scholarly journals Anti Thymocyte Globulin-Based Treatment for Acquired Bone Marrow Failure in Adults

Cells ◽  
2021 ◽  
Vol 10 (11) ◽  
pp. 2905
Author(s):  
Jennifer M.-L. Tjon ◽  
Saskia M. C. Langemeijer ◽  
Constantijn J. M. Halkes

Idiopathic acquired aplastic anemia can be successfully treated with Anti Thymocyte Globulin (ATG)-based immune suppressive therapy and is therefore considered a T cell-mediated auto immune disease. Based on this finding, several other forms of idiopathic acquired bone marrow failure are treated with ATG as well. For this review, we extensively searched the present literature for evidence that ATG can lead to enduring remissions in different forms of acquired multi- or single-lineage bone marrow failure. We conclude that ATG-based therapy can lead to an enduring hematopoietic response and increased overall survival (OS) in patients with acquired aplastic aplasia. In patients with hypocellular myelodysplastic syndrome, ATG can lead to a hematological improvement without changing the OS. ATG seems less effective in acquired single-lineage failure diseases like Pure Red Cell Aplasia, Amegakaryocytic Thrombocytopenia and Pure White Cell Aplasia, suggesting a different pathogenesis in these bone marrow failure states compared to aplastic anemia. T cell depletion is hypothesized to play an important role in the beneficial effect of ATG but, as ATG is a mixture of polyclonal antibodies binding to different antigens, other anti-inflammatory or immunomodulatory effects could play a role as well.

Blood ◽  
2013 ◽  
Vol 122 (14) ◽  
pp. 2453-2459 ◽  
Author(s):  
Andres Jerez ◽  
Michael J. Clemente ◽  
Hideki Makishima ◽  
Hanna Rajala ◽  
Ines Gómez-Seguí ◽  
...  

Key PointsSTAT3+ T cells are found not only in detected concomitant LGL-BMFs, but in cases in which an LGL expansion was not suspected. Transformation via acquisition of a somatic mutation in T cells may be a mechanism of immune, mainly hypoplastic, bone marrow failure.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 1141-1141
Author(s):  
Elena E. Solomou ◽  
Valeria Visconte ◽  
Federica Gibellini ◽  
Neal S. Young

Abstract Ligation of the signaling lymphocyte activation molecule (SLAM), a member of the immunoglobulin superfamily expressed in T and B cells, results in T cell activation and Th1 cytokine production. SAP is a small cytoplasmic protein expressed in T- and NK cells that controls the activation signals mediated by SLAM. On T cell activation, SAP binds to Fyn kinase; Fyn is activated and phosphorylates tyrosine residues on SLAM; phosphorylation results in the formation of a complex that selectively down-regulates co-stimulatory signals in activated T cells, resulting in inhibition of IFN-γ production. Thus SAP acts as a natural suppressor of SLAM-mediated T cell activation, and, in the absence of SAP, T cells are constitutively activated and overproduce IFN-γ. Mutations in the SAP gene lead to abnormal T cell activation and enhanced Th1 cytokine production in mouse models and in humans: about half of patients with X-linked lympoproliferative disease (XLP) have functionally disabling SAP mutations. Acquired aplastic anemia (AA) is a bone marrow failure syndrome in which hematopoietic cell destruction is effected by cytotoxic T cells and type 1 cytokines. We have recently shown that T cells from patients with AA have increased protein levels of T-bet, resulting in IFN-γ overproduction (Solomou EE et al, Blood2006; 107:3983). IFN-γ inhibits hematopoietic stem cell proliferation and induces Fas-mediated apoptosis; stem cell depletion results in marrow hypoplasia and peripheral blood pancytopenia. We examined SAP expression as an explanation for aberrant T cell activation and extreme Th1 polarization. SAP protein expression on immunoblot was very low to absent in unstimulated T cells from 16 of 20 AA patients examined, as compared to normal levels of expression in equivalent numbers of healthy donors (p<0.001). No significant differences were detected in Fyn and SLAM protein levels between AA and controls. SAP mRNA levels were also significantly decreased in T cells from those AA patients with low SAP protein levels, as determined by RT-PCR. Peripheral blood DNA samples from 18 patients with AA were analyzed for SAP mutations: three novel intronic mutations, not present in controls, were identified among 7 unrelated patients: one mutation was in the promoter region of SAP (position 106, C to T; 3 patients), and two mutations in the intron-exon junction between exons 1 and 2 (position 38975, C toT; 3 patients) and 3 and 4 (position 62771, C to A; 1 patient). IFN-γ, as measured by ELISA, in three patients with undetectable SAP protein levels was significantly increased compared to healthy controls (n=5, p<0.001). Increased IFN-γ levels and Th1 polarization in AA can in part be explained by functional SAP deficiency. SAP-deficient T cells in AA would be unable to block co-stimulatory signals, leading to an activated T cell phenotype and ultimately hematopoietic cell destruction and bone marrow failure. The SAP-deficient phenotype in T cells from patients with aplastic anemia may be secondary to subtle genetic alteration in the gene’s regulation (abnormal promoter binding sites or epigenetic modulation due to mutations in introns) or as yet unidentified aberrant upstream pathways (Ets-1 and Ets-2, the transcription factors that regulate SAP expression).


