Diamond Blackfan Anemia: New Paradigms for a “Not So Pure” Inherited Red Cell Aplasia

2006 ◽  
Vol 43 (3) ◽  
pp. 167-177 ◽  
Author(s):  
Jeffrey M. Lipton
Blood ◽  
2010 ◽  
Vol 116 (21) ◽  
pp. 4430-4430
Author(s):  
Adrianna Vlachos ◽  
Jason Farrar ◽  
Eva Atsidaftos ◽  
Ellen Muir ◽  
Thomas C. Markello ◽  
...  

Abstract Abstract 4430 Background: Diamond Blackfan anemia is a rare heritable red cell aplasia which usually presents in infancy but can also be diagnosed in childhood and even adulthood. Mutations or deletions in eleven ribosomal protein (RP) genes, resulting in protein haplo-insufficiency have been reported in about 54% of the patients. The 5q- syndrome is an acquired myelodysplastic syndrome (MDS) characterized by a similar erythroid failure. Another RP gene included in the 5q deleted region, RPS14, has been identified as a causal gene in 5q- MDS but has not been reported in DBA. Purpose: Array Comparative Genomic Hybridization has been used to identify large deletions in patients with DBA. This report demonstrates the use of Single Nucleotide Polymorphism (SNP) genotyping array hybridization to identify a patient, previously thought to have DBA, as having a 5q- deletion consistent with 5q- syndrome. Method: Seventy-five patient samples from the Diamond Blackfan Anemia Registry of North America, a patient database of now 608 patients designed to better understand the biology and epidemiology of DBA, underwent resequencing of 80 RP genes. Approximately 40% of the patients had no identifiable mutation. High resolution SNP array genotyping analysis was done on 23 probands from this cohort who did not have a mutation detected in either the resequencing project and/or the targeted sequencing efforts lead by Gazda and colleagues. Result: An acquired internal deletion on chromosome 5q involving RPS14 was identified in one patient with presumed DBA. The patient presented at 5 years 10 months of age with anemia noted on a routine blood count. The hemoglobin was 8.4 grams/dl, MCV 108.2 fL, and reticulocyte count 0.4%. The eADA was normal. The bone marrow showed decreased cellularity and megaloblastic changes in the erythroid series. There were adequate numbers of megakaryocytes with no hypolobulation. The cytogenetics performed at diagnosis in 1991 were reported as normal. The patient had no significant family history of anemia and was found to have no congenital physical anomalies. A diagnosis of non-classical DBA was presumed and the patient failed a trial of corticosteroids. At present the patient has marrow red cell aplasia and is on a chronic transfusion schedule. SNP array genotyping analysis identified mosaicism in two discrete regions covering ~17.7 Mb on 5q-, with an estimated 63.7% monosomy and 36.3% disomy in this region. The major region extends from 141.1M to 157.2M (hg18), including all of the 5q- syndrome commonly deleted region (CDR) at 5q33 though it excludes the 5q31 CDR associated with AML and more aggressive MDS as well as miR146a, a factor recently postulated to play a role in 5q- MDS. SNP array genotyping from purified peripheral blood populations indicated that lymphocytes were greater than 95% normal, while the myeloid cells were greater than 95% 5q-. CD34+ cells obtained from this patient showed a marked decrease in both myeloid and erythroid colony formation when compared with normal cells. Patient fibroblasts were normal and neither of the parents have any 5q anomalies by SNP array genotyping. Although the deletion was not identified in 1991 at the time of the diagnosis, the 46,XX,der(5)del(5)(q15q22)del(5)(q32q33) deletion was able to be detected on high resolution karyotyping in a post-SNP array genotyping marrow sample. Haploinsufficiency of RPS14 was confirmed by quantitative RT-PCR. Conclusion: Patients with non-classical DBA may have unique acquired 5q deletions with RPS14 haploinsufficiency. A search for other acquired somatic mutations or deletions in patients with DBA, in particular non-classical cases, is underway. SNP array genotyping is an essential diagnostic tool in this search. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 37 ◽  
pp. S59
Author(s):  
A. Vlachos ◽  
J.E. Farrar ◽  
E. Atsidaftos ◽  
E. Muir ◽  
A. Narla ◽  
...  

