Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 83
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Immunity ; 55(8): 1386-1401.e10, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35931086

ABSTRACT

Deleterious somatic mutations in DNA methyltransferase 3 alpha (DNMT3A) and TET mehtylcytosine dioxygenase 2 (TET2) are associated with clonal expansion of hematopoietic cells and higher risk of cardiovascular disease (CVD). Here, we investigated roles of DNMT3A and TET2 in normal human monocyte-derived macrophages (MDM), in MDM isolated from individuals with DNMT3A or TET2 mutations, and in macrophages isolated from human atherosclerotic plaques. We found that loss of function of DNMT3A or TET2 resulted in a type I interferon response due to impaired mitochondrial DNA integrity and activation of cGAS signaling. DNMT3A and TET2 normally maintained mitochondrial DNA integrity by regulating the expression of transcription factor A mitochondria (TFAM) dependent on their interactions with RBPJ and ZNF143 at regulatory regions of the TFAM gene. These findings suggest that targeting the cGAS-type I IFN pathway may have therapeutic value in reducing risk of CVD in patients with DNMT3A or TET2 mutations.


Subject(s)
Cardiovascular Diseases , DNA Methyltransferase 3A/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases/metabolism , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , DNA-Binding Proteins/genetics , Dioxygenases/genetics , Humans , Interferons/metabolism , Macrophages/metabolism , Mitochondria/genetics , Mutation/genetics , Nucleotidyltransferases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism
2.
Blood ; 140(21): 2228-2247, 2022 11 24.
Article in English | MEDLINE | ID: mdl-36130297

ABSTRACT

Myeloid neoplasms and acute leukemias derive from the clonal expansion of hematopoietic cells driven by somatic gene mutations. Although assessment of morphology plays a crucial role in the diagnostic evaluation of patients with these malignancies, genomic characterization has become increasingly important for accurate diagnosis, risk assessment, and therapeutic decision making. Conventional cytogenetics, a comprehensive and unbiased method for assessing chromosomal abnormalities, has been the mainstay of genomic testing over the past several decades and remains relevant today. However, more recent advances in sequencing technology have increased our ability to detect somatic mutations through the use of targeted gene panels, whole-exome sequencing, whole-genome sequencing, and whole-transcriptome sequencing or RNA sequencing. In patients with myeloid neoplasms, whole-genome sequencing represents a potential replacement for both conventional cytogenetic and sequencing approaches, providing rapid and accurate comprehensive genomic profiling. DNA sequencing methods are used not only for detecting somatically acquired gene mutations but also for identifying germline gene mutations associated with inherited predisposition to hematologic neoplasms. The 2022 International Consensus Classification of myeloid neoplasms and acute leukemias makes extensive use of genomic data. The aim of this report is to help physicians and laboratorians implement genomic testing for diagnosis, risk stratification, and clinical decision making and illustrates the potential of genomic profiling for enabling personalized medicine in patients with hematologic neoplasms.


Subject(s)
Hematologic Neoplasms , Leukemia, Myeloid, Acute , Myeloproliferative Disorders , Neoplasms , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Mutation , Genomics , Neoplasms/genetics , Hematologic Neoplasms/genetics , Clinical Decision-Making
3.
Blood ; 136(2): 157-170, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32347921

ABSTRACT

The 2016 revision of the World Health Organization classification of tumors of hematopoietic and lymphoid tissues is characterized by a closer integration of morphology and molecular genetics. Notwithstanding, the myelodysplastic syndrome (MDS) with isolated del(5q) remains so far the only MDS subtype defined by a genetic abnormality. Approximately half of MDS patients carry somatic mutations in spliceosome genes, with SF3B1 being the most commonly mutated one. SF3B1 mutation identifies a condition characterized by ring sideroblasts (RS), ineffective erythropoiesis, and indolent clinical course. A large body of evidence supports recognition of SF3B1-mutant MDS as a distinct nosologic entity. To further validate this notion, we interrogated the data set of the International Working Group for the Prognosis of MDS (IWG-PM). Based on the findings of our analyses, we propose the following diagnostic criteria for SF3B1-mutant MDS: (1) cytopenia as defined by standard hematologic values, (2) somatic SF3B1 mutation, (3) morphologic dysplasia (with or without RS), and (4) bone marrow blasts <5% and peripheral blood blasts <1%. Selected concomitant genetic lesions represent exclusion criteria for the proposed entity. In patients with clonal cytopenia of undetermined significance, SF3B1 mutation is almost invariably associated with subsequent development of overt MDS with RS, suggesting that this genetic lesion might provide presumptive evidence of MDS in the setting of persistent unexplained cytopenia. Diagnosis of SF3B1-mutant MDS has considerable clinical implications in terms of risk stratification and therapeutic decision making. In fact, this condition has a relatively good prognosis and may respond to luspatercept with abolishment of the transfusion requirement.


Subject(s)
Bone Marrow/metabolism , Erythropoiesis , Mutation , Myelodysplastic Syndromes , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Humans , Myelodysplastic Syndromes/classification , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/therapy , Prognosis , Risk Assessment
4.
Hematol Oncol ; 40(4): 491-504, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35368098

ABSTRACT

Despite much of the past 2 years being engulfed by the devastating consequences of the SAR-CoV-2 pandemic, significant progress, even breathtaking, occurred in the field of chronic myeloid malignancies. Some of this was show-cased at the 15th Post-American Society of Hematology (ASH) and the 25th John Goldman workshops on myeloproliferative neoplasms (MPN) held on 9th-10th December 2020 and 7th-10th October 2021, respectively. The inaugural Post-ASH MPN workshop was set out in 2006 by John Goldman (deceased) and Tariq Mughal to answer emerging translational hematology and therapeutics of patients with these malignancies. Rather than present a resume of the discussions, this perspective focuses on some of the pivotal translational hematology and therapeutic insights in these diseases.


Subject(s)
COVID-19 , Graft vs Host Disease , Hematology , Myeloproliferative Disorders , Hematopoietic Stem Cells , Humans , Myeloproliferative Disorders/drug therapy
5.
Blood ; 133(10): 1086-1095, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30670443

ABSTRACT

Myelodysplastic syndromes (MDS) are clonal diseases defined by clinical, morphologic, and genetic features often shared by related myeloid disorders. The diagnostic boundaries between these diseases can be arbitrary and not necessarily reflective of underlying disease biology or outcomes. In practice, measures that distinguish MDS from related disorders may be difficult to quantify and can vary as disease progression occurs. Patients may harbor findings that are not consistent with a single diagnostic category. Several overlap disorders have been formally described, such as the myelodysplastic/myeloproliferative neoplasms (MDS/MPNs). These disorders are characterized by hematopoietic dysplasia with increased proliferation of monocytes, neutrophils, or platelets. They may have mutational profiles that distinguish them from the disorders they resemble and reflect important differences in pathophysiology. MDS also shares diagnostic borders with other diseases. For example, aplastic anemia and hypoplastic MDS can be difficult to distinguish in patients with pancytopenia and bone marrow hypocellularity. Genetic features may help in this regard, because they can identify differences in prognosis and risk of progression. The boundary between MDS and secondary acute myeloid leukemia (sAML) is arbitrarily defined and has been redefined over the years. Genetic studies have demonstrated that sAML clones can precede clinical progression from MDS by many months, suggesting that MDS with excess blasts could be viewed as an overlap between a dysplastic bone marrow failure syndrome and an oligoblastic leukemia. This review will describe the diagnostic boundaries between MDS, MDS/MPNs, sAML, clonal hematopoiesis of indeterminate potential, clonal cytopenia of undetermined significance, and aplastic anemia and how genetic approaches may help to better define them.


Subject(s)
Anemia, Aplastic/diagnosis , Hematology/standards , Myelodysplastic Syndromes/diagnosis , Myeloproliferative Disorders/diagnosis , Bone Marrow/pathology , Clinical Trials as Topic , Diagnosis, Differential , Disease Progression , Hematology/methods , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/diagnosis , Mutation , Myelodysplastic-Myeloproliferative Diseases/diagnosis , Pancytopenia/diagnosis , Prognosis , Risk
6.
Blood ; 134(20): 1730-1744, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31434702

ABSTRACT

Heterozygous deletions within chromosome 20q, or del(20q), are frequent cytogenetic abnormalities detected in hematologic malignancies. To date, identification of genes in the del(20q) common deleted region that contribute to disease development have remained elusive. Through assessment of patient gene expression, we have identified STK4 (encoding Hippo kinase MST1) as a 20q gene that is downregulated below haploinsufficient amounts in myelodysplastic syndrome (MDS) and myeloproliferative neoplasm (MPN). Hematopoietic-specific gene inactivation in mice revealed Hippo kinase loss to induce splenomegaly, thrombocytopenia, megakaryocytic dysplasia, and a propensity for chronic granulocytosis; phenotypes that closely resemble those observed in patients harboring del(20q). In a JAK2-V617F model, heterozygous Hippo kinase inactivation led to accelerated development of lethal myelofibrosis, recapitulating adverse MPN disease progression and revealing a novel genetic interaction between these 2 molecular events. Quantitative serum protein profiling showed that myelofibrotic transformation in mice was associated with cooperative effects of JAK2-V617F and Hippo kinase inactivation on innate immune-associated proinflammatory cytokine production, including IL-1ß and IL-6. Mechanistically, MST1 interacted with IRAK1, and shRNA-mediated knockdown was sufficient to increase IRAK1-dependent innate immune activation of NF-κB in human myeloid cells. Consistent with this, treatment with a small molecule IRAK1/4 inhibitor rescued the aberrantly elevated IL-1ß production in the JAK2-V617F MPN model. This study identified Hippo kinase MST1 (STK4) as having a central role in the biology of del(20q)-associated hematologic malignancies and revealed a novel molecular basis of adverse MPN progression that may be therapeutically exploitable via IRAK1 inhibition.


Subject(s)
Hematologic Neoplasms/genetics , Myelodysplastic Syndromes/genetics , Myeloproliferative Disorders/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Chromosome Deletion , Chromosomes, Human, Pair 20/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/immunology , Humans , Immunity, Innate , Intracellular Signaling Peptides and Proteins , Mice , Myelodysplastic Syndromes/immunology , Myeloproliferative Disorders/immunology , Protein Serine-Threonine Kinases/immunology
7.
Cytotherapy ; 23(3): 236-241, 2021 03.
Article in English | MEDLINE | ID: mdl-33279399

ABSTRACT

BACKGROUND: Myelodysplastic syndromes (MDS) represent the most common type of acquired bone marrow failure in adults and is characterized by ineffective maturation of myeloid precursor cells and peripheral cytopenias associated with higher rates of infection, bleeding and transfusion dependence. In higher-risk patients with MDS who relapse or do not respond after standard hypomethylating agent (HMA) therapy, the 2-year survival rate is 15%. METHODS: Here the authors report the feasibility and safety of a novel experimental T-cell therapy called personalized adoptive cell therapy, which selects, immunizes and expands T cells against MDS-specific mutations and is targeted to patient-specific tumor cell neoantigens. Somatic mutations serve as the pathogenic drivers of cancer, including MDS, as these transformative genetic mutations may generate novel immunogenic proteins (i.e., neopeptides and possible neoantigens) that may be targeted therapeutically. RESULTS: The authors demonstrate that the adaptive immune system can be trained ex vivo to recognize neopeptides as neoantigens and that the infusion of culture-expanded, neoantigen-immunized autologous T cells has been feasible and safe in the three patients treated to date. DISCUSSION: The authors report on early results from their first-in-human phase 1 clinical trial that aims to assess the safety and tolerability of this novel form of adoptive T-cell immunotherapy for HMA-refractory patients with higher-risk MDS.


Subject(s)
Myelodysplastic Syndromes , Neoplasm Recurrence, Local , Aged , Cell- and Tissue-Based Therapy , Humans , Immunotherapy, Adoptive , Myelodysplastic Syndromes/therapy , T-Lymphocytes
8.
Cytotherapy ; 23(4): 320-328, 2021 04.
Article in English | MEDLINE | ID: mdl-33262074

ABSTRACT

Therapies that utilize immune checkpoint inhibition work by leveraging mutation-derived neoantigens and have shown greater clinical efficacy in tumors with higher mutational burden. Whether tumors with a low mutational burden are susceptible to neoantigen-targeted therapy has not been fully addressed. To examine the feasibility of neoantigen-specific adoptive T-cell therapy, the authors studied the T-cell response against somatic variants in five patients with myelodysplastic syndrome (MDS), a malignancy with a very low tumor mutational burden. DNA and RNA from tumor (CD34+) and normal (CD3+) cells isolated from the patients' blood were sequenced to predict patient-specific MDS neopeptides. Neopeptides representing the somatic variants were used to induce and expand autologous T cells ex vivo, and these were systematically tested in killing assays to determine the proportion of neopeptides yielding neoantigen-specific T cells. The authors identified a total of 32 somatic variants (four to eight per patient) and found that 21 (66%) induced a peptide-specific T-cell response and 19 (59%) induced a T-cell response capable of killing autologous tumor cells. Of the 32 somatic variants, 11 (34%) induced a CD4+ response and 11 (34%) induced a CD8+ response that killed the tumor. These results indicate that in vitro induction of neoantigen-specific T cells is feasible for tumors with very low mutational burden and that this approach warrants investigation as a therapeutic option for such patients.


Subject(s)
Myelodysplastic Syndromes , Neoplasms , Antigens, Neoplasm/genetics , Humans , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/therapy , T-Lymphocytes
10.
J Natl Compr Canc Netw ; 17(11.5): 1444-1447, 2019 11.
Article in English | MEDLINE | ID: mdl-31766023

ABSTRACT

Often unrecognized and underdiagnosed, myelodysplastic syndromes (MDS) are a rare group of cancers in which the bone marrow fails to produce sufficient healthy blood cells. Although patients with lower-risk MDS can live for >5 years, those with high-risk disease that evolves into acute myeloid leukemia is associated with significantly lower overall survival. At the NCCN 2019 Annual Congress: Hematologic Malignancies, Dr. Rafael Bejar summarized current standard treatment options for patients with MDS and discussed the importance of genetic testing to identify mutations that may impact treatment. Finally, Dr. Bejar described emerging personalized treatment strategies for the management of this disease.


Subject(s)
Myelodysplastic Syndromes/therapy , Precision Medicine/methods , Aged , Humans , Male
11.
Proc Natl Acad Sci U S A ; 113(39): 10750-8, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27638202

ABSTRACT

Prevention is an essential component of cancer eradication. Next-generation sequencing of cancer genomes and epigenomes has defined large numbers of driver mutations and molecular subgroups, leading to therapeutic advances. By comparison, there is a relative paucity of such knowledge in premalignant neoplasia, which inherently limits the potential to develop precision prevention strategies. Studies on the interplay between germ-line and somatic events have elucidated genetic processes underlying premalignant progression and preventive targets. Emerging data hint at the immune system's ability to intercept premalignancy and prevent cancer. Genetically engineered mouse models have identified mechanisms by which genetic drivers and other somatic alterations recruit inflammatory cells and induce changes in normal cells to create and interact with the premalignant tumor microenvironment to promote oncogenesis and immune evasion. These studies are currently limited to only a few lesion types and patients. In this Perspective, we advocate a large-scale collaborative effort to systematically map the biology of premalignancy and the surrounding cellular response. By bringing together scientists from diverse disciplines (e.g., biochemistry, omics, and computational biology; microbiology, immunology, and medical genetics; engineering, imaging, and synthetic chemistry; and implementation science), we can drive a concerted effort focused on cancer vaccines to reprogram the immune response to prevent, detect, and reject premalignancy. Lynch syndrome, clonal hematopoiesis, and cervical intraepithelial neoplasia which also serve as models for inherited syndromes, blood, and viral premalignancies, are ideal scenarios in which to launch this initiative.


Subject(s)
Neoplasms/immunology , Neoplasms/prevention & control , Precancerous Conditions/pathology , Germ Cells/metabolism , Humans , Immune System/pathology , Models, Biological , Neoplasm Proteins/metabolism , Tumor Microenvironment
12.
RNA ; 22(10): 1535-49, 2016 10.
Article in English | MEDLINE | ID: mdl-27492256

ABSTRACT

Myelodysplastic syndromes (MDS) are heterogeneous myeloid disorders with prevalent mutations in several splicing factors, but the splicing programs linked to specific mutations or MDS in general remain to be systematically defined. We applied RASL-seq, a sensitive and cost-effective platform, to interrogate 5502 annotated splicing events in 169 samples from MDS patients or healthy individuals. We found that splicing signatures associated with normal hematopoietic lineages are largely related to cell signaling and differentiation programs, whereas MDS-linked signatures are primarily involved in cell cycle control and DNA damage responses. Despite the shared roles of affected splicing factors in the 3' splice site definition, mutations in U2AF1, SRSF2, and SF3B1 affect divergent splicing programs, and interestingly, the affected genes fall into converging cancer-related pathways. A risk score derived from 11 splicing events appears to be independently associated with an MDS prognosis and AML transformation, suggesting potential clinical relevance of altered splicing patterns in MDS.


Subject(s)
Mutation , Myelodysplastic Syndromes/genetics , Phosphoproteins/genetics , RNA Splice Sites , RNA Splicing Factors/genetics , Serine-Arginine Splicing Factors/genetics , Splicing Factor U2AF/genetics , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Humans , Male , Middle Aged , Myelodysplastic Syndromes/pathology , RNA Splicing
13.
Curr Opin Hematol ; 24(2): 73-78, 2017 03.
Article in English | MEDLINE | ID: mdl-27875374

ABSTRACT

PURPOSE OF REVIEW: Myelodysplastic syndromes (MDS) have remarkably diverse somatic mutation patterns that can be challenging to interpret clinically. Yet, genetic information is increasingly available to physicians. This review will examine several implications of genetic diversity in MDS. RECENT FINDINGS: Somatic mutations can serve as clinically relevant biomarkers in MDS. Molecular subtypes may exist that share clinical features including risk of progression to acute myeloid leukemia, response to treatment, and overall survival. Several mutated genes are known to have prognostic value that is independent of common risk stratification tools. Mutations of several genes identify low-blast percentage patients with greater than predicted disease risk while only SF3B1 mutations predict lower disease risk than expected. Mutations of TP53 are associated with adverse features, yet demonstrate inferior outcomes than predicted by these risk factors. SF3B1 and TP53 mutations may identify clinically relevant subtypes of MDS and allow for better refinement of risk within these groups. Using somatic mutations to diagnose MDS is more challenging because they can occur in healthy individuals. Yet, patients with unexplained cytopenias have a high rate of clonal hematopoiesis that may be an important risk factor to identify clinically. SUMMARY: Patterns of somatic mutations are diverse in MDS, but can inform the prediction of prognosis and aid in its diagnosis.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Myelodysplastic Syndromes/genetics , Biomarkers , Genetic Association Studies , Humans , Mutation , Mutation Rate , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/mortality , Prognosis
14.
Blood ; 126(1): 9-16, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-25931582

ABSTRACT

Recent genetic analyses of large populations have revealed that somatic mutations in hematopoietic cells leading to clonal expansion are commonly acquired during human aging. Clonally restricted hematopoiesis is associated with an increased risk of subsequent diagnosis of myeloid or lymphoid neoplasia and increased all-cause mortality. Although myelodysplastic syndromes (MDS) are defined by cytopenias, dysplastic morphology of blood and marrow cells, and clonal hematopoiesis, most individuals who acquire clonal hematopoiesis during aging will never develop MDS. Therefore, acquisition of somatic mutations that drive clonal expansion in the absence of cytopenias and dysplastic hematopoiesis can be considered clonal hematopoiesis of indeterminate potential (CHIP), analogous to monoclonal gammopathy of undetermined significance and monoclonal B-cell lymphocytosis, which are precursor states for hematologic neoplasms but are usually benign and do not progress. Because mutations are frequently observed in healthy older persons, detection of an MDS-associated somatic mutation in a cytopenic patient without other evidence of MDS may cause diagnostic uncertainty. Here we discuss the nature and prevalence of CHIP, distinction of this state from MDS, and current areas of uncertainty regarding diagnostic criteria for myeloid malignancies.


Subject(s)
Clonal Evolution , Hematologic Neoplasms/diagnosis , Hematopoiesis , Myelodysplastic Syndromes/diagnosis , Diagnosis, Differential , Hematologic Neoplasms/classification , Hematopoiesis/genetics , Humans , Monoclonal Gammopathy of Undetermined Significance/diagnosis , Monoclonal Gammopathy of Undetermined Significance/pathology , Mutation , Myelodysplastic Syndromes/classification , Precancerous Conditions
15.
Blood ; 126(21): 2355-61, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26429975

ABSTRACT

Establishing a diagnosis in patients suspected of having a myelodysplastic syndrome (MDS) can be challenging and could be informed by the identification of somatic mutations. We performed a prospective study to examine the frequency and types of mutations encountered in 144 patients with unexplained cytopenias. Based on bone marrow findings, 17% were diagnosed with MDS, 15% with idiopathic cytopenias of undetermined significance (ICUS) and some evidence of dysplasia, and 69% with ICUS and no dysplasia. Bone marrow DNA was sequenced for mutations in 22 frequently mutated myeloid malignancy genes. Somatic mutations were identified in 71% of MDS patients, 62% of patients with ICUS and some dysplasia, and 20% of ICUS patients and no dysplasia. In total, 35% of ICUS patients carried a somatic mutation or chromosomal abnormality indicative of clonal hematopoiesis. We validated these results in a cohort of 91 lower-risk MDS and 249 ICUS cases identified over a 6-month interval. Mutations were found in 79% of those with MDS, in 45% of those with ICUS with dysplasia, and in 17% of those with ICUS without dysplasia. The spectrum of mutated genes was similar with the exception of SF3B1 which was rarely mutated in patients without dysplasia. Variant allele fractions were comparable between clonal ICUS (CCUS) and MDS as were mean age and blood counts. We demonstrate that CCUS is a more frequent diagnosis than MDS in cytopenic patients. Clinical and mutational features are similar in these groups and may have diagnostic utility once outcomes in CCUS patients are better understood.


Subject(s)
Alleles , Chromosome Aberrations , Gene Frequency , Hematopoiesis/genetics , Mutation , Myelodysplastic Syndromes , Age Factors , Female , Humans , Male , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , Prospective Studies , Retrospective Studies
16.
J Natl Compr Canc Netw ; 15(1): 60-87, 2017 01.
Article in English | MEDLINE | ID: mdl-28040720

ABSTRACT

The myelodysplastic syndromes (MDS) comprise a heterogenous group of myeloid disorders with a highly variable disease course. Diagnostic criteria to better stratify patients with MDS continue to evolve, based on morphology, cytogenetics, and the presence of cytopenias. More accurate classification of patients will allow for better treatment guidance. Treatment encompasses supportive care, treatment of anemia, low-intensity therapy, and high-intensity therapy. This portion of the guidelines focuses on diagnostic classification, molecular abnormalities, therapeutic options, and recommended treatment approaches.


Subject(s)
Anemia/drug therapy , Hematinics/therapeutic use , Hematopoietic Stem Cell Transplantation , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Anemia/etiology , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Humans , Immunologic Factors/therapeutic use , Induction Chemotherapy/methods , Medical Oncology/standards , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Survival Rate
17.
Blood ; 124(18): 2793-803, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25237199

ABSTRACT

Once thought to be rare disorders, the myelodysplastic syndromes (MDS) are now recognized as among the most common hematological neoplasms, probably affecting >30 000 patients per year in the United States. US regulatory approval of azacitidine, decitabine, and lenalidomide between 2004 and 2006 seemed to herald a new era in the development of disease-modifying therapies for MDS, but there have been no further drug approvals for MDS indications in the United States in the last 8 years. The available drugs are not curative, and few of the compounds that are currently in development are likely to be approved in the near future. As a result, MDS diagnoses continue to place a heavy burden on both patients and health care systems. Incomplete understanding of disease pathology, the inherent biological complexity of MDS, and the presence of comorbid conditions and poor performance status in the typical older patient with MDS have been major impediments to development of effective novel therapies. Here we discuss new insights from genomic discoveries that are illuminating MDS pathogenesis, increasing diagnostic accuracy, and refining prognostic assessment, and which will one day contribute to more effective treatments and improved patient outcomes.


Subject(s)
Myelodysplastic Syndromes/genetics , Biomarkers/metabolism , Genetic Predisposition to Disease , Humans , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/epidemiology , Myelodysplastic Syndromes/therapy , Prognosis
18.
Blood ; 124(17): 2705-12, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25224413

ABSTRACT

Only a minority of myelodysplastic syndrome (MDS) patients respond to hypomethylating agents (HMAs), but strong predictors of response are unknown. We sequenced 40 recurrently mutated myeloid malignancy genes in tumor DNA from 213 MDS patients collected before treatment with azacitidine (AZA) or decitabine (DEC). Mutations were examined for association with response and overall survival. The overall response rate of 47% was not different between agents. Clonal TET2 mutations predicted response (odds ratio [OR] 1.99, P = .036) when subclones unlikely to be detected by Sanger sequencing (allele fraction <10%) were treated as wild-type (WT). Response rates were highest in the subset of TET2 mutant patients without clonal ASXL1 mutations (OR 3.65, P = .009). Mutations of TP53 (hazard ratio [HR] 2.01, P = .002) and PTPN11 (HR 3.26, P = .006) were associated with shorter overall survival but not drug response. Murine-competitive bone marrow transplantation followed by treatment with AZA demonstrated that Tet2-null cells have an engraftment advantage over Tet2-WT cells. AZA significantly decreased this advantage for Tet2-null cells (P = .002) but not Tet2-WT cells (P = .212). Overall, Tet2 loss appears to sensitize cells to treatment with AZA in vivo, and TET2 mutations can identify patients more likely to respond to HMAs.


Subject(s)
DNA Methylation/drug effects , DNA-Binding Proteins/genetics , Mutation , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Proto-Oncogene Proteins/genetics , Aged , Animals , Azacitidine/analogs & derivatives , Azacitidine/therapeutic use , Biomarkers, Tumor/genetics , Bone Marrow Transplantation/methods , DNA Modification Methylases/antagonists & inhibitors , Decitabine , Dioxygenases , Enzyme Inhibitors/therapeutic use , Female , Gene Frequency , Genotype , Humans , Kaplan-Meier Estimate , Logistic Models , Male , Mice, Knockout , Repressor Proteins/genetics , Transplantation Chimera/genetics , Treatment Outcome , Tumor Suppressor Protein p53/genetics
19.
BMC Cancer ; 16: 652, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27538433

ABSTRACT

BACKGROUND: Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are myeloid neoplasms in which outgrowth of neoplastic clones disrupts normal hematopoiesis. Some patients with unexplained persistent cytopenias may not meet minimal diagnostic criteria for MDS but an alternate diagnosis is not apparent; the term idiopathic cytopenia of undetermined significance (ICUS) has been used to describe this state. MDS and AML occur primarily in older patients who are often treated outside the clinical trial setting. Consequently, our understanding of the patterns of diagnostic evaluation, management, and outcomes of these patients is limited. Furthermore, there are few natural history studies of ICUS. To better understand how patients who have MDS, ICUS, or AML are managed in the routine clinical setting, the Connect MDS/AML Disease Registry, a multicenter, prospective, observational cohort study of patients newly diagnosed with these conditions has been initiated. METHODS/DESIGN: The Connect MDS/AML Disease Registry will capture diagnosis, risk assessment, treatment, and outcomes data for approximately 1500 newly diagnosed patients from approximately 150 community and academic sites in the United States in 4 cohorts: (1) lower-risk MDS (International Prognostic Scoring System [IPSS] low and intermediate-1 risk), with and without del(5q); (2) higher-risk MDS (IPSS intermediate-2 and high risk); (3) ICUS; and (4) AML in patients aged ≥ 55 years (excluding acute promyelocytic leukemia). Diagnosis will be confirmed by central review. Baseline patient characteristics, diagnostic patterns, treatment patterns, clinical outcomes, health economics outcomes, and patient-reported health-related quality of life will be entered into an electronic data capture system at enrollment and quarterly for 8 years. A tissue substudy to explore the relationship between karyotypes, molecular markers, and clinical outcomes will be conducted, and is optional for patients. DISCUSSION: The Connect MDS/AML Disease Registry will be the first prospective, observational, non-interventional study in the United States to collect clinical information, patient-reported outcomes, and tissue samples from patients with MDS, ICUS, or AML receiving multiple therapies. Results from this registry may provide new insights into the relationship between diagnostic practices, treatment regimens, and outcomes in patients with these diseases and identify areas for future investigation. TRIAL REGISTRATION: Connect MDS/AML Disease Registry ( NCT01688011 ). Registered 14 September 2012.


Subject(s)
Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/therapy , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/therapy , Registries , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Prospective Studies , Quality of Life , Sample Size , United States
20.
Adv Exp Med Biol ; 907: 215-28, 2016.
Article in English | MEDLINE | ID: mdl-27256388

ABSTRACT

Many cancers demonstrate aberrant splicing patterns that contribute to their development and progression. Recently, recurrent somatic mutations of genes encoding core subunits of the spliceosome have been identified in several different cancer types. These mutations are most common in hematologic malignancies like the myelodysplastic syndromes (MDS), acute myeloid leukemia, and chronic lymphocytic leukemia, but also in occur in several solid tumors at lower frequency. The most frequent mutations occur in SF3B1, U2AF1, SRSF2, and ZRSR2 and are largely exclusive of each other. Mutations in SF3B1, U2AF1, and SRSF2 acquire heterozygous missense mutations in specific codons, resembling oncogenes. ZRSR2 mutations include clear loss-of-function variants, a pattern more common to tumor suppressor genes. These splicing factors are associated with distinct clinical phenotypes and patterns of mutation in different malignancies. Mutations have both diagnostic and prognostic relevance. Splicing factor mutations appear to affect only a minority of transcripts which show little overlap by mutation type. How differences in splicing caused by somatic mutations of spliceosome subunits lead to oncogenesis is not clear and may involve different targets in each disease type. However, cells with mutated splicing machinery may be particularly vulnerable to further disruption of the spliceosome suggesting a novel strategy for the targeted therapy of cancers.


Subject(s)
Mutation , Neoplasm Proteins/physiology , Neoplasms/genetics , RNA Splicing Factors/genetics , RNA Splicing/genetics , Hematologic Neoplasms/genetics , Humans , Myelodysplastic Syndromes/genetics , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phenotype , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/genetics , Phosphoproteins/physiology , RNA Splicing Factors/antagonists & inhibitors , RNA Splicing Factors/physiology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Ribonucleoproteins/genetics , Ribonucleoproteins/physiology , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/physiology , Splicing Factor U2AF/genetics , Splicing Factor U2AF/physiology
SELECTION OF CITATIONS
SEARCH DETAIL