS vasculitis were also described. The most prevalent manifestations among the 44 cases were vasculitis followed by seronegative arthritis and skin lesions (Table 4).

As shown in Table 3, the most prevalent laboratory abnormalities, in the five of ten large series with reported laboratory results, were hypergammaglobulinemia (35%) and ANA positivity (30%). The most common laboratory abnormalities associated with AD in the 44 cases were elevated ESR (88%) and CRP (78%) (Table 5). The difference in the laboratory abnormalities between the five of ten series with reported laboratory results and the case reports along with our series (44 patients) may reside in the fact that not all studies addressed the same spectrum of laboratory abnormalities.

Characteristics of MDS

As shown in Table 2, refractory anemia was the most common type of MDS (39%) in the ten large series and also among the 44 case series (36%). Four out of the cohort studies documented the karyotype for a total of 56 patients with both MDS and AD; 41% of the patients had normal karyotype followed by chromosome 7 abnormalities in 16% of the patients. Trisomy 8 (38) and del(5q) (10%) were less common (data not shown). Among the 44 cases reviewed, the most common cytogenetic presentation was normal diploid karyotype in 44% followed by having two or more abnormalities (13%). As a sole deletion, del(5q) was more common (11%) in comparison to del(20q) (9%).

It was difficult to draw a conclusion on International Prognostic Scoring System (IPSS) (39) from the available data since some of the series predate the IPSS; Dalamaga et al (9) reported six patients with low risk IPSS and 15 patients with high risk IPSS of 21 patients with MDS and AD that were reported in a cohort of 84 MDS patients. Concerning treatments, the patients received an assortment of modalities, all without hypomethylating agents, and therefore specific conclusions about the treatments could not be drawn.

Finally, transformation to acute myeloid leukemia occurred in 25% of the patients in both the large series and the 44 cases, which is not significantly different than the frequency of disease transformation in MDS without AD (40) in the pre-hypomethylating agents era.

Prognosis

Data on the prognosis of MDS patients who develop AD is scarce. Enright et al (4) showed that the median survival for patients with MDS (from MDS diagnosis) was 25 (range 3.5–142) months while the median survival for patients with MDS and AD was only 9 (range 1–143) months. In some of the patients in that series, a rapid deterioration occurred with the onset of AD. The median survival in MDS patients from diagnosis of AD was as short as 6 (1–143) months. Interestingly, those with chronic or isolated AD lived longer [23 (range 3–124) months]. The causes of death ranged from worsening pulmonary infiltrates to intracranial hemorrhage due to flare of vasculitis, gastrointestinal hemorrhage and sepsis. Okamoto et al (5) also showed that the outcome of MDS patients with AD was significantly worse than the outcome of MDS patients without AD, though the cause of death in this series was infection or leukemic progression. Similarly, Dalamaga et al (9) showed that MDS with AD was about 3.59 times more likely to be in the high-risk MDS category than in the low-risk range. Finally, Castro et al (8) also showed a worse outcome for MDS with AD with three of 16 patients dying after one year of follow-up. However, Marisavljević et al (7) showed similar incidence of low-risk MDS among patients with and without AD. Of note, the study (7) focused more on autoimmune serologic abnormalities rather than clinical presentations of AD in patients with MDS.

Discussion

AD occurring in the setting of MDS is important to recognize not only for the purpose of knowledge per se but also in regard to prognosis and treatment options. Therefore, our discussion concentrates on these latter issues.

Since most of these data precede the use of hypomethylating agents in MDS treatment, and observations from others (41) and our own (42) suggest that treating MDS with a hypomethylating agent may improve the outcome of AD, it is important to see whether AD will continue to be a negative prognosticator in MDS in the hypomethylating era. The role of hypomethylating agents as immunomodulators is still poorly understood. It could be explained by the effect noted on T-cells activation and proliferation, decrease in proinflammatory cytokines, and the up-regulation in genes such as p53 that lead to cell-growth arrest. The immunomodulatory concept was shown by using 5-azacitidine to control a graft-versus-host disease in a fully mismatched allogeneic transplant mouse model (43).