Blood ◽  
2016 ◽  
Vol 128 (21) ◽  
pp. 2504-2509 ◽  
Author(s):  
Robert T. Means

Abstract Pure red cell aplasia (PRCA) is a syndrome defined by a normocytic normochromic anemia with severe reticulocytopenia and marked reduction or absence of erythroid precursors from the bone marrow. Diamond-Blackfan anemia is a congenital form of PRCA. Acquired PRCA may be either a primary disorder or secondary to some other disorder or agent. Primary acquired PRCA is an autoimmune disorder that is frequently antibody-mediated. Myelodysplastic syndromes may also present with the morphologic appearance of PRCA. Secondary acquired PRCA may be associated with collagen vascular/autoimmune disorders such as systemic lupus erythematosus; lymphoproliferative disorders such as chronic lymphocytic leukemia or large granular lymphocyte leukemia; infections, particularly B19 parvovirus; thymoma and other solid tumors; or a variety of other disorders, drugs, or toxic agents. The therapeutic approach to PRCA typically involves immunosuppression, but specific pathogenic subtypes are associated with specific therapeutic approaches. Cyclosporine A, with or without concurrent corticosteroids, appears to be the single most effective immunosuppressive agent.


Hematology ◽  
2016 ◽  
Vol 2016 (1) ◽  
pp. 51-56 ◽  
Author(s):  
Robert T. Means

Abstract Pure red cell aplasia (PRCA) is a syndrome defined by a normocytic normochromic anemia with severe reticulocytopenia and marked reduction or absence of erythroid precursors from the bone marrow. Diamond-Blackfan anemia is a congenital form of PRCA. Acquired PRCA may be either a primary disorder or secondary to some other disorder or agent. Primary acquired PRCA is an autoimmune disorder that is frequently antibody-mediated. Myelodysplastic syndromes may also present with the morphologic appearance of PRCA. Secondary acquired PRCA may be associated with collagen vascular/autoimmune disorders such as systemic lupus erythematosus; lymphoproliferative disorders such as chronic lymphocytic leukemia or large granular lymphocyte leukemia; infections, particularly B19 parvovirus; thymoma and other solid tumors; or a variety of other disorders, drugs, or toxic agents. The therapeutic approach to PRCA typically involves immunosuppression, but specific pathogenic subtypes are associated with specific therapeutic approaches. Cyclosporine A, with or without concurrent corticosteroids, appears to be the single most effective immunosuppressive agent.


Blood ◽  
2015 ◽  
Vol 126 (23) ◽  
pp. 3615-3615 ◽  
Author(s):  
Ghadir S. Sasa ◽  
M. Tarek Elghetany ◽  
Katie Bergstrom ◽  
Sarah Nicholas ◽  
Ryan Himes ◽  
...  

