Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 103(6): 930-947, 2018 12 06.
Article in English | MEDLINE | ID: mdl-30503522

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare bone marrow failure disorder that affects 7 out of 1,000,000 live births and has been associated with mutations in components of the ribosome. In order to characterize the genetic landscape of this heterogeneous disorder, we recruited a cohort of 472 individuals with a clinical diagnosis of DBA and performed whole-exome sequencing (WES). We identified relevant rare and predicted damaging mutations for 78% of individuals. The majority of mutations were singletons, absent from population databases, predicted to cause loss of function, and located in 1 of 19 previously reported ribosomal protein (RP)-encoding genes. Using exon coverage estimates, we identified and validated 31 deletions in RP genes. We also observed an enrichment for extended splice site mutations and validated their diverse effects using RNA sequencing in cell lines obtained from individuals with DBA. Leveraging the size of our cohort, we observed robust genotype-phenotype associations with congenital abnormalities and treatment outcomes. We further identified rare mutations in seven previously unreported RP genes that may cause DBA, as well as several distinct disorders that appear to phenocopy DBA, including nine individuals with biallelic CECR1 mutations that result in deficiency of ADA2. However, no new genes were identified at exome-wide significance, suggesting that there are no unidentified genes containing mutations readily identified by WES that explain >5% of DBA-affected case subjects. Overall, this report should inform not only clinical practice for DBA-affected individuals, but also the design and analysis of rare variant studies for heterogeneous Mendelian disorders.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Adolescent , Child , Child, Preschool , Cohort Studies , Exome/genetics , Exons/genetics , Female , Gene Deletion , Genetic Association Studies/methods , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mutation/genetics , Phenotype , Ribosomal Proteins/genetics , Ribosomes/genetics , Sequence Analysis, RNA/methods , Exome Sequencing/methods
2.
Haematologica ; 106(5): 1303-1310, 2021 05 01.
Article in English | MEDLINE | ID: mdl-32241839

ABSTRACT

Diamond Blackfan anemia (DBA) is predominantly an autosomal dominant inherited red cell aplasia primarily caused by pathogenic germline variants in ribosomal protein genes. DBA due to pathogenic RPL35A variants has been associated with large 3q29 deletions and phenotypes not common in DBA. We conducted a multi-institutional genotype-phenotype study of 45 patients with DBA associated with pathogenic RPL35A germline variants and curated the variant data on 21 additional cases from the literature. Genotype-phenotype analyses were conducted comparing patients with large deletions versus all other pathogenic variants in RPL35A. Twenty-two of the 45 cases had large deletions in RPL35A. After adjusting for multiple tests, a statistically significant association was observed between patients with a large deletion and steroid-resistant anemia, neutropenia, craniofacial abnormalities, chronic gastrointestinal problems, and intellectual disabilities (p<0.01) compared with all other pathogenic variants. Non-large deletion pathogenic variants were spread across RPL35A with no apparent hot spot and 56% of the individual family variants were observed more than once. In this, the largest known study of DBA patients with pathogenic RPL35A variants, we determined that patients with large deletions have a more severe phenotype that is clinically different from those with non-large deletion variants. Genes of interest also deleted in the 3q29 region that could be associated with some of these phenotypic features include LMLN and IQCG. Management of DBA due to large RPL35A deletions may be challenging due to complex problems and require comprehensive assessments by multiple specialists including immunologic, gastrointestinal, and developmental evaluations to provide optimal multidisciplinary care.


Subject(s)
Anemia, Diamond-Blackfan , Anemia, Diamond-Blackfan/genetics , Genetic Association Studies , Humans , Mutation , Phenotype , Ribosomal Proteins/genetics
3.
Am J Hematol ; 96(11): 1491-1504, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34342889

ABSTRACT

The North American Pediatric Aplastic Anemia Consortium (NAPAAC) is a group of pediatric hematologist-oncologists, hematopathologists, and bone marrow transplant physicians from 46 institutions in North America with interest and expertise in aplastic anemia, inherited bone marrow failure syndromes, and myelodysplastic syndromes. The NAPAAC Bone Marrow Failure Diagnosis and Care Guidelines Working Group was established with the charge of harmonizing the approach to the diagnostic workup of aplastic anemia in an effort to standardize best practices in the field. This document outlines the rationale for initial evaluations in pediatric patients presenting with signs and symptoms concerning for severe aplastic anemia.