On the other hand, when examining the effect of immunomodulation on the outcome of AD, we noted that two patients from RPCI were treated with lenalidomide for their MDS, they both had bronchiolitis obliterans organizing pneumonia; one patient's pulmonary symptoms continued to deteriorate and the other had stable mild symptoms with the use of corticosteroids. It is difficult to draw a conclusion about lenalidomide from this limited number of patients. While improvement of AD symptoms has been observed in MDS patients post allogeneic stem cell transplantation (two patients from RPCI) or umbilical cord transplant (17); other reports suggested that lenalidomide is a possible cause for lymphocytic/eosinophilic immune myocarditis, hypersensitivity pneumonitis or autoimmune hemolytic anemia (4447) which should raise the possibility of exacerbating autoimmunity when lenalidomide is used in MDS.

The effect of immunosuppression on MDS was studied by Enright et al (4) where he identified 30 patients with AD in a cohort of 221 MDS patients. As most of the AD responded to immunosuppression (27 patients had corticosteroids, two sulfasalazine, one azathioprine, one cyclophosphamide and one with both azathioprine and cyclophosphamide). The authors noted improvement of peripheral cytopenia in ulcerative colitis patients that were responding to sulfasalazine. They also noted improved cytopenia in six patients treated with immunosuppression without using any MDS-specific therapy; and in the case of one patient with refractory anemia, cytogenetic clonal abnormalities disappeared with immunosuppressive therapy and the patient stayed alive after 13 years of follow up. The survival in the group of patients with hematologic improvement (n=6) to immunosuppression was better when compared to non-responders (59 vs. 9 months, P=0.02). Interestingly, Giannouli et al(3) did not find a similar association in a similarly designed study.

A crucial point in understanding the effect of AD on MDS outcome and for planning an appropriate treatment would be improved insights into the underlying pathophysiology of AD in the context of MDS. Kiladjian et al (2) have suggested that ineffective hematopoiesis in MDS causes increased apoptosis of dysplastic precursor cells in the bone marrow exposing tumor neo-antigen(s) (still unknown) that evoke an adaptive immune response. Support for this hypothesis comes from the fact that activated T-cells with clonal expansion ability were found in the majority of MDS patients studied. Although the exact significance of these T-cells remains unclear, differences in their percentages and their expansion capabilities between patients with MDS and AD and those with MDS without AD were detected. These authors demonstrated that the percentage of Gamma-Delta T-cells [which are involved in immune reactions against tumor cells by producing tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ)] were significantly lower in MDS patients with AD compared to either healthy donors or MDS patients without AD and were in the same low range as in patients with AD without MDS. Furthermore, they demonstrated that interleukin (IL)-2 and bromohalohydrin pyrophosphate, which stimulate expansion in 100% of T-cells, were only able to expand Gamma-Delta T-cells in 60% of MDS patients and 70% of the non-responding patients had AD, suggesting that the lack of response is related to the presence of AD (2).

Another cell population involved in MDS etiology is the regulatory T-cells (T-Regs, CD4+/CD25high/FOXP3+) (48). T-Regs are usually involved in maintaining immune tolerance and their expansion, in malignant disease, often results in suppression of host anti-tumor responses. The role of T-Regs in various autoimmune diseases is still unclear. While some authors such as Kordasti et al (48) demonstrated that MDS patients with a high IPSS had higher T-Reg percentages which allowed clonal expansion and disease progression when compared to MDS patients with low IPSS; others (49, 50) observed a higher percentage of functional T-Regs in patients with AD. We (42) therefore analyzed the percentages of T-Regs in a patient with MDS and AD before and after 5-azacitidine treatment. Our results demonstrated a decrease in TRegs following nine cycles of 5-azacitidine treatment along with clinical improvement in both MDS and AD. The key seems to be related to understanding the various immune effects of hypomethylating agents on this subset of cells. In both in-vivo and in-vitro studies, these agents demethylate the FOXP3 promoter thus increasing the number of T-Regs which could be beneficial in low risk MDS where T-Regs control seems to be lacking in addition to decrease in IFNγ production. On the other hand, the same agent could enhance in-vitro IFN-γ production by demethylation IFN-γ promoter (51). These discrepancies also support that there is more to define when studying T-Regs physiology in MDS and autoimmunity. Finally, concomitant treatment with corticosteroids could play a role in affecting the number of T-Regs in these cases.