Abstract Diamond Blackfan anemia (DBA) is an inherited pure red cell aplasia. Most cases present in the first year of life with elevation in erythrocyte adenosine deaminase (eADA) and frequently with increased mean corpuscular volume (MCV) and hemoglobin F (hgb F). Approximately 70 percent of cases are due to a mutation in one of several ribosomal protein (RP) genes or in GATA1, whereas the remaining cases are genetically uncharacterized. Here we report a child born with severe anemia and diagnosed with DBA at 2 months of age. His bone marrow was normocellular with a paucity of erythroid progenitors and scattered lymphocytes. An eADA level was not obtained prior to the first red cell transfusion. He was red cell transfusion dependent and his anemia did not respond to a steroid trial. His 4 year old sister, who had normal hemoglobin, MCV, hgb F, and eADA measurements, served as his HLA identical donor for hematopoietic stem cell transplantation (HSCT). HSCT resulted in 100% donor chimerism, but red cell engraftment was not achieved. He subsequently underwent a mismatched unrelated HSCT with trilineage engraftment. Ten years later, at the age of 14 years, the sister presented with profound hypoproductive normocytic anemia. The bone marrow showed absence of erythroid precursors and presence of lymphoid aggregates. Findings of immunodeficiency included numerous cutaneous warts, recurrent aphthous ulcers, Epstein Barr virus (EBV) reactivation, low IgM, and low numbers and percentages of CD19+ and CD3-56+16+ lymphocytes. The anemia and reticulocytopenia persisted despite resolution of EBV reactivation. Upon her presentation, levels of iron, ferritin, transferrin saturation, and liver transaminases were elevated. A liver biopsy obtained after transfusion of a total of 60 ml/kg packed red blood cells demonstrated 4.8 mg Fe/g dry liver weight with stage 2 portal fibrosis. Targeted DNA sequencing studies performed on the affected sister were negative for single nucleotide variants in any of 12 RP genes previously implicated in DBA and a genome wide chromosome microarray was normal. Whole exome analysis of her and her parents demonstrated that she carried compound heterozygous variants in CECR1 (cat eye syndrome chromosome region, candidate 1). The variant p.R169Q had been previously reported as pathogenic, while the p.G358R variant was of uncertain significance. These variants are present at frequencies of 4.9X10-4 and 2.6X10-5 in the Exome Aggregation Consortium database, respectively. Analysis of buccal swab DNA of the proband showed the same biallelic variants. An unaffected 16-year-old sibling had a normal genotype. CECR1 encodes adenosine deaminase 2 (ADA2) and ADA2 levels in the plasma of the affected sister were markedly low, consistent with a deficiency state. CECR1 is highly expressed in cells of myeloid origin and ADA2 is a secreted protein implicated in macrophage differentiation and proliferation. Deficiency of ADA2 (DADA2) results in aberrant monocyte differentiation favoring M1 over M2 macrophages, thereby resulting in a proinflammatory state. Recent descriptions of patients with DADA2 due to CECR1 mutations reported a spectrum of phenotypes including intermittent fevers, lacunar stroke in childhood, livedoid rash, polyarteritis nodosa, and immunodeficiency with B lymphopenia and low IgM levels. Our cases are similar to the report of one of two brothers, homozygous for CECR1 p.R169Q, described by van Montfrans, et al,. (NEJM, 2014). The eldest was given a diagnosis of atypical DBA (refractory pure red cell aplasia) in infancy and underwent a HSCT from his asymptomatic, HLA identical brother. This HSCT resulted in non-engraftment, necessitating a subsequent unrelated donor HSCT. The younger sibling donor later developed hepatosplenomegaly, profound lymphopenia, and evidence of an inflammatory state. Together, these three cases support pure red cell aplasia as a presentation of DADA2 and that this may be confused with DBA, particularly when manifest in infancy. We propose DADA2 should be considered in patients with genetically uncharacterized DBA. Differentiating features to suggest DADA2 may include normal eADA, MCV, and hgb F levels and findings of associated immunodeficiency. Additionally, the macrophage activation due to DADA2 may have played a role in the iron overload observed in our second patient prior to any red cell transfusion. Disclosures No relevant conflicts of interest to declare.


Blood ◽  
2009 ◽  
Vol 114 (22) ◽  
pp. 4219-4219
Author(s):  
Peter Joosten ◽  
Mels Hoogendoorn ◽  
Huib Storm ◽  
Robby Kibbelaar