Subject(s)
Anemia, Aplastic/diagnosis , Anemia, Aplastic/pathology , Bone Marrow/pathology , Child , Diagnosis, Differential , Fetal Hemoglobin/analysis , HLA Antigens/analysis , Humans , North America , Severity of Illness Index
4.
Pediatr Blood Cancer ; 68(8): e28984, 2021 08.
Article in English | MEDLINE | ID: mdl-34089224

ABSTRACT

Diamond Blackfan anemia (DBA) is a rare inherited bone marrow failure syndrome characterized by red cell failure, congenital anomalies, poor linear growth, and cancer predisposition. Two previous analyses from the Diamond Blackfan Anemia Registry have quantified DBA as a cancer predisposition syndrome of moderate cancer penetrance. Patients with DBA have a 4.8-fold higher relative risk of developing cancer with an overall cumulative incidence of 13.7% by age 45 years. The two most prevalent solid tumors are colorectal cancer (CRC) and osteogenic sarcoma. Current and evolving data support the institution of cancer screening and surveillance strategies for CRC in DBA.


Subject(s)
Anemia, Diamond-Blackfan , Bone Neoplasms , Colorectal Neoplasms , Anemia, Diamond-Blackfan/diagnosis , Anemia, Diamond-Blackfan/epidemiology , Anemia, Diamond-Blackfan/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/epidemiology , Disease Susceptibility , Early Detection of Cancer , Humans , Middle Aged , Mutation , Registries , Ribosomal Proteins/genetics
5.
Blood ; 131(4): 408-416, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29092827

ABSTRACT

Severe congenital neutropenia (SCN) and Shwachman-Diamond syndrome (SDS) are congenital neutropenia syndromes with a high rate of leukemic transformation. Hematopoietic stressors may contribute to leukemic transformation by increasing the mutation rate in hematopoietic stem/progenitor cells (HSPCs) and/or by promoting clonal hematopoiesis. We sequenced the exome of individual hematopoietic colonies derived from 13 patients with congenital neutropenia to measure total mutation burden and performed error-corrected sequencing on a panel of 46 genes on 80 patients with congenital neutropenia to assess for clonal hematopoiesis. An average of 3.6 ± 1.2 somatic mutations per exome was identified in HSPCs from patients with SCN compared with 3.9 ± 0.4 for healthy controls (P = NS). Clonal hematopoiesis due to mutations in TP53 was present in 48% (13/27) of patients with SDS but was not seen in healthy controls (0/17, P < .001) or patients with SCN (0/40, P < .001). Our SDS cohort was young (median age 6.3 years), and many of the patients had multiple TP53 mutations. Conversely, clonal hematopoiesis due to mutations of CSF3R was present in patients with SCN but was not detected in healthy controls or patients with SDS. These data show that hematopoietic stress, including granulocyte colony-stimulating factor, do not increase the mutation burden in HSPCs in congenital neutropenia. Rather, distinct hematopoietic stressors result in the selective expansion of HSPCs carrying specific gene mutations. In particular, in SDS there is enormous selective pressure to expand TP53-mutated HSPCs, suggesting that acquisition of TP53 mutations is an early, likely initiating event, in the transformation to myelodysplastic syndrome/acute myeloid leukemia in patients with SDS.


Subject(s)
Hematopoiesis , Hematopoietic Stem Cells/pathology , Mutation , Neutropenia/congenital , Adolescent , Adult , Child , Child, Preschool , Congenital Bone Marrow Failure Syndromes , Exome , Female , Hematopoietic Stem Cells/metabolism , Humans , Male , Mutation Rate , Neutropenia/genetics , Neutropenia/pathology , Neutropenia/physiopathology , Young Adult
6.
Pediatr Blood Cancer ; 67(12): e28737, 2020 12.
Article in English | MEDLINE | ID: mdl-33098753

ABSTRACT

The coagulopathy of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is well documented in adults, with increases in D-dimer and prothrombin time found to be strong predictors of mortality, and anticoagulation shown to decrease this mortality. Viscoelastic parameters such as elevations in maximum clot firmness (MCF) on rotational thromboelastometry (ROTEM) have correlated with a hypercoagulable state in adults with SARS-CoV-2. We report our experience in children infected with SARS-CoV-2, with noted elevations in D-dimer and MCF on ROTEM (indicating hypercoagulability). Exploration of viscoelastic testing to provide additional laboratory-based evidence for pediatric-specific risk assessment for thromboprophylaxis in SARS-CoV-2 is warranted.