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 180-180
Author(s):  
Gloria T Shan ◽  
Ivy Tran ◽  
Ashley R Sandy ◽  
Ann Friedman ◽  
Yi Zhang ◽  
...  

Abstract Abstract 180 Aplastic anemia is a severe bone marrow disorder characterized by the loss of hematopoietic stem cells (HSC). HSC destruction is thought to be T cell-mediated in a majority of patients with aplastic anemia. Global immunosuppression and HSC transplantation can induce disease remission, but these treatments are not effective in all patients and can promote life-threatening complications. Thus, novel immunomodulatory approaches are needed in this disorder. Notch is a conserved cell-cell communication pathway that can regulate T cell differentiation and function with context-dependent effects. To study the role of Notch signaling in pathogenic T cells causing immune-mediated bone marrow failure, we inhibited canonical Notch signaling in mature T cells through conditional expression of the pan-Notch inhibitor DNMAML (ROSA-DNMAMLf × Cd4-Cre mice). We used two complementary mouse models of immune-mediated bone marrow failure that mimic features of aplastic anemia: administration of C57BL/6 (B6) T cells into sublethally irradiated (500 rads) minor histocompatibility antigen mismatched BALB/b recipients (Chen et al., J Immunol 2007; 178:4159), or infusion of B6 lymphocytes into unirradiated MHC-mismatched B6×DBA F1 recipients. In contrast to control B6 T cells which led to lethal bone marrow failure in virtually all recipients, DNMAML-expressing Notch-deprived T cells were profoundly deficient at inducing HSC loss in both disease models, leading to markedly improved long-term survival (>90%). Notch-deficient T cells showed a modest decrease in overall expansion within secondary lymphoid organs, but their accumulation in the target bone marrow was preserved. Upon restimulation with anti-CD3 and anti-CD28 antibodies, DNMAML T cells had decreased production of IL-2 and interferon gamma. Activated CD4+ and CD8+ DNMAML T cells had reduced interferon gamma, granzyme B, and perforin transcripts despite preserved induction of the master transcription factors Tb×21 (encoding T-bet) and Eomes. In vivo infusion of CFSE-labeled host-type target cells revealed a decreased cytotoxicity in DNMAML as compared to control B6 T cell recipients. These observations point to a novel spectrum and mechanism of Notch action in mature T cells. Since we have shown recently that canonical Notch signaling is dispensable for the maintenance of adult HSCs (Maillard et al., Cell Stem Cell 2008, 2:356), our findings suggest that Notch inhibition could represent a novel therapeutic modality to target the T cell response and reverse immune-mediated HSC destruction in aplastic anemia. Disclosures: Shan: American Society of Hematology: Research Funding. Zhang:University of Michigan Comprehensive Cancer Center: Research Funding; Damon Runyon Cancer Research Foundation: Research Funding. Maillard:Damon Runyon Cancer Research Foundation: Research Funding; American Society of Hematology: Research Funding; University of Michigan Comprehensive Cancer Center: Research Funding.


2017 ◽  
Vol 9 (2) ◽  
pp. 127-128
Author(s):  
Anuja Nanda ◽  
Vineeta Gupta ◽  
Prachi Maheshwari ◽  
Archna Tandon

ABSTRACT Aplastic anemia is a rare disease caused by destruction of pluripotent stem cells in bone marrow. The etiology may be radiation exposure, chemotherapy, environmental toxins or it could be autoimmune. During pregnancy it can be life threatening for both mother and child. Treatment options are erythrocytes and platelet transfusions and immune suppressive therapy. We report a series of obstetric events in the life of a woman whose pregnancy was complicated due to aplastic anemia but her subsequent obstetric outcome improved after successful bone marrow transplantation. How to cite this article Gupta V, Maheshwari P, Tandon A, Nanda A. Obstetric Outcome in a Patient with Aplastic Anemia. J South Asian Feder Obst Gynae 2017;9(2):121-122.


2020 ◽  
Vol 13 (1) ◽  
pp. 76-78 ◽  
Author(s):  
Marina Vitorino ◽  
Filipa Nunes ◽  
Mariana Costa ◽  
Beatriz Porteiro ◽  
Alexys Reis Borges ◽  
...  

Pure red cell aplasia (PRCA) is a rare bone marrow failure characterized by a progressive normocytic anemia and reticulocytopenia without leukopenia and thrombocytopenia. It can be associated with various hematological disorders but exceedingly rarely with angioimmunoblastic T-cell lymphoma (AITL). We report the case of a 72-year-old woman with PRCA associated with AITL. The patient presented with severe anemia (hemoglobin 2.6 g/dL) and a low reticulocyte count 0.7%. Direct and indirect Coombs tests were positive. A CT scan of the chest, abdomen, and pelvis revealed multiple lymphadenopathies. A cervical lymph node biopsy was compatible with AITL. A bone marrow biopsy showed medullary involvement by AITL and a severe erythroid hypoplasia with a myeloid:erythroid ratio of 19.70. The patient was started on CHOP and after 6 cycles the PET scan confirmed complete remission.


2021 ◽  
Vol 4 (3) ◽  
pp. 235-238
Author(s):  
Sreethish Sasi ◽  
Mohamed A. Yassin ◽  
Arun P. Nair ◽  
Afraa M. Fadul ◽  
Mohammed A. Abukhattab

<b><i>Introduction:</i></b> Aplastic anemia (AA) and paroxysmal nocturnal hemoglobinuria (PNH) are bone marrow failure syndromes. A 20–40% of patients with AA have a PNH clone at diagnosis. To date, there are little data about the course of COVID-19 in patients with AA and PNH. <b><i>Case Presentation:</i></b> A 36-year-old gentleman, who was previously diagnosed as a case of AA with PNH clones off immune-suppressive therapy, presented with fever and cough and was diagnosed with mild pneumonia due to COVID-19 with positive nasopharyngeal swab polymerase chain reaction (PCR) for severe acute respiratory syndrome coronavirus 2. His clinical course was benign except transient thrombocytopenia. He was asymptomatic after day 4, and viral PCR was negative on day 21. <b><i>Discussion:</i></b> Though studies have shown that COVID-19 is associated with lymphopenia, our patient had a normal to high lymphocyte count. The neutrophil to lymphocyte ratio (NLR) was &#x3c;1 during COVID-19, which correlates with the mild course of the disease. To know whether elevated lymphocyte count, low NLR, and benign course of COVID-19 is a standard feature for all patients with underlying AA, we need more case reports and series. The significance of this case report is that it describes the course of COVID-19 in a patient with AA and PNH clones and, up to our knowledge, is the first report showcasing the association between these rare combinations of diseases.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 3483-3483
Author(s):  
Kazuya Sato ◽  
Xingmin Feng ◽  
Jichun Chen ◽  
Marie J. Desierto ◽  
Keyvan Keyvanfar ◽  
...  

Abstract Abstract 3483 In aplastic anemia (AA), the marrow is not “empty” but replaced by fat; the increase in adipocytes number and size is most obvious on three dimensional reconstructions of the marrow in human and murine (Takaku T, Blood. 2008). A reciprocal relationship exists between adipogenesis and osteogenesis, and osteoblasts constitute the hematopoietic niche and play an active role in the regulation of stem cells and progenitors. Fat in the marrow has been considered an epiphenomenon in AA. However, a recent report suggested that bone marrow (BM) adipocytes negatively regulated hematopoiesis in mouse models (Naveiras O, Nature. 2009). Peroxisome proliferator-activated receptor-g (PPAR-g) is a key transcription factor for adipogenesis, and blocking PPAR-g signaling inhibited adipogenesis in vitro (Wright HM, J Biol Chem. 2000). To examine the role of BM adipocytes, we investigated the effects of PPAR-g antagonists, bisphenol A diglycidyl ether (BADGE, 30 mg/kg/day) and GW9662 (1 mg/kg/day), on hematopoiesis in a mouse model of immune-mediated BM failure (Chen J, J Immunol. 2007). We induced BM failure by infusion of lymph node (LN) cells from C57BL/6 mice into sublethally irradiated C.B10-H2(b)/LilMcd (C.B10) recipient mice that were matched at major histocompatibility antigens but differed in multiple minor histocompatibility antigens. In adaptation of the “runt” disease model, mice uniformly develop progressive and fatal pancytopenia, closely resembling human BM failure, without other evidence of graft-versus-host disease. We treated recipient mice with BADGE, GW9662, or control vehicle from day -1 to day 14. On day 14, mice were sacrificed and evaluated by peripheral blood (PB) cell counting and BM cellularity, as well as morphology of marrow adipocytes. Mice in the BADGE- and GW9662-treated groups showed higher numbers of leukocytes, neutrophils, and platelets in PB and higher total nucleated cells and Lin- Sca1+ c-kit+ stem cells in BM than did animals in the control group. Both confocal microscopic imaging and hematoxylin and eosin staining of BM also showed significantly higher numbers of nucleated cells and many fewer and smaller adipocytes in the treated groups (Figure 1). We also investigated dose response of BADGE in the treatment of AA mice. Low dose of BADGE (15 mg/kg/day) had no effect while high dose of BADGE (60 mg/kg/day) seemed to have no extra benefit for the BM hematopoiesis compared with the medium dose (30 mg/kg/day). However, we also noted in PPAR-g antagonist-treated groups that there was significantly less CD8+ T cell infiltration of BM, as determined by flow cytometry. We speculated that PPAR- g antagonists might also negatively affect activation of cytotoxic T cells. By magnetic beads-based multiplex assay, we found the concentrations of inflammation-related cytokines in the plasma, including Interleukin-6, tumor necrosis factor alpha, monocyte chemotactic protein-1 were markedly decreased in PPAR- g antagonist-treated groups. When we performed PCR arrays focusing on adipogenesis and inflammasome pathways, we found that expression of adipogenesis genes was greatly decreased in the treated groups, including Agt (−149 folds), Cebpa (−4.7 folds), Acacb (−11.7 folds), Fabp4 (−3.2 folds), Adig (−14.2 folds), and Bmp2 (−12.9 folds). The expression of inflammation- or inflammasome-related genes including Nlrc4 (−11.3 folds), Mapk12 (−4.8 folds), Ptgs2 (−8.7 folds), and Rela (−5.9 folds) was also decreased while apoptosis inhibitor genes including Xiap (+17.5 folds), Mapk1 (+6.6 folds), and Bcl2l1 (+3.9 folds) were increased in the treated groups. In vitro, BADGE and GW9662 inhibited activation and proliferation of T cells stimulated with anti-CD3/CD28 or phorbol myristate acetate/ionomycin. These data suggested that BADGE and GW9662 inhibition was not specific for adipogenesis but affected T cell activation. Indeed, PPAR-g antagonists failed to ameliorate pancytopenia and BM hypoplasia in the mice exposed to either a lethal or sublethal dose of total body irradiation. PPAR-g antagonists may act to attenuate murine immune mediated marrow failure by mechanism of inhibition of T-cell activation. Figure 1. Histology of femurs from untreated bone marrow failure mice and PPAR-g antagonists treated mice. Both BADGE and GW9662 inhibited adipogenesis and increased cellularity in the bone marrow of AA mice. Figure 1. Histology of femurs from untreated bone marrow failure mice and PPAR-g antagonists treated mice. Both BADGE and GW9662 inhibited adipogenesis and increased cellularity in the bone marrow of AA mice. Disclosures: No relevant conflicts of interest to declare.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 2195-2195
Author(s):  
Nicholas C.J. Lee ◽  
Bhavisha A. Patel ◽  
Taha Bat ◽  
Ibrahim F. Ibrahim ◽  
Madhuri Vusirikala ◽  
...  

Abstract Introduction: Aplastic anemia (AA) is a life-threatening disorder characterized by pancytopenia and a hypocellular bone marrow. Pure red cell aplasia (PRCA) is a similar disorder with primary reduction in the red blood cell population and virtual absence of erythroid precursors in the bone marrow. While the etiology of immune mediated marrow failure is multifactorial, preceding viral infections have been associated with the disease; these include parvovirus B19, cytomegalovirus, and Epstein-Barr virus. We present four cases of immune mediated marrow failure with either preceding or simultaneous SARS-CoV-2 infection. Methods: The medical records of patients treated for AA or PRCA at the University of Texas Southwestern Medical Center, Parkland Hospital, and the National Institutes of Health (NIH) were reviewed for SARS-CoV-2 infection. Four patients without prior hematological diseases were identified who had SARS-CoV-2 infection prior to or with simultaneous the diagnosis of AA or PRCA. Results: Patient #1 was a 22-year-old white female who was diagnosed with asymptomatic COVID-19 10 days prior to her pancytopenia and AA diagnosis was confirmed by bone marrow biopsy (5% cellularity; Table 1). Her extensive work-up including HIV, hepatitis panel, immunoglobulins, B12 and folate was negative, and she underwent HLA-matched family donor hematopoietic stem cell transplant. Patient #2 was a 69-year-old Asian female who presented to her primary care physician with symptoms of fatigue and was found to be pancytopenic. CBC from a few months prior was completely normal. Further work-up was positive for COVID-19 and negative for HIV, nutritional deficiency, or hemolysis. She did not have respiratory symptoms, was eventually diagnosed with pRBC and platelet transfusion-dependent severe AA (5-10% cellularity on bone marrow), and underwent treatment with cyclosporine, equine antithymocyte globulin, and eltrombopag. She has had a partial response to this therapy. Both patients had bone marrow specimens stained for SARS-CoV-2 by immunohistochemistry that were negative. Patient #3 was a 76-year-old white male who was diagnosed with COVID-19 4 months prior to presenting with a non-ST segment myocardial infarction and found to be profoundly anemic, requiring pRBC transfusion. He re-presented with chest pain one week later and was found to be anemic again, and required transfusion. A trial of darbepoetin alfa was unsuccessful. Extensive work-up for malignancy, infection, and autoimmune etiologies were negative. He was diagnosed with PRCA based on the bone marrow biopsy and initiated treatment with cyclosporine. Patient # 4 was diagnosed with severe AA (presenting as pancytopenia) and COVID-19 infection. He had fatigue for one month and fever, chills and sore throat one-week prior seeking medical care. Testing for hepatitis, HIV, EBV, and CMV was negative. He was treated on a clinical trial (NCT04304820) at NIH with cyclosporine and eltrombopag until SARS-CoV-2 PCR was negative then received equine anti-thymocyte globulin. He has achieved a complete hematologic response at 6 months and remains well at last follow-up. Conclusion: The four patients described had minimal respiratory COVID-19 symptoms, but they presented with cytopenia and were eventually diagnosed with bone marrow failure. It is possible that this is co-incidental due to the high prevalence of SARS-CoV-2. However, there is emerging evidence that COVID-19 pneumonia is a hyperinflammatory and immune dysregulated state improved by dexamethasone therapy. Other immune mediated hematologic conditions, such as autoimmune hemolytic anemia and immune thrombocytopenia, have been reported. The onset from infection to cytopenia appears rapid, although patients often presented with symptoms for many days prior to diagnosis and thus testing may have been delayed from the onset of infection. This case series does not provide a mechanistic link between SARS-CoV-2 infection and bone marrow failure, but it raises the possibility that SARS-CoV-2 may mediate an immunologic response that contributes to marrow failure. Patients appear to respond well to standard immunosuppressive treatment. Further cases and studies are needed to determine if this is directly linked to SARS-CoV-2 and whether the natural history and response to standard therapy is different than idiopathic cases. Figure 1 Figure 1. Disclosures Young: Novartis: Research Funding.


Blood ◽  
2007 ◽  
Vol 110 (11) ◽  
pp. 1695-1695
Author(s):  
Lifang Huang ◽  
Min Dai ◽  
Wenli Liu ◽  
Jianfeng Zhou ◽  
Hanying Sun ◽  
...  

Abstract Aplastic anemia(AA) is an unusual hematologic disease and the paradigm of the human bone marrow failure syndromes. Abnormal immunological responses have been found in most aplastic patients. Activated type 1 cytotoxic T cells may be the main contributing factor of pathogenesis. IDO is a rate-limiting enzyme in Tryptophan(Trp) metabolism. The proliferation of Th1 cells is specifically inhibited by the over-expression of IDO, which can degrade the Trp in local environment, hamper the conductioin of the activating signal in T cell and induce immune tolerance. 3,4-DAA is an IDO agonist and can activate IDO. In our experiment, Balb/c mice were irradiated (5.0Gy of 60Co), and then infused with 5×106 lymph node(LN) cells from DBA/2 mice in 4 hours. Blood count was monitored and marrow damage was assessed by histogical study. Concentrations of serum IFN-γ were measured by ELISA. The levels of Tryptophan and kynurenine were evaluated by high performance liquid chromatography (HPLC). CD4+CD25+ T cells in spleen were analyzed by flow cytometry. The level of Foxp3 in CD4+CD25+ T cell was measured by RT-PCR. Irradiation and infusion of LN cells led to rapid development of severe pancytopenia and bone marrow hypoplasia. Bone marrow of affected mice showed lymphocyte infiltration. Serum IFN-γ concentration increased 3.7 fold at d6 post infusion. The recipient mice were divided into 4 different treating groups as follow: 0.9% Sodium Chloride as control; Cyclosporin A (CsA) (50ug/g/d ×5d, peritoneal injection); 3,4-DAA(5mg/d, orally daily); the combination of CsA and 3,4-DAA. The effects of CsA, IDO agonist and CsA combined IDO agonist were analyzed at day 6,10,14,21,24 and 28 after LN cells infusion. The white cell and the platelet recovered to near normal, respectively (4.2±0.32)×109, (937±190.47)×1012 at d21 in the combination group. Early stage treatment with CsA can improve periphery blood cells and BM nucleated cells, but long term effect was not remarkable. In contrast, the 3,4-DAA group exhibited slow and gradually enhanced role. Periphery blood cells and BM nucleated cells were improved remarkably in the combination group. The number and function of CD4+CD25+T cells also increased remarkably. In the treatment of AA, abnormal immune response in bone marrow was inhibited by CsA, meanwhile immune tolerance was induced through up-regulating the regulatory T cells(Treg) by 3,4-DAA. In this way, the balance of immune in bone marrow could be reestablished quickly. CsA combined IDO agonist could provide a new strategy for the management of AA.


Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 2479-2479 ◽  
Author(s):  
Sabrina Martyr ◽  
Arun Balakumuran ◽  
Aldemar Montero ◽  
Cynthia E. Dunbar ◽  
Elizabeth M. Kang ◽  
...  

Abstract Background Pure red cell aplasia (PRCA), Diamond-Blackfan anemia (DBA) and moderate aplastic anemia (MAA) are all bone marrow failure syndromes that are immune-mediated or may respond to immunosuppressive therapies (IST). Anti-thymocyte globulin, cyclosporine and corticosteroids have been used with some success but have significant toxicities. The humanized monoclonal antibody to the interleukin-2 receptor on T cells, daclizumab, showed efficacy in MAA and PRCA patients with some patients achieving transfusion independence (Sloand et al, Haematologica 2010). However, this agent has since been withdrawn from the market. It is increasing recognized that the anti-CD20 chimeric monoclonal antibody, rituximab, may modulate T cell immunity in addition to its known depletion of B cells (Staci, Seminars in Hematology 2010). There are anecdotal case reports of rituximab, showing benefit in PRCA. Here, we summarize our experience using rituximab in PRCA, DBA and MAA. Design and Methods We enrolled 11 patients with PRCA (n = 7), DBA (n = 1), and MAA (n = 3) who had failed at least one prior immunosuppressive regimen to receive rituximab 375 mg/m2intravenous infusions weekly times 4 doses (NCT00229619). Responses were evaluated at 3, 6 and 12 months. Patients with MAA, DBA or PRCA were eligible for trial participation. MAA was defined as a hypocellular marrow without evidence of an underlying disease process and depression of at least two of three cell lines (an absolute neutrophil count (ANC) ≤ 1200/µL, a platelet count ≤ 70,000/µL, and a hemoglobin ≤ 8.5 g/dL or absolute reticulocyte count (ARC) ≤ 60, 000/µL in transfusion-dependent patients) but who do not fulfill criteria for severe aplastic anemia (i.e. bone marrow cellularity < 30% and depression of two of the three peripheral counts: ANC < 500/µL, a platelet count < 20,000/µL and an ARC < 60,000/µL). DBA and PRCA were defined as anemia, reticulocytopenia (ARC ≤ 50, 000/µL) and absent or decreased marrow erythroid precursors. Patients with Fanconi’s anemia, other congenital bone marrow failure syndromes, cytologic abnormalities indicating myelodysplasia or recent/ongoing parvovirus infection were excluded. Complete response (CR) was defined as return of blood counts to normal. Partial response (PR) for MAA was defined as improvement in two of the three depressed blood counts that qualified patient for participation. PR for DBA/PRCA was defined as an increase in hemoglobin by 1.5 g/dl of blood and or ARC ≥ 50,000/µL but not meeting criteria for normal counts. Results Overall, 5/11 (45%) patients responded to rituximab, all achieving PR. At 3 months, one patient with PRCA had responded. At 6 months, two additional patients responded (one with PRCA, one with MAA). At 12 months, an additional two responses were confirmed (one PRCA, one MAA). One PRCA patient lost his response between the 6 and 12 month endpoint. Among the three responding PRCA patients, the mean reticulocyte count at study initiation was 4400/µL; this increased to 54,000/µL at 6 months and further increased to 61,000/µL at 12 months (including patient who lost his response). The study was terminated early for poor accrual; many eligible patients received alternate treatments at home. Due to early study termination, the duration of responses for majority of the patients is unknown. Given the reports of daclizumab efficacy in these diseases, 90% of our patients were previously treated with daclizumab. Notably, 3 of the patients responding to rituximab had previously not responded to daclizumab. Safety The most common toxicity of rituximab observed was an infusion related reaction affecting (8/11) 73% of patients with the first infusion of rituximab. One patient developed serum sickness after the third cycle which precluded the administration of the last dose. An expected decrease in quantitative immunoglobulin levels was observed; at the 6 month evaluation there was an 11% decrease in IgG and IgA; a greater decrease (48%) was observed in IgM. Conclusions Rituximab is a viable treatment option in the armamentarium for patients with PRCA and MAA. Rituximab is safe, effective, and easily administered. Responses can be delayed to beyond 6 months therefore we suggest observation for at least 6 months after rituximab administration. Disclosures: Off Label Use: Rituximab is not FDA approved for the treatment of Pure Red Cell Aplasia, Diamond-Blackfan Anemia or Moderate Aplastic Anemia.


Sign in / Sign up

Export Citation Format

Share Document