Abstract Abstract 4219 Background Diamond-Blackfan Anemia (DBA) is an autosomal dominant inherited bone marrow failure syndrome due to a defect in the ribosomal protein (RP) synthesis. Diagnostic criteria consist of presentation of anemia before birth with near normal or slightly decreased neutrophil counts, variable platelet counts, reticulocytopenia, macrocytosis, and normal marrow cellularity with a paucity of red cell precursors (Diamond et al 1976). Patient characteristics In 2002 a 34 year old man was presented with a hemoglobin (Hb) level of 2.4 mmol/l, a MCV of 117 fl, no reticulocytes and a normal leukocyte and platelet count. Except shortness of breath he had no other complaints. He did not use any medication. On physical examination there was only a short stature. Marrow cytology showed 20% erythropoesis with some dyserythropoesis. Marrow histology and cytogenetic were normal. A recent parvovirus infection was excluded. His medical history started in 1968 at the age of five weeks. A DBA was diagnosed and treated successfully with corticosteroids. There was a relapse at the age of 3 with again a good response on corticosteroids. In 2002 he was initially treated with 6 units of red blood cells, resulting in a rise of the Hb to 6.4 mmol/l. After five months he had a Hb of 3.8 mmol/l, a normal MCV and 50.109/l reticulocytes. Kidney and adrenal function were normal, there was no hypogonadism and no splenomegaly. Hb electrophoresis showed an elevated HbF of 6.4%. By exclusion of other causes of anemia, it was concluded that the anemia had to be considered as a relapse DBA. Corticosteroids (1 mg/kg) for 6 months did not show any improvement. Cyclosporine 100 mg two times a day raised the Hb above 8 mmol/l from November 2003 till May 2004. While on cyclosporine he relapsed again and became red blood cell transfusion dependent from February 2006. A search for a HLA identical donor at that time was unsuccessful. ATG, cyclosporine and corticosteroids did not diminish the need for red blood cell transfusion. In 2007 the RPS19 (ribosomal protein S19) mutation was demonstrated in this patient, which definitively confirmed the diagnosis made 35 years ago. In 2008, still red blood cell transfusion dependent, treatment with lenalidomide 10 mg/day for 21 days during each 28 days was initiated. Treatment considerations All mutated genes in DBA cause a decreased synthesis of ribosomal proteins. As a consequence erythroid progenitors and precursors are highly sensitive to apoptosis (Perdahl et al 1994). One of the mechanisms is activation of p53 (Danilova et al 2008). Both the 5q- syndrome and DBA show haploinsufficiency for closely related ribosomal proteins RPS-19 and RPS-14. It is assumed that the pathophysiology of the 5q- syndrome and DBA are the same (Ebert et al 2007). Because lenalidomide is effective in patients with the 5q- syndrome, lenalidomide was considered as treatment option in DBA at the Annual DBA International Consensus Conferences in 2008 and 2009. Results Due to severe pancytopenia, lenalidomide was stopped after 5 weeks. During the next 6 months a slow recovery of white blood cells and platelets was observed. Before and 3 months after the start of the lenalidomide treatment marrow cytology, histology and cytogenetics were done. Both times there was a red cell aplasia, no signs of myelodysplasia and no cytogenetic abnormalities. Rechallenge with lenalidomide in a dose of 10 mg each fourth day resulted again in a pancytopenia in 6 weeks time. Each 3 weeks 3 units of red blood cells are necessary ever since. Discussion and Conclusion Treatment of DBA with lenalidomide in this patient was unsuccessful and resulted in a temporarily and severe neutropenia and thrombocytopenia. These adverse effects are also documented in 62% of the 5q- syndrome patients treated with 10 mg Lenalidomide. Moreover, in retrospect, we doubt if lenalidomide can be effective when red cell aplasia is already present as a late complication of DBA. Disclosures: No relevant conflicts of interest to declare.


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.


1994 ◽  
Vol 16 (3) ◽  
pp. 260-265 ◽  
Author(s):  
F. Madanat ◽  
M. Arnaout ◽  
A. Hasan ◽  
M. Tarawneh ◽  
M. Shomaf ◽  
...  

2000 ◽  
Vol 111 (4) ◽  
pp. 1010-1022 ◽  
Author(s):  
Paul Fisch ◽  
Rupert Handgretinger ◽  
Hans-Eckart Schaefer

Sign in / Sign up

Export Citation Format

Share Document