Subject(s)
Coronavirus Infections , Pandemics , Pneumonia, Viral , Severe Acute Respiratory Syndrome , Severe acute respiratory syndrome-related coronavirus , Thrombosis , Venous Thromboembolism , Adult , Anticoagulants , Betacoronavirus , COVID-19 , Child , Humans , SARS-CoV-2
7.
Pediatr Blood Cancer ; 67(12): e28748, 2020 12.
Article in English | MEDLINE | ID: mdl-33025707

ABSTRACT

BACKGROUND: Diamond-Blackfan anemia (DBA) is an inherited bone marrow failure syndrome characterized by anemia, short stature, congenital anomalies, and cancer predisposition. Most cases are due to mutations in genes encoding ribosomal proteins (RP) leading to RP haploinsufficiency. Effective treatments for the anemia of DBA include chronic red cell transfusions, long-term corticosteroid therapy, or hematopoietic stem cell transplantation. In a small patient series and in animal models, there have been hematologic responses to L-leucine with amelioration of anemia. The study objectives of this clinical trial were to determine feasibility, safety, and efficacy of L-leucine in transfusion-dependent patients with DBA. PROCEDURE: Patients ≥2 years of age received L-leucine 700 mg/m2 orally three times daily for nine months to determine a hematologic response and any improvement in growth (NCT01362595). RESULTS: This multicenter, phase I/II study enrolled 55 subjects; 43 were evaluable. There were 21 males; the median age at enrollment was 10.4 years (range, 2.5-46.1 years). No significant adverse events were attributable to L-leucine. Two subjects had a complete erythroid response and five had a partial response. Nine of 25, and 11 of 25, subjects experienced a positive weight and height percentile change, respectively, at the end of therapy. CONCLUSIONS: L-leucine is safe, resulted in an erythroid response in 16% of subjects with DBA, and led to an increase in weight and linear growth velocity in 36% and 44% of evaluable subjects, respectively. Further studies will be critical to understand the role of L-leucine in the management of patients with DBA.


Subject(s)
Anemia, Diamond-Blackfan/therapy , Blood Transfusion/methods , Leucine/therapeutic use , Adolescent , Adult , Anemia, Diamond-Blackfan/pathology , Child , Child, Preschool , Combined Modality Therapy , Feasibility Studies , Female , Follow-Up Studies , Humans , Male , Middle Aged , Pilot Projects , Prognosis , Young Adult
8.
Blood ; 129(23): 3111-3120, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28377399

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital bone marrow failure syndrome characterized by erythroid hypoplasia, usually without perturbation of other hematopoietic lineages. Approximately 65% of DBA patients with autosomal dominant inheritance have heterozygous mutations or deletions in ribosomal protein (RP) genes while <1% of patients with X-linked inheritance have been identified with mutations in the transcription factor GATA1 Erythroid cells from patients with DBA have not been well characterized, and the mechanisms underlying the erythroid specific effects of either RP or GATA1 associated DBA remain unclear. We have developed an ex vivo culture system to expand peripheral blood CD34+ progenitor cells from patients with DBA and differentiate them into erythroid cells. Cells from patients with RP or GATA1 mutations showed decreased proliferation and delayed erythroid differentiation in comparison with controls. RNA transcript analyses of erythroid cells from controls and patients with RP or GATA1 mutations showed distinctive differences, with upregulation of heme biosynthesis genes prominently in RP-mediated DBA and failure to upregulate components of the translational apparatus in GATA1-mediated DBA. Our data show that dysregulation of translation is a common feature of DBA caused by both RP and GATA1 mutations. This trial was registered at www.clinicaltrials.gov as #NCT00106015.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Adolescent , Adult , Anemia, Diamond-Blackfan/blood , Anemia, Diamond-Blackfan/metabolism , Case-Control Studies , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Child , Child, Preschool , Erythroid Cells/metabolism , Erythroid Cells/pathology , Erythropoiesis/genetics , Female , GATA1 Transcription Factor/genetics , Genes, Dominant , Genes, X-Linked , Humans , Male , Models, Genetic , Mutation , Ribosomal Proteins/genetics , Transcriptome , Young Adult
9.
Haematologica ; 104(10): 1974-1983, 2019 10.
Article in English | MEDLINE | ID: mdl-30948484

ABSTRACT

Quality of response to immunosuppressive therapy and long-term outcomes for pediatric severe aplastic anemia remain incompletely characterized. Contemporary evidence to inform treatment of relapsed or refractory severe aplastic anemia for pediatric patients is also limited. The clinical features and outcomes for 314 children treated from 2002 to 2014 with immunosuppressive therapy for acquired severe aplastic anemia were analyzed retrospectively from 25 institutions in the North American Pediatric Aplastic Anemia Consortium. The majority of subjects (n=264) received horse anti-thymocyte globulin (hATG) plus cyclosporine (CyA) with a median 61 months follow up. Following hATG/CyA, 71.2% (95%CI: 65.3,76.6) achieved an objective response. In contrast to adult studies, the quality of response achieved in pediatric patients was high, with 59.8% (95%CI: 53.7,65.8) complete response and 68.2% (95%CI: 62.2,73.8) achieving at least a very good partial response with a platelet count ≥50×109L. At five years post-hATG/CyA, overall survival was 93% (95%CI: 89,96), but event-free survival without subsequent treatment was only 64% (95%CI: 57,69) without a plateau. Twelve of 171 evaluable patients (7%) acquired clonal abnormalities after diagnosis after a median 25.2 months (range: 4.3-71 months) post treatment. Myelodysplastic syndrome or leukemia developed in 6 of 314 (1.9%). For relapsed/refractory disease, treatment with a hematopoietic stem cell transplant had a superior event-free survival compared to second immunosuppressive therapy treatment in a multivariate analysis (HR=0.19, 95%CI: 0.08,0.47; P=0.0003). This study highlights the need for improved therapies to achieve sustained high-quality remission for children with severe aplastic anemia.


Subject(s)
Anemia, Aplastic/drug therapy , Antilymphocyte Serum/administration & dosage , Cyclosporine/administration & dosage , Immunosuppression Therapy , Anemia, Aplastic/epidemiology , Anemia, Aplastic/pathology , Antilymphocyte Serum/adverse effects , Child, Preschool , Cyclosporine/adverse effects , Female , Follow-Up Studies , Humans , Infant , Male , Retrospective Studies , United States/epidemiology
10.
Hum Mutat ; 39(8): 1102-1111, 2018 08.
Article in English | MEDLINE | ID: mdl-29766597

ABSTRACT

Diamond-Blackfan anemia (DBA) is a rare genetic hypoplasia of erythroid progenitors characterized by mild to severe anemia and associated with congenital malformations. Clinical manifestations in DBA patients are quite variable and genetic testing has become a critical factor in establishing a diagnosis of DBA. The majority of DBA cases are due to heterozygous loss-of-function mutations in ribosomal protein (RP) genes. Causative mutations are fairly straightforward to identify in the case of large deletions and frameshift and nonsense mutations found early in a protein coding sequence, but diagnosis becomes more challenging in the case of missense mutations and small in-frame indels. Our group recently characterized the phenotype of lymphoblastoid cell lines established from DBA patients with pathogenic lesions in RPS19 and observed that defective pre-rRNA processing, a hallmark of the disease, was rescued by lentiviral vectors expressing wild-type RPS19. Here, we use this complementation assay to determine whether RPS19 variants of unknown significance are capable of rescuing pre-rRNA processing defects in these lymphoblastoid cells as a means of assessing the effects of these sequence changes on the function of the RPS19 protein. This approach will be useful in differentiating pathogenic mutations from benign polymorphisms in identifying causative genes in DBA patients.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Ribosomal Proteins/genetics , Cell Line , Codon, Nonsense/genetics , Computational Biology , DNA, Complementary/genetics , Frameshift Mutation/genetics , Humans , Mutation/genetics , Phenotype
11.
Blood ; 127(11): 1481-92, 2016 Mar 17.
Article in English | MEDLINE | ID: mdl-26679864

ABSTRACT

Current therapeutic strategies for sickle cell anemia are aimed at reactivating fetal hemoglobin. Pomalidomide, a third-generation immunomodulatory drug, was proposed to induce fetal hemoglobin production by an unknown mechanism. Here, we report that pomalidomide induced a fetal-like erythroid differentiation program, leading to a reversion of γ-globin silencing in adult human erythroblasts. Pomalidomide acted early by transiently delaying erythropoiesis at the burst-forming unit-erythroid/colony-forming unit-erythroid transition, but without affecting terminal differentiation. Further, the transcription networks involved in γ-globin repression were selectively and differentially affected by pomalidomide including BCL11A, SOX6, IKZF1, KLF1, and LSD1. IKAROS (IKZF1), a known target of pomalidomide, was degraded by the proteasome, but was not the key effector of this program, because genetic ablation of IKZF1 did not phenocopy pomalidomide treatment. Notably, the pomalidomide-induced reprogramming was conserved in hematopoietic progenitors from individuals with sickle cell anemia. Moreover, multiple myeloma patients treated with pomalidomide demonstrated increased in vivo γ-globin levels in their erythrocytes. Together, these data reveal the molecular mechanisms by which pomalidomide reactivates fetal hemoglobin, reinforcing its potential as a treatment for patients with ß-hemoglobinopathies.


Subject(s)
Hematopoietic Stem Cells/drug effects , Thalidomide/analogs & derivatives , Transcription, Genetic/drug effects , gamma-Globins/genetics , Adult , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/genetics , Carrier Proteins/blood , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/drug effects , Erythroid Precursor Cells/metabolism , Erythropoiesis/drug effects , Fetal Hemoglobin/biosynthesis , Gene Expression Regulation, Developmental , Genetic Vectors/genetics , Hematopoietic Stem Cells/metabolism , Histone Demethylases/blood , Humans , Ikaros Transcription Factor/blood , Ikaros Transcription Factor/drug effects , Kruppel-Like Transcription Factors/blood , Lentivirus/genetics , Multiple Myeloma/blood , Multiple Myeloma/genetics , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Proteins/blood , Proteasome Endopeptidase Complex/metabolism , RNA Interference , RNA, Small Interfering/genetics , Repressor Proteins , SOXD Transcription Factors/blood , Thalidomide/pharmacology , beta-Globins/biosynthesis , beta-Globins/genetics , gamma-Globins/biosynthesis
12.
Biol Blood Marrow Transplant ; 23(5): 726-735, 2017 May.
Article in English | MEDLINE | ID: mdl-28115275

ABSTRACT

Fanconi anemia (FA), dyskeratosis congenita (DC), and Diamond Blackfan anemia (DBA) are 3 of the most common inherited bone marrow failure syndromes (IBMFS), in which the hematologic manifestations can be cured with hematopoietic cell transplantation (HCT). Later in life, these patients face a variety of medical conditions, which may be a manifestation of underlying disease or due to pre-HCT therapy, the HCT, or a combination of all these elements. Very limited long-term follow-up data exist in these populations, with FA the only IBMFS that has specific published data. During the international consensus conference sponsored by the Pediatric Blood and Marrow Transplant Consortium entitled "Late Effects Screening and Recommendations following Allogeneic Hematopoietic Cell Transplant (HCT) for Immune Deficiency and Nonmalignant Hematologic Disease" held in Minneapolis, Minnesota in May of 2016, a half-day session was focused specifically on the unmet needs for these patients with IBMFS. A multidisciplinary group of experts discussed what is currently known, outlined an agenda for future research, and laid out long-term follow-up guidelines based on a combination of evidence in the literature as well as expert opinion. This article addresses the state of science in that area as well as consensus regarding the agenda for future research, with specific screening guidelines to follow in the next article from this group.


Subject(s)
Anemia, Aplastic/therapy , Bone Marrow Diseases/therapy , Hematopoietic Stem Cell Transplantation/methods , Hemoglobinuria, Paroxysmal/therapy , Long Term Adverse Effects , Anemia, Aplastic/complications , Anemia, Diamond-Blackfan/complications , Anemia, Diamond-Blackfan/therapy , Biomedical Research/methods , Biomedical Research/trends , Bone Marrow Diseases/complications , Bone Marrow Failure Disorders , Child , Dyskeratosis Congenita/complications , Dyskeratosis Congenita/therapy , Fanconi Anemia/complications , Fanconi Anemia/therapy , Forecasting , Hematopoietic Stem Cell Transplantation/adverse effects , Hemoglobinuria, Paroxysmal/complications , Humans , Practice Guidelines as Topic/standards
13.
Biol Blood Marrow Transplant ; 23(9): 1422-1428, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28533057

ABSTRACT

Patients with inherited bone marrow failure syndromes (IBMFS), such as Fanconi anemia (FA), dyskeratosis congenita (DC), or Diamond Blackfan anemia (DBA), can have hematologic manifestations cured through hematopoietic cell transplantation (HCT). Subsequent late effects seen in these patients arise from a combination of the underlying disease, the pre-HCT therapy, and the HCT process. During the international consensus conference sponsored by the Pediatric Blood and Marrow Transplant Consortium on late effects screening and recommendations following allogeneic hematopoietic cell transplantation for immune deficiency and nonmalignant hematologic diseases held in Minneapolis, Minnesota in May 2016, a half-day session was focused specifically on the unmet needs for these patients with IBMFS. This multidisciplinary group of experts in rare diseases and transplantation late effects has already published on the state of the science in this area, along with discussion of an agenda for future research. This companion article outlines consensus disease-specific long-term follow-up screening guidelines for patients with IMBFS.


Subject(s)
Anemia, Aplastic/diagnosis , Anemia, Diamond-Blackfan/diagnosis , Bone Marrow Diseases/diagnosis , Dyskeratosis Congenita/diagnosis , Fanconi Anemia/diagnosis , Hematopoietic Stem Cell Transplantation , Hemoglobinuria, Paroxysmal/diagnosis , Anemia, Aplastic/immunology , Anemia, Aplastic/pathology , Anemia, Aplastic/therapy , Anemia, Diamond-Blackfan/immunology , Anemia, Diamond-Blackfan/mortality , Anemia, Diamond-Blackfan/therapy , Bone Marrow Diseases/immunology , Bone Marrow Diseases/pathology , Bone Marrow Diseases/therapy , Bone Marrow Failure Disorders , Child , Consensus , Consensus Development Conferences as Topic , Dyskeratosis Congenita/immunology , Dyskeratosis Congenita/mortality , Dyskeratosis Congenita/therapy , Fanconi Anemia/immunology , Fanconi Anemia/mortality , Fanconi Anemia/therapy , Hemoglobinuria, Paroxysmal/immunology , Hemoglobinuria, Paroxysmal/pathology , Hemoglobinuria, Paroxysmal/therapy , Humans , International Cooperation , Survival Analysis , Transplantation, Homologous
15.
Pediatr Blood Cancer ; 63(2): 306-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26496000

ABSTRACT

BACKGROUND: Diamond-Blackfan anemia (DBA) is a rare inherited bone marrow failure syndrome. The mainstays of treatment involve chronic red cell transfusions, long-term glucocorticoid therapy, and stem cell transplantation. Systematic data concerning endocrine function in DBA are limited. We studied patients in the DBA Registry (DBAR) of North America to assess the prevalence of various endocrinopathies. PROCEDURE: In a pilot study, retrospective data were collected for 12 patients with DBA. Subsequently, patients with DBA aged 1-39 years were recruited prospectively. Combined, 57 patients were studied; 38 chronically transfused, 12 glucocorticoid-dependent, and seven in remission. Data were collected on anthropometric measurements, systematic screening of pituitary, thyroid, parathyroid, adrenal, pancreatic, and gonadal function, and ferritin levels. Descriptive statistics were tabulated and group differences were assessed. RESULTS: Fifty-three percent of patients had ≥ 1 endocrine disorder, including adrenal insufficiency (32%), hypogonadism (29%), hypothyroidism (14%), growth hormone dysfunction (7%), diabetes mellitus (2%), and/or diabetes insipidus (2%). Ten of the 33 patients with available heights had height standard deviation less than -2. Low 25-hydroxy vitamin D (25(OH)D) levels were present in 50%. A small proportion also had osteopenia, osteoporosis, or hypercalciuria. Most with adrenal insufficiency were glucocorticoid dependent; other endocrinopathies were more common in chronically transfused patients. CONCLUSIONS: Endocrine dysfunction is common in DBA, as early as the teenage years. Although prevalence is highest in transfused patients, patients taking glucocorticoids or in remission also have endocrine dysfunction. Longitudinal studies are needed to better understand the etiology and true prevalence of these disorders.


Subject(s)
Anemia, Diamond-Blackfan/complications , Endocrine System Diseases/epidemiology , Endocrine System Diseases/etiology , Adolescent , Adult , Anemia, Diamond-Blackfan/therapy , Blood Transfusion , Child , Child, Preschool , Female , Glucocorticoids/therapeutic use , Humans , Infant , Male , Pilot Projects , Prevalence , Registries , Retrospective Studies , Young Adult
19.
Blood ; 122(14): 2487-90, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-23943650

ABSTRACT

Classical 5q- syndrome is an acquired macrocytic anemia of the elderly. Similar to Diamond Blackfan anemia (DBA), an inherited red cell aplasia, the bone marrow is characterized by a paucity of erythroid precursors. RPS14 deletions in combination with other deletions in the region have been implicated as causative of the 5q- syndrome phenotype. We asked whether smaller, less easily detectable deletions could account for a syndrome with a modified phenotype. We employed single-nucleotide polymorphism array genotyping to identify small deletions in patients diagnosed with DBA and other anemias lacking molecular diagnoses. Diminutive mosaic deletions involving RPS14 were identified in a 5-year-old patient with nonclassical DBA and in a 17-year-old patient with myelodysplastic syndrome. Patients with nonclassical DBA and other hypoproliferative anemias may have somatically acquired 5q deletions with RPS14 haploinsufficiency not identified by fluorescence in situ hybridization or cytogenetic testing, thus refining the spectrum of disorders with 5q- deletions.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Macrocytic/genetics , Cytogenetic Analysis/methods , Ribosomal Proteins/genetics , Adolescent , Anemia, Diamond-Blackfan/diagnosis , Anemia, Macrocytic/diagnosis , Anemia, Macrocytic/drug therapy , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , Female , Genotype , Humans , Immunologic Factors/therapeutic use , Lenalidomide , Phenotype , Polymorphism, Single Nucleotide , Real-Time Polymerase Chain Reaction , Thalidomide/analogs & derivatives , Thalidomide/therapeutic use
20.
Blood ; 119(16): 3815-9, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22362038

ABSTRACT

Diamond Blackfan anemia (DBA) is an inherited bone marrow failure syndrome characterized by red cell aplasia and congenital anomalies. A predisposition to cancer has been suggested but not quantified by case reports. The DBA Registry of North America (DBAR) is the largest established DBA patient cohort, with prospective follow-up since 1991. This report presents the first quantitative assessment of cancer incidence in DBA. Among 608 patients with 9458 person-years of follow-up, 15 solid tumors, 2 acute myeloid leukemias, and 2 cases of myelodysplastic syndrome were diagnosed at a median age of 41 years in patients who had not received a bone marrow transplant. Cancer incidence in DBA was significantly elevated. The observed-to- expected ratio for all cancers combined was 5.4 (P < .05); significant observed-to-expected ratios were 287 for myelodysplastic syndrome, 28 for acute myeloid leukemia, 36 for colon carcinoma, 33 for osteogenic sarcoma, and 12 for female genital cancers. The median survival was 56 years, and the cumulative incidence of solid tumor/leukemia was approximately 20% by age 46 years. As in Fanconi anemia and dyskeratosis congenita, DBA is both an inherited bone marrow failure syndrome and a cancer predisposition syndrome; cancer risks appear lower in DBA than in Fanconi anemia or dyskeratosis congenita. This trial was registered at www.clinicaltrials.gov as #NCT00106015.


Subject(s)
Anemia, Diamond-Blackfan/mortality , Anemia, Diamond-Blackfan/therapy , Bone Marrow Transplantation , Hemoglobinuria, Paroxysmal/mortality , Neoplasms/mortality , Registries/statistics & numerical data , Adolescent , Adult , Aged , Anemia, Aplastic , Bone Marrow Diseases , Bone Marrow Failure Disorders , Child , Child, Preschool , Comorbidity , Female , Follow-Up Studies , Hemoglobinuria, Paroxysmal/therapy , Humans , Incidence , Infant , Male , Middle Aged , Risk Factors , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL