Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 276
Filter
1.
Leuk Lymphoma ; 63(8): 1964-1969, 2022 08.
Article in English | MEDLINE | ID: mdl-35343360

ABSTRACT

We examine changes in population level incidence and survival of patients diagnosed with myelodysplastic syndrome (MDS) in the United States in 2001-2016. Data were extracted from the Surveillance, Epidemiology, and End Results (SEER)-18 database. Period analysis was used to calculate one-, two-, and five-year survival. The incidence peaked at 5.6 per 100,000 in 2010 then decreased to 3.9 by 2016, with a decrease in the diagnoses of refractory anemia (RA) and RA with ringed sideroblasts (RARS) and a relative increase in RA with excess blasts (RAEB). Overall, one-, two-, and five-year relative survival decreased over time, going from 74.3%, 60.9%, and 42.3%, respectively, in 2008-2010 to 70.9%, 55.9%, and 37.6%, respectively, in 2014-2016. When survival was examined by histology, patients with RA/RARS and RAEB had similar survival expectations in 2008-2010 versus 2014-2016 and a decrease was observed for 5q-MDS. Our results highlight the need for new treatment options in MDS.


Subject(s)
Anemia, Refractory, with Excess of Blasts , Anemia, Refractory , Myelodysplastic Syndromes , Anemia, Refractory/genetics , Anemia, Refractory, with Excess of Blasts/genetics , Chromosome Deletion , Humans , Incidence , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/therapy
2.
Leuk Res ; 112: 106757, 2022 01.
Article in English | MEDLINE | ID: mdl-34864369

ABSTRACT

We studied 79 patients with AML-MRC or RAEB-T, who were later reclassified according to the WHO classification. Marrow slides were examined cytomorphologically with regard to dysplasia. Patients were followed up until March 2020. Thirty-one patients underwent allogeneic stem cell transplantation (median survival (ms) 16 months), 14 were treated with induction chemotherapy (ms 8.4 months), 18 received hypomethylating agents (ms 9.2 months), 16 received low dose chemotherapy or best supportive care (ms 2.4 months). Only 30.4 % fulfilled the morphologic WHO criteria. 46.8 % were classified as AML-MRC by an antecedent MDS, 54.4 % of the pts were classified by MDS-related chromosomal abnormalities. 5 % did not fulfill any of the criteria and were entered based on 20-29 % medullary blasts. There was no difference in ms between pts presenting with > 50 % dysplasia as compared to pts with dysplasia between 10 % and 50 % (ms 9.1 vs 9.9 months, p = n.s.) or for pts with antecedent MDS (ms 9.1 vs 8.9 months, p = n.s.). Myelodysplasia-related cytogenetic abnormalities were associated with a worse outcome (ms 8.1 vs 13.5 months, p = 0.026). AML-MRC in its current definition is a heterogenous entity. Dysplasia of ≥ 50 % in ≥ two lineages is not helpful for diagnostics and prognostication and therefore should be deleted in future classifications. We recommend utilizing the WHO guidelines for defining dysplasia (10 % or greater in ≥ 1 of the three myeloid cell lines) assisting in establishing the diagnosis of MDS.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid/genetics , Myelodysplastic Syndromes/genetics , Acute Disease , Aged , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/therapy , Chromosome Aberrations/statistics & numerical data , Female , Humans , Leukemia, Myeloid/diagnosis , Leukemia, Myeloid/therapy , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Outcome Assessment, Health Care/methods , Retrospective Studies , Survival Analysis , World Health Organization
3.
Cancer Sci ; 112(5): 1888-1898, 2021 May.
Article in English | MEDLINE | ID: mdl-33560542

ABSTRACT

Circular RNAs (circRNAs) have been identified to exert vital biological functions and can be used as new biomarkers in a number of tumors. However, little is known about the functions of circRNAs in myelodysplastic syndrome (MDS). Here, we aimed to investigate circRNA expression profiles and to investigate the functional and clinical value of circRNAs in MDS. Differential expression of circRNAs between MDS and control subjects was analyzed using circRNA arrays, in which we identified 145 upregulated circRNAs and 224 downregulated circRNAs. Validated by real-time quantitative PCR between 100 MDS patients and 20 controls, three upregulated (hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_104634) and three downregulated (hsa_circRNA_103846, hsa_circRNA_102817, and hsa_circRNA_102526) circRNAs matched the arrays. The receiver operating characteristic curve analysis of these circRNAs showed that the area under the curve was 0.7266, 0.8676, 0.7349, 0.7091, 0.8806, and 0.7472, respectively. Kaplan-Meier survival analysis showed that only hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_102817 were significantly associated with overall survival. Furthermore, we generated a competing endogenous RNA network focused on hsa_circRNA_100352, hsa_circRNA_104056, and hsa_circRNA_102817. Analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes showed that the three circRNAs were linked with some important cancer-related functions and pathways.


Subject(s)
Biomarkers, Tumor/metabolism , Myelodysplastic Syndromes/metabolism , RNA, Circular/metabolism , Aged , Anemia, Refractory/genetics , Anemia, Refractory/metabolism , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Anemia, Sideroblastic/genetics , Anemia, Sideroblastic/metabolism , Area Under Curve , Biomarkers, Tumor/genetics , Bone Marrow/metabolism , Case-Control Studies , Down-Regulation , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Myelodysplastic Syndromes/blood , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , RNA, Circular/genetics , ROC Curve , Real-Time Polymerase Chain Reaction , Statistics, Nonparametric , Up-Regulation
4.
Clin Lymphoma Myeloma Leuk ; 21(2): e206-e211, 2021 02.
Article in English | MEDLINE | ID: mdl-33293239

ABSTRACT

BACKGROUND: Recent data suggest significant underutilization of hypomethylating agents (HMAs) that are recommended treatments for patients with myelodysplastic syndromes (MDS) with refractory anemia with excess blasts (RAEB). The study objective was to assess the degree of HMA use and predictors of HMA underuse in this population. PATIENTS AND METHODS: This was a retrospective study including patients diagnosed with the RAEB form of MDS between January 2011 and December 2015 using the Surveillance, Epidemiology, and End Results-Medicare linked database. Patients were excluded if they had < 1 year of continuous enrollment before diagnosis or received stem cell transplant or lenalidomide during the follow-up period. HMA non-peristence was defined as use of < 4 cycles (3-10 HMA days/28 days) of HMAs or a gap of ≥ 90 days between consecutive cycles. Patients were characterized as HMA never-users, HMA-persistent users, and HMA-non-persistent users. Descriptive statistics were used to summarize patient characteristics. Multivariable logistic regression was used to assess predictors of HMA underuse and persistence. RESULTS: Of the 1190 patients, 526 (44%) were never-users, 295 (25%) were non-persistent users, and 369 (31%) were persistent users. Age at diagnosis (eg, 66-70 years vs. ≥ 80 years; odds ratio [OR], 2.36; 95% confidence interval [CI], 1.56-3.56), marital status (single vs. married; OR, 0.67; 95% CI, 0.51-0.89), National Cancer Institute comorbidity index (≥ 3 vs. 0-1; OR, 0.62; 95% CI, 0.46-0.83), and performance status (poor vs. good; OR, 0.67; 95% CI, 0.51-0.87) were significantly associated with HMA underuse. CONCLUSION: Several demographic and clinical factors were associated with underuse of HMAs. There is need for a better understanding of suboptimal HMA use and its relationship with clinical response.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antimetabolites, Antineoplastic/therapeutic use , Azacitidine/therapeutic use , DNA Methylation/drug effects , Drug Utilization/statistics & numerical data , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/genetics , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Retrospective Studies , SEER Program/statistics & numerical data , United States
5.
Hematol Oncol ; 38(4): 531-540, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32469434

ABSTRACT

Patients with acute myeloid leukemia (AML) evolving from myelodysplastic syndrome (MDS) or higher-risk MDS have limited treatment options and poor prognosis. Our previous single-center study of decitabine followed by low dose idarubicin and cytarabine (D-IA) in patients with myeloid neoplasms showed promising primary results. We therefore conducted a multicenter study of D-IA regimen in AML evolving from MDS and higher-risk MDS. Patients with AML evolving from MDS or refractory anemia with excess blasts type 2 (RAEB-2) (based on the 2008 WHO classification) were included. The D-IA regimen (decitabine, 20 mg/m2 daily, days 1 to 3; idarubicin, 6 mg/m2 daily, days 4 to 6; cytarabine 25 mg/m2 every 12 hours, days 4 to 8; granulocyte colony stimulating factor [G-CSF], 5 µg/kg, from day 4 until neutrophil count increased to 1.0 × 109 /L) was administered as induction chemotherapy. Seventy-one patients were enrolled and treated, among whom 44 (62.0%) had AML evolving from MDS and 27 (38.0%) had RAEB-2. Twenty-eight (63.6%) AML patients achieved complete remission (CR) or complete remission with incomplete blood count recovery (CRi): 14 (31.8%) patients had CR and 14 (31.8%) had CRi. Six (22.2%) MDS patients had CR and 15 (55.6%) had marrow complete remission. The median overall survival (OS) was 22.4 months for the entire group, with a median OS of 24.2 months for AML and 20.0 months for MDS subgroup. No early death occurred. In conclusion, the D-IA regimen was effective and well tolerated, representing an alternative option for patients with AML evolving from MDS or MDS subtype RAEB-2.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Clonal Evolution , Epigenesis, Genetic , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Adult , Aged , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/pathology , Cytarabine/administration & dosage , Decitabine/administration & dosage , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , High-Throughput Nucleotide Sequencing , Humans , Idarubicin/administration & dosage , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Prognosis , Prospective Studies , Survival Rate , Young Adult
6.
Blood ; 135(25): 2271-2285, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32202636

ABSTRACT

SETD2, the histone H3 lysine 36 methyltransferase, previously identified by us, plays an important role in the pathogenesis of hematologic malignancies, but its role in myelodysplastic syndromes (MDSs) has been unclear. In this study, low expression of SETD2 correlated with shortened survival in patients with MDS, and the SETD2 levels in CD34+ bone marrow cells of those patients were increased by decitabine. We knocked out Setd2 in NUP98-HOXD13 (NHD13) transgenic mice, which phenocopies human MDS, and found that loss of Setd2 accelerated the transformation of MDS into acute myeloid leukemia (AML). Loss of Setd2 enhanced the ability of NHD13+ hematopoietic stem and progenitor cells (HSPCs) to self-renew, with increased symmetric self-renewal division and decreased differentiation and cell death. The growth of MDS-associated leukemia cells was inhibited though increasing the H3K36me3 level by using epigenetic modifying drugs. Furthermore, Setd2 deficiency upregulated hematopoietic stem cell signaling and downregulated myeloid differentiation pathways in the NHD13+ HSPCs. Our RNA-seq and chromatin immunoprecipitation-seq analysis indicated that S100a9, the S100 calcium-binding protein, is a target gene of Setd2 and that the addition of recombinant S100a9 weakens the effect of Setd2 deficiency in the NHD13+ HSPCs. In contrast, downregulation of S100a9 leads to decreases of its downstream targets, including Ikba and Jnk, which influence the self-renewal and differentiation of HSPCs. Therefore, our results demonstrated that SETD2 deficiency predicts poor prognosis in MDS and promotes the transformation of MDS into AML, which provides a potential therapeutic target for MDS-associated acute leukemia.


Subject(s)
Anemia, Refractory, with Excess of Blasts/pathology , Calgranulin B/physiology , Histone-Lysine N-Methyltransferase/deficiency , Histone-Lysine N-Methyltransferase/physiology , Leukemia, Myeloid, Acute/etiology , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Animals , Calgranulin B/biosynthesis , Calgranulin B/genetics , Cell Transformation, Neoplastic , Cells, Cultured , Decitabine/pharmacology , Down-Regulation , Gene Expression Regulation, Leukemic , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/pathology , Histone Code/drug effects , Histone-Lysine N-Methyltransferase/biosynthesis , Histone-Lysine N-Methyltransferase/genetics , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelodysplastic Syndromes/pathology , Nuclear Pore Complex Proteins/genetics , Oncogene Proteins, Fusion/genetics , Prognosis , Recombinant Proteins/therapeutic use , Time Factors , Tissue Array Analysis , Transcriptome
8.
Leuk Lymphoma ; 61(5): 1178-1187, 2020 05.
Article in English | MEDLINE | ID: mdl-31878809

ABSTRACT

Hypomethylating agents (HMA) showed overall survival (OS) benefits in patients with higher-risk myelodysplastic syndromes (HR-MDS) in clinical trials. We conducted a retrospective cohort study of Surveillance, Epidemiology, and End Results (SEER)-Medicare data of patients ≥66 years diagnosed with refractory anemia with excess blasts (RAEB), a proxy for HR-MDS, in 01/2001-04/2004 (pre-period) or 01/2006-12/2011 (post-period). Association between post-period diagnosis and OS was examined using propensity scores (PS)-matched samples. Among 1876 RAEB patients, median OS was 9 months and 30.8% received HMAs (3.6% in pre-period; 43.0% in post-period) with no association between post-period diagnosis and OS. In the top PS quartile, post-period diagnosis was associated with a 74% lower risk of death (Hazard ratio [HR] = 0.26, 95%-CI: 0.10-0.69, p = 0.007), while outcomes were worse in the lowest PS quartile (HR = 2.80, 95%-CI: 1.06-7.36, p = 0.037). HMA lead to a 3-month OS benefit for patients most likely to receive HMA but not for unselected RAEB cohort.


Subject(s)
Anemia, Refractory, with Excess of Blasts , Myelodysplastic Syndromes , Aged , Anemia, Refractory, with Excess of Blasts/drug therapy , Anemia, Refractory, with Excess of Blasts/genetics , Azacitidine , Humans , Medicare , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Propensity Score , Retrospective Studies , United States/epidemiology
10.
Cancer ; 123(14): 2661-2670, 2017 Jul 15.
Article in English | MEDLINE | ID: mdl-28324640

ABSTRACT

BACKGROUND: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only treatment with a curative potential for myelodysplastic syndrome (MDS) patients. Allo-HSCT has substantial risks, particularly in the elderly, and its role for older MDS patients has yet to be defined. METHODS: We analyzed 88 MDS patients aged ≥ 60 years with allo-HSCT after reduced intensity conditioning regimens over the last decade. The study cohort had high risk features; 47 of 88 (53.4%) patients were > 65 years of age, 24 (27%) patients had cytogenetic abnormalities consistent with monosomal karyotype (MKpos), 33 (38%) patients had histological subtype of RAEB-1 and RAEB-2 at diagnosis, and 45 (51%) patients had a hematopoietic cell transplantation-comorbidity index (HCT-CI) of ≥ 3. RESULTS: The 3-year incidence of progression, transplant-related mortality (TRM), and overall survival (OS) were 26% (95% confidence interval [CI], 18%-37%), 35% (95% CI, 26%-47%), and 41% (95% CI, 30%-52%), respectively. MKpos was the only prognostic factor that increased the risk of disease progression compared with good-risk cytogenetics (hazard ratio [HR] = 9.5, P = .003) as well as MKneg (HR = 3.3, P = .01). For TRM, HCT-CI ≥ 3, but not age >65 years, was associated with worse outcomes (HR = 3.1, P = .007). Cytogenetics and HCT-CI enabled us to identify prognostic groups for OS. MKpos patients had the worst 3-year OS (17%), whereas patients with good-risk cytogenetics and HCT-CI < 3 had the best OS (92%). CONCLUSION: Our results confirm that allo-HSCT can provide long-term survival in older MDS patients. Cytogenetics and HCT-CI identify prognostic risk groups and guide selection of older MDS patients who are candidates for allo-HSCT. Cancer 2017;123:2661-70. © 2017 American Cancer Society.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Myelodysplastic Syndromes/therapy , Transplantation Conditioning/methods , Transplantation, Homologous/methods , Aged , Anemia, Refractory, with Excess of Blasts/epidemiology , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/therapy , Cause of Death , Chromosome Aberrations , Comorbidity , Disease Progression , Female , Graft vs Host Disease/epidemiology , Humans , Leukemia, Myelomonocytic, Chronic/epidemiology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/therapy , Male , Middle Aged , Monosomy/genetics , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/genetics , Prognosis , Proportional Hazards Models , Retrospective Studies , Risk Factors , Survival Rate , Treatment Outcome
11.
J Cancer Res Clin Oncol ; 143(5): 873-882, 2017 May.
Article in English | MEDLINE | ID: mdl-28108816

ABSTRACT

PURPOSE: The aim of this study was to examine whether decitabine priming prior to low-dose chemotherapeutic regimens could improve outcomes in patients with myelodysplastic syndromes-refractory anemia with excess of blasts (MDS-RAEB). METHODS: The current retrospective analysis included all MDS-RAEB patients receiving idarubicin/cytarabine (IA) or aclacinomycin/cytarabine (AA), with or without decitabine priming during a period from February 2010 to May 2015. Treatment response and toxicity were compared between patients receiving decitabine priming and those who did not. A panel of 6 MDS-related genes was examined using bone marrow specimens. RESULTS: A total of 81 patients were included in the analysis: 40 received decitabine priming prior to chemotherapy (decitabine priming group). The median follow-up was 10.9 months (IQR: 6.2-21.9). The rate of overall response (OR) and complete remission (CR) was significantly higher in the decitabine priming group than in the chemotherapy group (OR: 75.0 vs. 51.2%, p = 0.027; CR: 55.0 vs. 29.3%, p = 0.019). Overall survival (OS) did not differ significantly between the two groups (19.5 vs. 14.7 months, p = 0.082). In a subgroup analysis that included only patients at < 60 years of age, the CR rate in the decitabine priming group was significantly higher than in the chemotherapy group (65.5 vs. 31.0%, p = 0.009). Survival benefit of decitabine priming was apparent in patients at < 60 years of age (22.4 months with 95% CI of 6.7-38.1 vs. 14.7 months with 95% CI of 11.4-18.0 months in the chemotherapy group, p = 0.028), patients with intermediate and unfavorable karyotypes (22.4 months with 95% CI of 15.1-29.7 vs. 11.9 months with 95% CI of 4.0-19.8 months in the chemotherapy group, p = 0.042), and patients with mutated splicing factor genes (35.3 months with 95% CI of 21.4-49.2 vs. 17.8 months with 95% CI of 13.8-21.8 months in the chemotherapy group, p = 0.039). Grade 3-4 hematological and non-hematological toxicities were not significantly different between the two groups. CONCLUSIONS: Decitabine priming prior to low-dose chemotherapy could improve treatment responses in patients with MDS-RAEB.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Aclarubicin/administration & dosage , Aclarubicin/analogs & derivatives , Adult , Anemia, Refractory, with Excess of Blasts/genetics , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Cytarabine/administration & dosage , Decitabine , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Humans , Idarubicin/administration & dosage , Karyotype , Male , Middle Aged , Mutation , Retrospective Studies
12.
Pril (Makedon Akad Nauk Umet Odd Med Nauki) ; 38(3): 157-162, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29668469

ABSTRACT

Myelodysplastic syndrome (MDS) is a diverse group of clonal hematologic neoplasms. The only curative treatment for MDS is allogeneic stem cell transplantation (SCT). Epigenetic changes play an important role in the pathogenesis of MDS and treatment with DNA methyl transferase inhibitors, Azacitidine, significantly prolong the survival of high-risk MDS patients. Here we report a case of a 58-year-old male presented with pancytopenia, macrocytosis, and hyperplastic bone marrow with 3-lineage dysplasia with ~14% of myeloid blasts. Cytogenetic studies with G banding showed normal karyotype. Multiplex ligation-dependent probe amplification (MLPA) screening for most predictive cytogenetic abnormalities of MDS showed loss of the Y chromosome. Those findings later were confirmed with Quantitative Fluorescent (QF)-PCR and specific MLPA for Y chromosome, showing loss of the Y chromosome in >80% of cells. He was diagnosed with MDS-RAEB2 according to 2008 WHO classification and stratified into high risk group (IPSS score 5). Unrelated allogeneic SCT was planed and bridging treatment with Azacitidine at a dose of 75mg/m2/daily subcutaneously for 7 days every 28 days was initiated. Hematologic improvements, according to the International Working Group 2006 criteria, were observed after 4 cycles of Azacitidine treatment. After 6 cycles, complete hematological remission was achieved. Interestingly, molecular analysis performed after the 8th cycle showed normal presence of Y chromosome indicating a cytogenetic remission, molecularly confirmed. Maintenance treatment with Azacitidine was assigned, and the scheduled SCT was postponed. Experience from our case showed that the loss of the Y chromosome was related to the disease onset, and indicated that Azacitidine might be consider as effective treatment for MDS cases associated with good cytogenetic.


Subject(s)
Anemia, Refractory, with Excess of Blasts/drug therapy , Azacitidine/administration & dosage , Chromosomes, Human, Y , Cytogenetic Analysis , DNA Modification Methylases/antagonists & inhibitors , Enzyme Inhibitors/administration & dosage , Myelodysplastic Syndromes/drug therapy , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/genetics , Bone Marrow Examination , Drug Administration Schedule , Humans , Male , Middle Aged , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Predictive Value of Tests , Remission Induction , Treatment Outcome
14.
Hematology Am Soc Hematol Educ Program ; 2016(1): 598-604, 2016 Dec 02.
Article in English | MEDLINE | ID: mdl-27913534

ABSTRACT

Myelodysplastic syndrome (MDS) and myeloproliferative disorders are rare in children; they are divided into low-grade MDS (refractory cytopenia of childhood [RCC]), advanced MDS (refractory anemia with excess blasts in transformation), and juvenile myelomonocytic leukemia (JMML), each with different characteristics and management strategies. Underlying genetic predisposition is recognized in an increasing number of patients. Germ line GATA2 mutation is found in 70% of adolescents with MDS and monosomy 7. It is challenging to distinguish RCC from aplastic anemia, inherited bone marrow failure, and reactive conditions. RCC is often hypoplastic and may respond to immunosuppressive therapy. In case of immunosuppressive therapy failure, hypercellular RCC, or RCC with monosomy 7, hematopoietic stem cell transplantation (HSCT) using reduced-intensity conditioning regimens is indicated. Almost all patients with refractory anemia with excess blasts are candidates for HSCT; children age 12 years or older have a higher risk of treatment-related death, and the conditioning regimens should be adjusted accordingly. Unraveling the genetics of JMML has demonstrated that JMML in patients with germ line PTPN11 and CBL mutations often regresses spontaneously, and therapy is seldom indicated. Conversely, patients with JMML and neurofibromatosis type 1, somatic PTPN11, KRAS, and most of those with NRAS mutations have a rapidly progressive disease, and early HSCT is indicated. The risk of relapse after HSCT is high, and prophylaxis for graft-versus-host disease and monitoring should be adapted to this risk.


Subject(s)
Anemia, Aplastic , Anemia, Refractory, with Excess of Blasts , Leukemia, Myelomonocytic, Juvenile , Adolescent , Anemia, Aplastic/diagnosis , Anemia, Aplastic/genetics , Anemia, Aplastic/immunology , Anemia, Aplastic/therapy , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/immunology , Anemia, Refractory, with Excess of Blasts/therapy , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 7/genetics , Chromosomes, Human, Pair 7/immunology , Female , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/immunology , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/immunology , Humans , Immunosuppression Therapy/methods , Infant , Leukemia, Myelomonocytic, Juvenile/diagnosis , Leukemia, Myelomonocytic, Juvenile/genetics , Leukemia, Myelomonocytic, Juvenile/immunology , Leukemia, Myelomonocytic, Juvenile/therapy , Male , Membrane Proteins/genetics , Membrane Proteins/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/immunology , Proto-Oncogene Proteins c-cbl/genetics , Proto-Oncogene Proteins c-cbl/immunology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology
16.
Am J Hematol ; 91(5): 492-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26874914

ABSTRACT

Refractory anemia with ring sideroblasts and thrombocytosis (RARS-T) shares overlapping features of myelodysplastic syndromes (MDS) and myeloproliferative neoplasms (MPN). RARS-T is characterized by SF3B1 and JAK2 mutations and prognosis is considered to be better than MDS but not as good as MPN. The objective of the study was to identify predictors of survival in RARS-T. We analyzed clinical and laboratory variables in 82 patients and applied a 27-gene NGS assay to 48 marrow samples obtained at diagnosis. 94% of patients had ≥1 mutations; common mutations being: SF3B1 85%, JAK2V617F 33%, ASXL1 29%, DNMT3A 13%, SETBP1 13% and TET2 10%. In a multivariable survival analysis (n = 82), anemia (P = 0.02) [HB< 10 gm/dl: HR 2.3, 95% CI 1.2-4.6] and abnormal karyotype (P =.01) [HR 6.1, 95% CI 2.7-13.8] were independently prognostic for inferior survival. In patients with NGS information (n = 48), univariate analysis showed association between poor survival and presence of SETBP1 (P = 0.04) or ASXL1 (P = 0.08) mutations whereas the absence of these mutations (ASXL1wt/SETBP1wt) was favorable (P = 0.04); the number of concurrent mutations did not provide additional prognostication (P = 0.3). We developed a HR-weighted prognostic model, with 2 points for an abnormal karyotype, 1 point for either ASXL1 and/or SETBP1 mutations, and 1 point for a HB level < 10 gm/dl, which effectively stratified patients into three risk categories; low (0 points), intermediate (1 point) and high (≥2 points), with median survivals of 80, 42 and 11 months respectively (P = 0.01). In summary, we confirm the unique mutational landscape in RARS-T and provide a novel mutation-enhanced prognostic model.


Subject(s)
Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Sideroblastic/mortality , Codon, Nonsense , DNA Mutational Analysis/methods , Frameshift Mutation , Sequence Analysis, DNA/methods , Thrombocytosis/etiology , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/complications , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Sideroblastic/complications , Anemia, Sideroblastic/genetics , Bone Marrow/chemistry , Bone Marrow/pathology , Carrier Proteins/genetics , Chromosome Aberrations , Disease Progression , Female , Hemoglobins/analysis , Humans , Janus Kinase 2/genetics , Kaplan-Meier Estimate , Karyotyping , Male , Middle Aged , Nuclear Proteins/genetics , Phosphoproteins/genetics , Prognosis , Proportional Hazards Models , RNA Splicing Factors , Repressor Proteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Thrombocytosis/genetics
19.
Leukemia ; 29(5): 1092-103, 2015 May.
Article in English | MEDLINE | ID: mdl-25428262

ABSTRACT

The splicing factor SF3B1 is the most commonly mutated gene in the myelodysplastic syndrome (MDS), particularly in patients with refractory anemia with ring sideroblasts (RARS). We investigated the functional effects of SF3B1 disruption in myeloid cell lines: SF3B1 knockdown resulted in growth inhibition, cell cycle arrest and impaired erythroid differentiation and deregulation of many genes and pathways, including cell cycle regulation and RNA processing. MDS is a disorder of the hematopoietic stem cell and we thus studied the transcriptome of CD34(+) cells from MDS patients with SF3B1 mutations using RNA sequencing. Genes significantly differentially expressed at the transcript and/or exon level in SF3B1 mutant compared with wild-type cases include genes that are involved in MDS pathogenesis (ASXL1 and CBL), iron homeostasis and mitochondrial metabolism (ALAS2, ABCB7 and SLC25A37) and RNA splicing/processing (PRPF8 and HNRNPD). Many genes regulated by a DNA damage-induced BRCA1-BCLAF1-SF3B1 protein complex showed differential expression/splicing in SF3B1 mutant cases. This is the first study to determine the target genes of SF3B1 mutation in MDS CD34(+) cells. Our data indicate that SF3B1 has a critical role in MDS by affecting the expression and splicing of genes involved in specific cellular processes/pathways, many of which are relevant to the known RARS pathophysiology, suggesting a causal link.


Subject(s)
Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/cytology , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Stem Cells/cytology , Alternative Splicing , Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Refractory, with Excess of Blasts/metabolism , Antigens, CD34/metabolism , Cell Cycle , Cell Differentiation , Cell Proliferation , Exons , Female , Gene Expression Profiling , Gene Knockdown Techniques , Heterozygote , Homeostasis , Humans , K562 Cells , Male , Mutation , Myelodysplastic Syndromes/metabolism , Phosphoproteins/metabolism , Point Mutation , RNA/genetics , RNA Splicing , RNA Splicing Factors , Ribonucleoprotein, U2 Small Nuclear/metabolism , Sequence Analysis, RNA
20.
Eur J Haematol ; 94(5): 413-8, 2015 May.
Article in English | MEDLINE | ID: mdl-25200248

ABSTRACT

TET2, a member of the ten-eleven-translocation (TET) family genes that modify DNA by converting 5-methylcytosine (5-mC) to 5-hydroxymethylcytosine (5-hmC), is located in chromosome 4q24 and is frequently mutated in myeloid malignancies. The impact of TET2 mutation on survival outcomes is still controversial; however, functional studies have proved that it is a loss-of-function mutation that impairs myeloid cell differentiation and contributes to the phenotype of myeloid neoplasia. We, herein, aimed to investigate TET2 expression in patients with myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). A significantly decreased TET2 expression was observed in bone marrow cells from AML (n = 53) and patients with MDS (n = 64), compared to normal donors (n = 22). In MDS, TET2 expression was significantly reduced in RAEB-1/RAEB-2 compared to other WHO 2008 classifications, and a lower TET2 expression was observed at the time of MDS disease progression in four of five patients. In multivariate analysis, low TET2 expression (P = 0.03), male gender (P = 0.02), and WHO 2008 classification (P < 0.0001) were independent predictors of poorer overall survival. These results suggest that defective TET2 expression plays a role in the MDS pathophysiology and predicts survival outcomes in this disease.


Subject(s)
Anemia, Refractory, with Excess of Blasts/genetics , Anemia, Sideroblastic/genetics , DNA-Binding Proteins/genetics , Leukemia, Myeloid, Acute/genetics , Proto-Oncogene Proteins/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Refractory, with Excess of Blasts/diagnosis , Anemia, Refractory, with Excess of Blasts/mortality , Anemia, Refractory, with Excess of Blasts/pathology , Anemia, Sideroblastic/diagnosis , Anemia, Sideroblastic/mortality , Anemia, Sideroblastic/pathology , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Case-Control Studies , Chromosomes, Human, Pair 4 , Dioxygenases , Down-Regulation , Female , Gene Expression , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Multivariate Analysis , Mutation , Prognosis , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...