Also, it has been shown that monocyte-derived dendritic cells (MoDCs) from MDS patients are numerically and functionally defective (1, 52). DCs are considered one of the most potent antigen presenting cells (APC), They usually differentiate from circulating monocytes under “danger” mediators' effect such as lipopolysaccharadies, TNF-α or other cytokines. In-vitro, DCs are being generated from peripheral blood monocytes by culturing with IL-4 and granulocyte-macrophage colony-stimulating factor. When compared with healthy donor samples, MDS patients' MoDC had impaired/low differentiation and low endocytic capacity. They were also suggested to be derived from the same MDS clone (95% of DCs from 2 patients with 5q deletion had the same clonal abnormalities). For all that, it is believed MoDCs in these patients are ineffective in clearing the increased apoptotic bodies in the dysplastic bone marrow, which could attribute to triggering autoimmune manifestations.

Finally, interferon regulatory factor-1 (IRF-1) was previously suggested to be a transcription activator for IFN-β and to activate parts of the immune response (IL-2 and Th1 differentiation); when truncated IRF-1 becomes non-functional and involved in myelodysplasia (53, 54). Its loss was shown to cause leukemogenesis (55). Giannouli et al (56) have studied its role as a possible link between autoimmunity and myelodysplasia by comparing IRF-1 full length mRNA (IRF-1 FL) (as a surrogate for alternative splicing that gives rise into nonfunctional IRF-1) in samples from MDS and AD, MDS without AD, vasculitis and normal control patients. They confirmed the low expression of IRF-1 FL in MDS samples; those with MDS and AD had high expression of IRF-1 FL similar to those with vasculitis alone (without MDS). With those findings it was concluded that inhibiting IRF-1 could play a therapeutic role in controlling autoimmunity in MDS patients.

In summary, though the exact mechanism(s) underlying MDS and AD are not well established, it is important to look for early signs of autoimmunity in MDS patients. Treatment options include treating MDS only as suggested by others (41) and our observation (42) of treating both diseases concomitantly with 5-azacitidine.

Acknowledgments

Funding: Supported partially by grants from the National Cancer Institute Grant CA16056 (OAU, LAF, SNJS, AWB, MB, CEV, JET, ESW, MW), the Szefel Foundation (ESW), the Leonard S. LuVullo Endowment for Leukemia Research (MW), the Nancy C. Cully Endowment for Leukemia Research (MW), the Babcock Family Endowment (MW) and the Heidi Leukemia Research Fund, Buffalo, NY (MW).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Authors' contribution: O.A.U. reviewed the literature and all the cases and wrote the manuscript; L.A.F constructed the database; S.J.N.S. and A.W.B. reviewed all the karyotype analyses; M.B. reviewed the pathology specimens; C.E.V, E.A.G., J.E.T. and E.S.W. contributed to the MDS care of the patients; J.A.Jr. contributed to the AD care of the patients; M.W. oversaw the conduct of the study, contributed to the care of the patients and to the manuscript preparation All authors approved the final version submitted.

Financial disclosures: The authors have no financial disclosures.

References

  • 1.Micheva I, Thanopoulou E, Michalopoulou S, Karakantza M, Kouraklis-Symeonidis A, Mouzaki A, et al. Defective tumor necrosis factor alpha-induced maturation of monocyte-derived dendritic cells in patients with myelodysplastic syndromes. Clin Immunol. 2004 Dec;113(3):310–7. doi: 10.1016/j.clim.2004.08.007. [DOI] [PubMed] [Google Scholar]
  • 2.Kiladjian JJ, Visentin G, Viey E, Chevret S, Eclache V, Stirnemann J, et al. Activation of cytotoxic T-cell receptor gammadelta T lymphocytes in response to specific stimulation in myelodysplastic syndromes. Haematologica. 2008 Mar;93(3):381–9. doi: 10.3324/haematol.11812. [DOI] [PubMed] [Google Scholar]
  • 3.Giannouli S, Voulgarelis M, Zintzaras E, Tzioufas AG, Moutsopoulos HM. Autoimmune phenomena in myelodysplastic syndromes: a 4-yr prospective study. Rheumatology (Oxford) 2004 May;43(5):626–32. doi: 10.1093/rheumatology/keh136. [DOI] [PubMed] [Google Scholar]
  • 4.Enright H, Jacob HS, Vercellotti G, Howe R, Belzer M, Miller W. Paraneoplastic autoimmune phenomena in patients with myelodysplastic syndromes: response to immunosuppressive therapy. British journal of haematology. 1995 Oct;91(2):403–8. doi: 10.1111/j.1365-2141.1995.tb05310.x. [DOI] [PubMed] [Google Scholar]
  • 5.Okamoto T, Okada M, Mori A, Saheki K, Takatsuka H, Wada H, et al. Correlation between immunological abnormalities and prognosis in myelodysplastic syndrome patients. International journal of hematology. 1997 Oct;66(3):345–51. doi: 10.1016/s0925-5710(97)00042-x. [DOI] [PubMed] [Google Scholar]
  • 6.Billstrom R, Johansson H, Johansson B, Mitelman F. Immune-mediated complications in patients with myelodysplastic syndromes--clinical and cytogenetic features. European journal of haematology. 1995 Jul;55(1):42–8. doi: 10.1111/j.1600-0609.1995.tb00231.x. [DOI] [PubMed] [Google Scholar]
  • 7.Marisavljevic D, Kraguljac N, Rolovic Z. Immunologic abnormalities in myelodysplastic syndromes: clinical features and characteristics of the lymphoid population. Med Oncol. 2006;23(3):385–91. doi: 10.1385/MO:23:3:385. [DOI] [PubMed] [Google Scholar]
  • 8.Castro M, Conn DL, Su WP, Garton JP. Rheumatic manifestations in myelodysplastic syndromes. The Journal of rheumatology. 1991 May;18(5):721–7. [PubMed] [Google Scholar]
  • 9.Dalamaga M, Karmaniolas K, Matekovits A, Migdalis I, Papadavid E. Cutaneous manifestations in relation to immunologic parameters in a cohort of primary myelodysplastic syndrome patients. Journal of the European Academy of Dermatology and Venereology : JEADV. 2008 May;22(5):543–8. doi: 10.1111/j.1468-3083.2007.02520.x. [DOI] [PubMed] [Google Scholar]
  • 10.Hamidou MA, Derenne S, Audrain MA, Berthelot JM, Boumalassa A, Grolleau JY. Prevalence of rheumatic manifestations and antineutrophil cytoplasmic antibodies in haematological malignancies. A prospective study. Rheumatology (Oxford) 2000 Apr;39(4):417–20. doi: 10.1093/rheumatology/39.4.417. [DOI] [PubMed] [Google Scholar]
  • 11.Chandran G, Ahern MJ, Seshadri P, Coghlan D. Rheumatic manifestations of the myelodysplastic syndromes: a comparative study. Australian and New Zealand journal of medicine. 1996 Oct;26(5):683–8. doi: 10.1111/j.1445-5994.1996.tb02940.x. [DOI] [PubMed] [Google Scholar]
  • 12.Anderson LA, Pfeiffer RM, Landgren O, Gadalla S, Berndt SI, Engels EA. Risks of myeloid malignancies in patients with autoimmune conditions. British journal of cancer. 2009 Mar 10;100(5):822–8. doi: 10.1038/sj.bjc.6604935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Funato K, Kuriyama Y, Uchida Y, Suzuki A, Miyazawa K, Ohyashiki K. Myelodysplastic syndrome accompanied by Addison's disease and multiple autoimmune phenomena: steroid therapy resolved cytopenias and all immune disorders. Intern Med. 2001 Oct;40(10):1041–4. doi: 10.2169/internalmedicine.40.1041. [DOI] [PubMed] [Google Scholar]
  • 14.Manganelli P, Delsante G, Bianchi G, Fietta P, Quaini F. Remitting seronegative symmetrical synovitis with pitting oedema in a patient with myelodysplastic syndrome and relapsing polychondritis. Clin Rheumatol. 2001;20(2):132–5. doi: 10.1007/pl00011192. [DOI] [PubMed] [Google Scholar]
  • 15.Namba Y, Koizumi H, Nakamura H, Tarumi T, Sawada K, Ohkawara A. Specific cutaneous lesions of the scalp in myelodysplastic syndrome with deletion of 20q. J Dermatol. 1999 Apr;26(4):220–4. doi: 10.1111/j.1346-8138.1999.tb03460.x. [DOI] [PubMed] [Google Scholar]
  • 16.Varkonyi J, Jakab L, Szombathy T, Vamos R, Zalatnai A, Nagy P. [Relapsing polychondritis associated with myelodysplasia and acute eosinophilic leukemia] Orv Hetil. 1997 Apr 13;138(15):945–8. [PubMed] [Google Scholar]
  • 17.Tsuji G, Maekawa S, Saigo K, Nobuhara Y, Nakamura T, Kawano S, et al. Dermatomyositis and myelodysplastic syndrome with myelofibrosis responding to methotrexate therapy. Am J Hematol. 2003 Nov;74(3):175–8. doi: 10.1002/ajh.10430. [DOI] [PubMed] [Google Scholar]
  • 18.Tomonari A, Tojo A, Takahashi T, Iseki T, Ooi J, Takahashi S, et al. Resolution of Behcet's disease after HLA-mismatched unrelated cord blood transplantation for myelodysplastic syndrome. Annals of hematology. 2004 Jul;83(7):464–6. doi: 10.1007/s00277-003-0819-6. [DOI] [PubMed] [Google Scholar]
  • 19.Tsochatzis E, Vassilopoulos D, Deutsch M, Asvesti C, Sevastos N, Archimandritis AJ. Myelodysplastic syndrome presenting as acquired ichthyosis. Eur J Intern Med. 2006 Aug;17(5):368–9. doi: 10.1016/j.ejim.2005.12.016. [DOI] [PubMed] [Google Scholar]
  • 20.Giagounidis AA, Haase S, Germing U, Heinsch M, Aul C. Autoimmune disorders in two patients with myelodysplastic syndrome and 5q deletion. Acta Haematol. 2005;113(2):146–9. doi: 10.1159/000083454. [DOI] [PubMed] [Google Scholar]
  • 21.Pollack SM, Gutierrez G, Ascensao J. Pulmonary alveolar proteinosis with myeloproliferative syndrome with myelodysplasia: bronchoalveolar lavage reduces white blood cell count. Am J Hematol. 2006 Aug;81(8):634–8. doi: 10.1002/ajh.20670. [DOI] [PubMed] [Google Scholar]
  • 22.Steurer M, Fritsche G, Tzankov A, Gotwald T, Sturm W, Konwalinka G, et al. Large-vessel arteritis and myelodysplastic syndrome: report of two cases. Eur J Haematol. 2004 Aug;73(2):128–33. doi: 10.1111/j.1600-0609.2004.00265.x. [DOI] [PubMed] [Google Scholar]
  • 23.Espinosa G, Font J, Munoz-Rodriguez FJ, Cervera R, Ingelmo M. Myelodysplastic and myeloproliferative syndromes associated with giant cell arteritis and polymyalgia rheumatica: a coincidental coexistence or a causal relationship? Clin Rheumatol. 2002 Aug;21(4):309–13. doi: 10.1007/s100670200081. [DOI] [PubMed] [Google Scholar]
  • 24.Tanvetyanon T, Stiff PJ. Recurrent steroid-responsive pancreatitis associated with myelodysplastic syndrome and transformations. Leuk Lymphoma. 2005 Jan;46(1):151–4. doi: 10.1080/10428190400011617. [DOI] [PubMed] [Google Scholar]
  • 25.Coha B, Fustar-Preradovic L, Sekelj S, Sekelj A. Total hearing loss and blindness caused by relapsing polychondritis and myelodysplastic syndrome. Eur Arch Otorhinolaryngol. 2007 Dec;264(12):1517–9. doi: 10.1007/s00405-007-0387-9. [DOI] [PubMed] [Google Scholar]
  • 26.Isoda A, Sakurai A, Ogawa Y, Miyazawa Y, Saito A, Matsumoto M, et al. Chronic inflammatory demyelinating polyneuropathy accompanied by chronic myelomonocytic leukemia: possible pathogenesis of autoimmunity in myelodysplastic syndrome. Int J Hematol. 2009 Sep;90(2):239–42. doi: 10.1007/s12185-009-0375-5. [DOI] [PubMed] [Google Scholar]
  • 27.Lopez FF, Vaidyan PB, Mega AE, Schiffman FJ. Aortitis as a manifestation of myelodysplastic syndrome. Postgrad Med J. 2001 Feb;77(904):116–8. doi: 10.1136/pmj.77.904.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Akoum R, Michel S, Wafic T, Emile B, Marwan M, Khaled H, et al. Myelodysplastic syndrome and pancytopenia responding to treatment of hyperthyroidism: peripheral blood and bone marrow analysis before and after antihormonal treatment. J Cancer Res Ther. 2007 Jan-Mar;3(1):43–6. doi: 10.4103/0973-1482.31972. [DOI] [PubMed] [Google Scholar]
  • 29.Incalzi RA, Arena V, Capelli A, Gambassi G. Isolated PACNS-like presentation of a systemic vasculitis complicating a myelodysplastic syndrome. J Intern Med. 2004 Jun;255(6):674–9. doi: 10.1111/j.1365-2796.2004.01310.x. [DOI] [PubMed] [Google Scholar]
  • 30.Mobeireek A, Al-Mohareb F, Al-Dayel F, Al-Gazlan S, Alabdely H, Khalid M. Obliterative bronchiolitis in a patient with myelodysplastic syndrome before bone marrow transplantation. Respir Med. 2007 Feb;101(2):359–62. doi: 10.1016/j.rmed.2006.04.025. [DOI] [PubMed] [Google Scholar]
  • 31.Farmakis D, Polymeropoulos E, Polonifi A, Deftereos S, Giakoumi X, Floudas H, et al. Myelodysplastic syndrome associated with multiple autoimmune disorders. Clin Rheumatol. 2005 Aug;24(4):428–30. doi: 10.1007/s10067-004-1059-4. [DOI] [PubMed] [Google Scholar]
  • 32.Della Rossa A, Tavoni A, Tognetti A, Testi C, Bombardieri S. Behcet's disease with gastrointestinal involvement associated with myelodysplasia in a patient with congenital panhypopituitarism. Clin Rheumatol. 1998;17(6):515–7. doi: 10.1007/BF01451290. [DOI] [PubMed] [Google Scholar]
  • 33.Tabata R, Tabata C, Okamoto T, Omori K, Terada M, Nagai T. Autoimmune pancreatitis associated with myelodysplastic syndrome. Int Arch Allergy Immunol. 2010;151(2):168–72. doi: 10.1159/000236007. [DOI] [PubMed] [Google Scholar]
  • 34.Cohen MJ, Shyman A, Klein M, Ben-Yehuda A, Rubinow A, Or R, et al. Large vessel (Takayasu's) arteritis in a patient with myelodysplastic syndrome: is there a common pathogenesis? Clinical lymphoma, myeloma & leukemia. 2011 Feb;11(1):60–3. doi: 10.3816/CLML.2011.n.008. [DOI] [PubMed] [Google Scholar]
  • 35.Das M, Chhabra R, Hinton SW. Cutaneous leukocytoclastic vasculitis and myelodysplastic syndrome with little or no evidence of associated autoimmune disorders-a case report and a brief review of the literature. Am J Med Sci. 2008 Oct;336(4):368–71. doi: 10.1097/MAJ.0b013e31815e8c0b. [DOI] [PubMed] [Google Scholar]
  • 36.Pinheiro RF, Silva MR, Chauffaille Mde L. The 5q- syndrome and autoimmune phenomena: report of three cases. Leuk Res. 2006 Apr;30(4):507–10. doi: 10.1016/j.leukres.2005.08.025. [DOI] [PubMed] [Google Scholar]
  • 37.Lee J, Kim H, Ahn JK, Hwang JW, Jang JH, Koh EM, et al. Ankylosing spondylitis in a patient with myelodysplastic syndrome: an association with HLA-B27 or coincidence? Rheumatol Int. 2009 Apr;29(6):689–92. doi: 10.1007/s00296-008-0735-0. [DOI] [PubMed] [Google Scholar]
  • 38.Agard C, Pottier P, Hamidou M, Papo T, Genereau T, de Faucal P, et al. [Empirical treatment of granulomatous hepatitis of unknown origin: practice investigation in the French National Society of Internal Medicine] La Revue de medecine interne / fondee par la Societe nationale francaise de medecine interne. 2006 Apr;27(4):276–84. doi: 10.1016/j.revmed.2006.01.005. [DOI] [PubMed] [Google Scholar]
  • 39.Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997 Mar 15;89(6):2079–88. [PubMed] [Google Scholar]
  • 40.Komrokji RS, Zhang L, Bennett JM. Myelodysplastic syndromes classification and risk stratification. Hematology/oncology clinics of North America. 2010 Apr;24(2):443–57. doi: 10.1016/j.hoc.2010.02.004. [DOI] [PubMed] [Google Scholar]
  • 41.Pilorge S, Doleris LM, Dreyfus F, Park S. The autoimmune manifestations associated with myelodysplastic syndrome respond to 5-azacytidine: a report on three cases. British journal of haematology. 2011 Jun;153(5):664–5. doi: 10.1111/j.1365-2141.2010.08557.x. [DOI] [PubMed] [Google Scholar]
  • 42.Al Ustwani O, Francis J, Wallace PK, Ambrus J, Jr., Wetzler M. Treating myelodysplastic syndrome improves an accompanying autoimmune disease along with a reduction in regulatory T-cells. Leuk Res. 2011 May;35(5):e35–6. doi: 10.1016/j.leukres.2010.12.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sanchez-Abarca LI, Gutierrez-Cosio S, Santamaria C, Caballero-Velazquez T, Blanco B, Herrero-Sanchez C, et al. Immunomodulatory effect of 5-azacytidine (5-azaC): potential role in the transplantation setting. Blood. 2010 Jan 7;115(1):107–21. doi: 10.1182/blood-2009-03-210393. [DOI] [PubMed] [Google Scholar]
  • 44.Carver JR, Nasta S, Chong EA, Stonecypher M, Wheeler JE, Ahmadi T, et al. Myocarditis during lenalidomide therapy. The Annals of pharmacotherapy. 2010 Nov;44(11):1840–3. doi: 10.1345/aph.1P044. [DOI] [PubMed] [Google Scholar]
  • 45.Thornburg A, Abonour R, Smith P, Knox K, Twigg HL., 3rd Hypersensitivity pneumonitis-like syndrome associated with the use of lenalidomide. Chest. 2007 May;131(5):1572–4. doi: 10.1378/chest.06-1734. [DOI] [PubMed] [Google Scholar]
  • 46.Darabi K, Kantamnei S, Wiernik PH. Lenalidomide-induced warm autoimmune hemolytic anemia. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2006 Dec 10;24(35):e59. doi: 10.1200/JCO.2006.08.4053. [DOI] [PubMed] [Google Scholar]
  • 47.Brauer DL, Edelman B, Rapoport AP, Hess JR, Akpek G. Plasma exchange and rituximab treatment for lenalidomide-associated cold agglutinin disease. Transfusion. 2012 Nov;52(11):2432–5. doi: 10.1111/j.1537-2995.2012.03608.x. [DOI] [PubMed] [Google Scholar]
  • 48.Kordasti SY, Ingram W, Hayden J, Darling D, Barber L, Afzali B, et al. CD4+CD25high Foxp3+ regulatory T cells in myelodysplastic syndrome (MDS) Blood. 2007 Aug 1;110(3):847–50. doi: 10.1182/blood-2007-01-067546. [DOI] [PubMed] [Google Scholar]
  • 49.Cao D, Malmstrom V, Baecher-Allan C, Hafler D, Klareskog L, Trollmo C. Isolation and functional characterization of regulatory CD25brightCD4+ T cells from the target organ of patients with rheumatoid arthritis. European journal of immunology. 2003 Jan;33(1):215–23. doi: 10.1002/immu.200390024. [DOI] [PubMed] [Google Scholar]
  • 50.Azab NA, Bassyouni IH, Emad Y, Abd El-Wahab GA, Hamdy G, Mashahit MA. CD4+CD25+ regulatory T cells (TREG) in systemic lupus erythematosus (SLE) patients: the possible influence of treatment with corticosteroids. Clin Immunol. 2008 May;127(2):151–7. doi: 10.1016/j.clim.2007.12.010. [DOI] [PubMed] [Google Scholar]
  • 51.Aggarwal S, van de Loosdrecht AA, Alhan C, Ossenkoppele GJ, Westers TM, Bontkes HJ. Role of immune responses in the pathogenesis of low-risk MDS and high-risk MDS: implications for immunotherapy. British journal of haematology. 2011 Jun;153(5):568–81. doi: 10.1111/j.1365-2141.2011.08683.x. [DOI] [PubMed] [Google Scholar]
  • 52.Ma L, Ceuppens J, Kasran A, Delforge M, Boogaerts M, Vandenberghe P. Immature and mature monocyte-derived dendritic cells in myelodysplastic syndromes of subtypes refractory anemia or refractory anemia with ringed sideroblasts display an altered cytokine profile. Leuk Res. 2007 Oct;31(10):1373–82. doi: 10.1016/j.leukres.2006.11.007. [DOI] [PubMed] [Google Scholar]
  • 53.Harada H, Kondo T, Ogawa S, Tamura T, Kitagawa M, Tanaka N, et al. Accelerated exon skipping of IRF-1 mRNA in human myelodysplasia/leukemia; a possible mechanism of tumor suppressor inactivation. Oncogene. 1994 Nov;9(11):3313–20. [PubMed] [Google Scholar]
  • 54.Salkowski CA, Kopydlowski K, Blanco J, Cody MJ, McNally R, Vogel SN. IL-12 is dysregulated in macrophages from IRF-1 and IRF-2 knockout mice. J Immunol. 1999 Aug 1;163(3):1529–36. [PubMed] [Google Scholar]
  • 55.Liebermann DA, Hoffman B. Myeloid differentiation (MyD)/growth arrest DNA damage (GADD) genes in tumor suppression, immunity and inflammation. Leukemia : official journal of the Leukemia Society of America, Leukemia Research Fund, UK. 2002 Apr;16(4):527–41. doi: 10.1038/sj.leu.2402477. [DOI] [PubMed] [Google Scholar]
  • 56.Giannouli S, Tzoanopoulos D, Ritis K, Kartalis G, Moutsopoulos HM, Voulgarelis M. Autoimmune manifestations in human myelodysplasia: a positive correlation with interferon regulatory factor-1 (IRF-1) expression. Annals of the rheumatic diseases. 2004 May;63(5):578–82. doi: 10.1136/ard.2003.012948. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES