21 results on '"Viny AD"'
Search Results
2. Molecular predictors of immunophenotypic measurable residual disease clearance in acute myeloid leukemia.
- Author
-
Stahl M, Derkach A, Farnoud N, Bewersdorf JP, Robinson T, Famulare C, Cho C, Devlin S, Menghrajani K, Patel MA, Cai SF, Miles LA, Bowman RL, Geyer MB, Dunbar A, Epstein-Peterson ZD, McGovern E, Schulman J, Glass JL, Taylor J, Viny AD, Stein EM, Getta B, Arcila ME, Gao Q, Barker J, Shaffer BC, Papadopoulos EB, Gyurkocza B, Perales MA, Abdel-Wahab O, Levine RL, Giralt SA, Zhang Y, Xiao W, Pai N, Papaemmanuil E, Tallman MS, Roshal M, and Goldberg AD
- Subjects
- Humans, Prognosis, Stem Cell Transplantation, Remission Induction, Transplantation, Homologous, Neoplasm, Residual genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Measurable residual disease (MRD) is a powerful prognostic factor in acute myeloid leukemia (AML). However, pre-treatment molecular predictors of immunophenotypic MRD clearance remain unclear. We analyzed a dataset of 211 patients with pre-treatment next-generation sequencing who received induction chemotherapy and had MRD assessed by serial immunophenotypic monitoring after induction, subsequent therapy, and allogeneic stem cell transplant (allo-SCT). Induction chemotherapy led to MRD- remission, MRD+ remission, and persistent disease in 35%, 27%, and 38% of patients, respectively. With subsequent therapy, 34% of patients with MRD+ and 26% of patients with persistent disease converted to MRD-. Mutations in CEBPA, NRAS, KRAS, and NPM1 predicted high rates of MRD- remission, while mutations in TP53, SF3B1, ASXL1, and RUNX1 and karyotypic abnormalities including inv (3), monosomy 5 or 7 predicted low rates of MRD- remission. Patients with fewer individual clones were more likely to achieve MRD- remission. Among 132 patients who underwent allo-SCT, outcomes were favorable whether patients achieved early MRD- after induction or later MRD- after subsequent therapy prior to allo-SCT. As MRD conversion with chemotherapy prior to allo-SCT is rarely achieved in patients with specific baseline mutational patterns and high clone numbers, upfront inclusion of these patients into clinical trials should be considered., (© 2022 Wiley Periodicals LLC.)
- Published
- 2023
- Full Text
- View/download PDF
3. Subtype-specific 3D genome alteration in acute myeloid leukaemia.
- Author
-
Xu J, Song F, Lyu H, Kobayashi M, Zhang B, Zhao Z, Hou Y, Wang X, Luan Y, Jia B, Stasiak L, Wong JH, Wang Q, Jin Q, Jin Q, Fu Y, Yang H, Hardison RC, Dovat S, Platanias LC, Diao Y, Yang Y, Yamada T, Viny AD, Levine RL, Claxton D, Broach JR, Zheng H, and Yue F
- Subjects
- Humans, Chromatin genetics, DNA Methylation, Promoter Regions, Genetic, Enhancer Elements, Genetic, Gene Silencing, Reproducibility of Results, CRISPR-Cas Systems, Sequence Analysis, DNA (Cytosine-5-)-Methyltransferases, Gene Expression Regulation, Leukemic, Leukemia, Myeloid, Acute genetics, Genome, Human genetics
- Abstract
Acute myeloid leukaemia (AML) represents a set of heterogeneous myeloid malignancies, and hallmarks include mutations in epigenetic modifiers, transcription factors and kinases
1-5 . The extent to which mutations in AML drive alterations in chromatin 3D structure and contribute to myeloid transformation is unclear. Here we use Hi-C and whole-genome sequencing to analyse 25 samples from patients with AML and 7 samples from healthy donors. Recurrent and subtype-specific alterations in A/B compartments, topologically associating domains and chromatin loops were identified. RNA sequencing, ATAC with sequencing and CUT&Tag for CTCF, H3K27ac and H3K27me3 in the same AML samples also revealed extensive and recurrent AML-specific promoter-enhancer and promoter-silencer loops. We validated the role of repressive loops on their target genes by CRISPR deletion and interference. Structural variation-induced enhancer-hijacking and silencer-hijacking events were further identified in AML samples. Hijacked enhancers play a part in AML cell growth, as demonstrated by CRISPR screening, whereas hijacked silencers have a downregulating role, as evidenced by CRISPR-interference-mediated de-repression. Finally, whole-genome bisulfite sequencing of 20 AML and normal samples revealed the delicate relationship between DNA methylation, CTCF binding and 3D genome structure. Treatment of AML cells with a DNA hypomethylating agent and triple knockdown of DNMT1, DNMT3A and DNMT3B enabled the manipulation of DNA methylation to revert 3D genome organization and gene expression. Overall, this study provides a resource for leukaemia studies and highlights the role of repressive loops and hijacked cis elements in human diseases., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
4. The Impact of Inflammation-Induced Tumor Plasticity during Myeloid Transformation.
- Author
-
Yeaton A, Cayanan G, Loghavi S, Dolgalev I, Leddin EM, Loo CE, Torabifard H, Nicolet D, Wang J, Corrigan K, Paraskevopoulou V, Starczynowski DT, Wang E, Abdel-Wahab O, Viny AD, Stone RM, Byrd JC, Guryanova OA, Kohli RM, Cisneros GA, Tsirigos A, Eisfeld AK, Aifantis I, and Guillamot M
- Subjects
- Animals, Hematopoiesis genetics, Inflammation genetics, Mutation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Myeloproliferative Disorders genetics, Myeloproliferative Disorders pathology
- Abstract
Clonal hematopoiesis (CH) is an aging-associated condition characterized by the clonal outgrowth of mutated preleukemic cells. Individuals with CH are at an increased risk of developing hematopoietic malignancies. Here, we describe a novel animal model carrying a recurrent TET2 missense mutation frequently found in patients with CH and leukemia. In a fashion similar to CH, animals show signs of disease late in life when they develop a wide range of myeloid neoplasms, including acute myeloid leukemia (AML). Using single-cell transcriptomic profiling of the bone marrow, we show that disease progression in aged animals correlates with an enhanced inflammatory response and the emergence of an aberrant inflammatory monocytic cell population. The gene signature characteristic of this inflammatory population is associated with poor prognosis in patients with AML. Our study illustrates an example of collaboration between a genetic lesion found in CH and inflammation, leading to transformation and the establishment of blood neoplasms., Significance: Progression from a preleukemic state to transformation, in the presence of TET2 mutations, is coupled with the emergence of inflammation and a novel population of inflammatory monocytes. Genes characteristic of this inflammatory population are associated with the worst prognosis in patients with AML. These studies connect inflammation to progression to leukemia. See related commentary by Pietras and DeGregori, p. 2234 . This article is highlighted in the In This Issue feature, p. 2221., (©2022 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
5. PU.1-Dependent Enhancer Inhibition Separates Tet2-Deficient Hematopoiesis from Malignant Transformation.
- Author
-
Aivalioti MM, Bartholdy BA, Pradhan K, Bhagat TD, Zintiridou A, Jeong JJ, Thiruthuvanathan VJ, Pujato M, Paranjpe A, Zhang C, Levine RL, Viny AD, Wickrema A, Verma A, and Will B
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Cytosine, Enhancer Elements, Genetic, Hematopoiesis genetics, Humans, Mice, DNA-Binding Proteins genetics, Dioxygenases genetics, Leukemia, Myeloid, Acute genetics, Proto-Oncogene Proteins genetics, Trans-Activators genetics
- Abstract
Cytosine hypermethylation in and around DNA-binding sites of master transcription factors, including PU.1, occurs in aging hematopoietic stem cells following acquired loss-of-function mutations of DNA methyl-cytosine dioxygenase ten-eleven translocation-2 (TET2), albeit functional relevance has been unclear. We show that Tet2-deficient mouse hematopoietic stem and progenitor cells undergo malignant transformation upon compromised gene regulation through heterozygous deletion of an upstream regulatory region (UREΔ/WT) of the PU.1 gene. Although compatible with multilineage blood formation at young age, Tet2-deficient PU.1 UREΔ/WT mice develop highly penetrant, transplantable acute myeloid leukemia (AML) during aging. Leukemic stem and progenitor cells show hypermethylation at putative PU.1-binding sites, fail to activate myeloid enhancers, and are hallmarked by a signature of genes with impaired expression shared with human AML. Our study demonstrates that Tet2 and PU.1 jointly suppress leukemogenesis and uncovers a methylation-sensitive PU.1-dependent gene network as a unifying molecular vulnerability associated with AML., Significance: We identify moderately impaired PU.1 mRNA expression as a biological modality predisposing Tet2-deficient hematopoietic stem and progenitor cells to malignant transformation. Our study furthermore uncovers a methylation-sensitive PU.1 gene network as a common feature of myeloid leukemia potentially allowing for the identification of patients at risk for malignant transformation. See related commentary by Schleicher and Pietras, p. 378. This article is highlighted in the In This Issue feature, p. 369., (©2022 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
6. Combinatorial genetics reveals the Dock1-Rac2 axis as a potential target for the treatment of NPM1;Cohesin mutated AML.
- Author
-
Meyer AE, Stelloh C, Pulakanti K, Burns R, Fisher JB, Heimbruch KE, Tarima S, Furumo Q, Brennan J, Zheng Y, Viny AD, Vassiliou GS, and Rao S
- Subjects
- Animals, Cell Cycle Proteins, Chromosomal Proteins, Non-Histone, Mice, Mutation, Nucleophosmin, Precision Medicine, Transcription Factors genetics, rac GTP-Binding Proteins, Cohesins, RAC2 GTP-Binding Protein, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
Acute myeloid leukemia (AML) is driven by mutations that occur in numerous combinations. A better understanding of how mutations interact with one another to cause disease is critical to developing targeted therapies. Approximately 50% of patients that harbor a common mutation in NPM1 (NPM1cA) also have a mutation in the cohesin complex. As cohesin and Npm1 are known to regulate gene expression, we sought to determine how cohesin mutation alters the transcriptome in the context of NPM1cA. We utilized inducible Npm1
cAflox/+ and core cohesin subunit Smc3flox/+ mice to examine AML development. While Npm1cA/+ ;Smc3Δ/+ mice developed AML with a similar latency and penetrance as Npm1cA/+ mice, RNA-seq suggests that the Npm1cA/+ ; Smc3Δ/+ mutational combination uniquely alters the transcriptome. We found that the Rac1/2 nucleotide exchange factor Dock1 was specifically upregulated in Npm1cA/+ ;Smc3Δ/+ HSPCs. Knockdown of Dock1 resulted in decreased growth and adhesion and increased apoptosis only in Npm1cA/+ ;Smc3Δ/+ AML. Higher Rac activity was also observed in Npm1cA/+ ;Smc3Δ/+ vs. Npm1cA/+ AMLs. Importantly, the Dock1/Rac pathway is targetable in Npm1cA/+ ;Smc3Δ/+ AMLs. Our results suggest that Dock1/Rac represents a potential target for the treatment of patients harboring NPM1cA and cohesin mutations and supports the use of combinatorial genetics to identify novel precision oncology targets., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
7. DNMT3A-Mutant Leukemia Cells Primed to "Fork It Over" under DNA Damage.
- Author
-
Viny AD
- Subjects
- DNA (Cytosine-5-)-Methyltransferases genetics, DNA Damage, DNA Methyltransferase 3A, Humans, Mutation, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy, Myelodysplastic Syndromes genetics, Myelodysplastic Syndromes therapy
- Abstract
Mutations in the gene DNMT3A have been identified in various hematopoietic conditions, including clonal hematopoiesis, myelodysplastic syndrome, and acute myeloid leukemia. The clinical significance of this early mutation and the resultant enhanced clonal fitness have been a focus for therapeutic intervention. See related article by Venugopal et al., p. 756., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
8. Recurrent Mutations in Cyclin D3 Confer Clinical Resistance to FLT3 Inhibitors in Acute Myeloid Leukemia.
- Author
-
Smith CC, Viny AD, Massi E, Kandoth C, Socci ND, Rapaport F, Najm M, Medina-Martinez JS, Papaemmanuil E, Tarver TC, Hsu HH, Le MH, West B, Bollag G, Taylor BS, Levine RL, and Shah NP
- Subjects
- Cell Line, Tumor, Humans, Aminopyridines therapeutic use, Cyclin D3 genetics, Drug Resistance, Neoplasm genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Mutation, Protein Kinase Inhibitors therapeutic use, Pyrroles therapeutic use, fms-Like Tyrosine Kinase 3 antagonists & inhibitors
- Abstract
Purpose: Biomarkers of response and resistance to FLT3 tyrosine kinase inhibitors (TKI) are still emerging, and optimal clinical combinations remain unclear. The purpose of this study is to identify co-occurring mutations that influence clinical response to the novel FLT3 inhibitor pexidartinib (PLX3397)., Experimental Design: We performed targeted sequencing of pretreatment blasts from 29 patients with FLT3 internal tandem duplication (ITD) mutations treated on the phase I/II trial of pexidartinib in relapsed/refractory FLT3 -ITD+ acute myeloid leukemia (AML). We sequenced 37 samples from 29 patients with available material, including 8 responders and 21 non-responders treated at or above the recommended phase II dose of 3,000 mg., Results: Consistent with other studies, we identified mutations in NRAS, TP53, IDH2 , and a variety of epigenetic and transcriptional regulators only in non-responders. Among the most frequently mutated genes in non-responders was Cyclin D3 ( CCND3) . A total of 3 individual mutations in CCND3 (Q276*, S264R, and T283A) were identified in 2 of 21 non-responders (one patient had both Q276* and S264R). No CCND3 mutations were found in pexidartinib responders. Expression of the Q276* and T283A mutations in FLT3 -ITD MV4;11 cells conferred resistance to apoptosis, decreased cell-cycle arrest, and increased proliferation in the presence of pexidartinib and other FLT3 inhibitors. Inhibition of CDK4/6 activity in CCND3 mutant MV4;11 cells restored pexidartinib-induced cell-cycle arrest but not apoptosis., Conclusions: Mutations in CCND3 , a gene not commonly mutated in AML, are a novel cause of clinical primary resistance to FLT3 inhibitors in AML and may have sensitivity to CDK4/6 inhibition., (©2021 American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
9. The prognosis and durable clearance of RAS mutations in patients with acute myeloid leukemia receiving induction chemotherapy.
- Author
-
Ball BJ, Hsu M, Devlin SM, Arcila M, Roshal M, Zhang Y, Famulare CA, Goldberg AD, Cai SF, Dunbar A, Epstein-Peterson Z, Menghrajani KN, Glass JL, Taylor J, Viny AD, Giralt SS, Gyurkocza B, Shaffer BC, Tamari R, Levine RL, Tallman MS, and Stein EM
- Subjects
- Alleles, Bone Marrow pathology, Clone Cells, High-Throughput Nucleotide Sequencing, Humans, Kaplan-Meier Estimate, Leukemia, Myeloid, Acute genetics, Middle Aged, Prognosis, Progression-Free Survival, Proto-Oncogene Proteins p21(ras) genetics, Recurrence, Remission Induction, Signal Transduction genetics, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Gain of Function Mutation, Genes, ras, Induction Chemotherapy, Leukemia, Myeloid, Acute drug therapy, Mutation
- Published
- 2021
- Full Text
- View/download PDF
10. DOT1L inhibitors block abnormal self-renewal induced by cohesin loss.
- Author
-
Heimbruch KE, Fisher JB, Stelloh CT, Phillips E, Reimer MH Jr, Wargolet AJ, Meyer AE, Pulakanti K, Viny AD, Loppnow JJ, Levine RL, Pulikkan JA, Zhu N, and Rao S
- Subjects
- Animals, Benzimidazoles pharmacology, Cell Cycle Proteins deficiency, Cells, Cultured, Chromosomal Proteins, Non-Histone deficiency, Enzyme Inhibitors pharmacology, Epigenesis, Genetic, Hematopoietic Stem Cells metabolism, Hematopoietic Stem Cells physiology, Histone-Lysine N-Methyltransferase genetics, Histone-Lysine N-Methyltransferase metabolism, Histones metabolism, Homeodomain Proteins metabolism, Humans, Leukemia, Myeloid, Acute genetics, Mice, Cohesins, Cell Cycle Proteins genetics, Cell Self Renewal, Chromosomal Proteins, Non-Histone genetics, Hematopoietic Stem Cells drug effects, Histone-Lysine N-Methyltransferase antagonists & inhibitors, Leukemia, Myeloid, Acute metabolism
- Abstract
Acute myeloid leukemia (AML) is a high-risk malignancy characterized by a diverse spectrum of somatic genetic alterations. The mechanisms by which these mutations contribute to leukemia development and how this informs the use of targeted therapies is critical to improving outcomes for patients. Importantly, how to target loss-of-function mutations has been a critical challenge in precision medicine. Heterozygous inactivating mutations in cohesin complex genes contribute to AML in adults by increasing the self-renewal capacity of hematopoietic stem and progenitor cells (HSPCs) by altering PRC2 targeting to induce HOXA9 expression, a key self-renewal transcription factor. Here we sought to delineate the epigenetic mechanism underpinning the enhanced self-renewal conferred by cohesin-haploinsufficiency. First, given the substantial difference in the mutational spectrum between pediatric and adult AML patients, we first sought to identify if HOXA9 was also elevated in children. Next, using primary HSPCs as a model we demonstrate that abnormal self-renewal due to cohesin loss is blocked by DOT1L inhibition. In cohesin-depleted cells, DOT1L inhibition is associated with H3K79me2 depletion and a concomitant increase in H3K27me3. Importantly, we find that there are cohesin-dependent gene expression changes that promote a leukemic profile, including HoxA overexpression, that are preferentially reversed by DOT1L inhibition. Our data further characterize how cohesin mutations contribute to AML development, identifying DOT1L as a potential therapeutic target for adult and pediatric AML patients harboring cohesin mutations.
- Published
- 2021
- Full Text
- View/download PDF
11. Clinical and molecular predictors of response and survival following venetoclax therapy in relapsed/refractory AML.
- Author
-
Stahl M, Menghrajani K, Derkach A, Chan A, Xiao W, Glass J, King AC, Daniyan AF, Famulare C, Cuello BM, Horvat TZ, Abdel-Wahab O, Levine RL, Viny AD, Stein EM, Cai SF, Roshal M, Tallman MS, and Goldberg AD
- Subjects
- Bridged Bicyclo Compounds, Heterocyclic, Decitabine therapeutic use, Humans, Nucleophosmin, Retrospective Studies, Sulfonamides, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics
- Abstract
Azacitidine + venetoclax, decitabine + venetoclax, and low-dose cytarabine + venetoclax are now standard treatments for newly diagnosed older or unfit patients with acute myeloid leukemia (AML). Although these combinations are also commonly used in relapsed or refractory AML (RR-AML), clinical and molecular predictors of response and survival in RR-AML are incompletely understood. We retrospectively analyzed clinical and molecular characteristics and outcomes for 86 patients with RR-AML who were treated with venetoclax combinations. The complete remission (CR) or CR with incomplete hematologic recovery (CRi) rate was 24%, and the overall response rate was 31% with the inclusion of a morphologic leukemia-free state. Azacitidine + venetoclax resulted in higher response rates compared with low-dose cytarabine + venetoclax (49% vs 15%; P = .008). Median overall survival (OS) was 6.1 months, but it was significantly longer with azacitidine + venetoclax compared with low-dose cytarabine + venetoclax (25 vs 3.9 months; P = .003). This survival advantage of azacitidine + venetoclax over low-dose cytarabine + venetoclax persisted when patients were censored for subsequent allogeneic stem cell transplantation (8.1 vs 3.9 months; P = .035). Mutations in NPM1 were associated with higher response rates, whereas adverse cytogenetics and mutations in TP53, KRAS/NRAS, and SF3B1 were associated with worse OS. Relapse was driven by diverse mechanisms, including acquisition of novel mutations and an increase in cytogenetic complexity. Venetoclax combination therapy is effective in many patients with RR-AML, and pretreatment molecular characteristics may predict outcomes. Trials that evaluate novel agents in combination with venetoclax therapy in patients with RR-AML that have adverse risk genomic features are warranted., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
12. A cohesive look at leukemogenesis: The cohesin complex and other driving mutations in AML.
- Author
-
Heimbruch KE, Meyer AE, Agrawal P, Viny AD, and Rao S
- Subjects
- Animals, Carrier Proteins, Cell Cycle Proteins chemistry, Cell Cycle Proteins metabolism, Chromatin genetics, Chromatin metabolism, Chromatin Assembly and Disassembly, Chromosomal Proteins, Non-Histone chemistry, Chromosomal Proteins, Non-Histone metabolism, Genetic Predisposition to Disease, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Nucleophosmin, Protein Binding, Protein Multimerization, Structure-Activity Relationship, Cohesins, Cell Cycle Proteins genetics, Chromosomal Proteins, Non-Histone genetics, Disease Susceptibility, Leukemia, Myeloid, Acute etiology, Mutation, Oncogenes
- Abstract
Acute myeloid leukemia (AML) affects tens of thousands of patients a year, yet survival rates are as low as 25% in certain populations. This poor survival rate is partially due to the vast genetic diversity of the disease. Rarely do 2 patients with AML have the same mutational profile, which makes the development of targeted therapies particularly challenging. However, a set of recurrent mutations in chromatin modifiers have been identified in many patients, including mutations in the cohesin complex, which have been identified in up to 20% of cases. Interestingly, the canonical function of the cohesin complex in establishing sister chromatid cohesin during mitosis is unlikely to be the affected role in leukemogenesis. Instead, the cohesin complex's role in DNA looping and gene regulation likely facilitates disease. The epigenetic mechanisms by which cohesin complex mutations promote leukemia are not completely elucidated, but alterations of enhancer-promoter interactions and differential histone modifications have been shown to drive oncogenic gene expression changes. Such changes commonly include HoxA upregulation, which may represent a common pathway that could be therapeutically targeted. As cohesin mutations rarely occur alone, examining the impact of common co-occurring mutations, including those in NPM1, the core-binding factor complex, FLT3, and ASXL1, will yield additional insight. While further study of these mutational interactions is required, current research suggests that the use of combinatorial genetics could be the key to uncovering new targets, allowing for the treatment of AML patients based on their individual genetic profiles., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
13. Driver mutations in acute myeloid leukemia.
- Author
-
Kishtagari A, Levine RL, and Viny AD
- Subjects
- Humans, Leukemia, Myeloid, Acute pathology, Methylation, Mutation, Transcription Factors genetics, Epigenomics methods, Leukemia, Myeloid, Acute genetics
- Abstract
Purpose of Review: The mutational landscape of acute myeloid leukemia (AML) has revised diagnostic, prognostic, and therapeutic schemata over the past decade. Recurrently mutated AML genes have functional consequences beyond typical oncogene-driven growth and loss of tumor suppresser function., Recent Findings: Large-scale genomic sequencing efforts have mapped the complexity of AML and trials of mutation-based targeted therapy has led to several FDA-approved drugs for mutant-specific AML. However, many recurrent mutations have been identified across a spectrum from clonal hematopoiesis to myelodysplasia to overt AML, such as effectors of DNA methylation, chromatin modifiers, and spliceosomal machinery. The functional effects of these mutations are the basis for substantial discovery., Summary: Understanding the molecular and pathophysiologic functions of key genes that exert leukemogenic potential is essential towards translating these findings into better treatment for AML.
- Published
- 2020
- Full Text
- View/download PDF
14. Sphingolipid metabolism determines the therapeutic efficacy of nanoliposomal ceramide in acute myeloid leukemia.
- Author
-
Barth BM, Wang W, Toran PT, Fox TE, Annageldiyev C, Ondrasik RM, Keasey NR, Brown TJ, Devine VG, Sullivan EC, Cote AL, Papakotsi V, Tan SF, Shanmugavelandy SS, Deering TG, Needle DB, Stern ST, Zhu J, Liao J, Viny AD, Feith DJ, Levine RL, Wang HG, Loughran TP Jr, Sharma A, Kester M, and Claxton DF
- Subjects
- Antineoplastic Agents, Phytogenic pharmacology, Antineoplastic Agents, Phytogenic therapeutic use, Ceramides administration & dosage, Drug Delivery Systems, Humans, Leukemia, Myeloid, Acute metabolism, Nanostructures, Treatment Outcome, Vinblastine pharmacology, Vinblastine therapeutic use, Ceramides therapeutic use, Drug Carriers chemistry, Leukemia, Myeloid, Acute drug therapy, Liposomes chemistry, Sphingolipids metabolism
- Published
- 2019
- Full Text
- View/download PDF
15. Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia.
- Author
-
Xiao W, Goldberg AD, Famulare CA, Devlin SM, Nguyen NT, Sim S, Kabel CC, Patel MA, McGovern EM, Patel A, Schulman J, Dunbar AJ, Epstein-Peterson ZD, Menghrajani KN, Getta BM, Cai SF, Geyer MB, Glass JL, Taylor J, Viny AD, Levine RL, Zhang Y, Giralt SA, Klimek V, Tallman MS, and Roshal M
- Subjects
- Adult, Aged, Case-Control Studies, Combined Modality Therapy, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute pathology, Leukemia, Myeloid, Acute therapy, Male, Middle Aged, Neoplasm Recurrence, Local pathology, Neoplasm Recurrence, Local therapy, Neoplasm, Residual pathology, Neoplasm, Residual therapy, Prognosis, Retrospective Studies, Survival Rate, Dendritic Cells pathology, Leukemia, Myeloid, Acute mortality, Neoplasm Recurrence, Local mortality, Neoplasm, Residual mortality
- Abstract
Measurable residual disease is associated with inferior outcomes in patients with acute myeloid leukemia (AML). Measurable residual disease monitoring enhances risk stratification and may guide therapeutic intervention. The European LeukemiaNet working party recently came to a consensus recommendation incorporating leukemia associated immunophenotype-based different from normal approach by multi-color flow cytometry for measurable residual disease evaluation. However, the analytical approach is highly expertise-dependent and difficult to standardize. Here we demonstrate that loss of plasmacytoid dendritic cell differentiation after 7+3 induction in AML is highly specific for measurable residual disease positivity (specificity 97.4%) in a uniformly treated patient cohort. Moreover, loss of plasmacytoid dendritic cell differentiation as determined by a blast-to-plasmacytoid dendritic cell ratio >10 was strongly associated with inferior overall and relapse-free survival (RFS) [Hazard ratio 2.79, 95% confidence interval (95%CI): 0.98-7.97; P =0.077) and 3.83 (95%CI: 1.51-9.74; P =0.007), respectively), which is similar in magnitude to measurable residual disease positivity. Importantly, measurable residual disease positive patients who reconstituted plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio <10) showed a higher rate of measurable residual disease clearance at later pre-transplant time points compared to patients with loss of plasmacytoid dendritic cell differentiation (blast/ plasmacytoid dendritic cell ratio <10) (6 of 12, 50% vs 2 of 18, 11%; P =0.03). Furthermore pre-transplant plasmacytoid dendritic cell recovery was associated with superior outcome in measurable residual disease positive patients. Our study provides a novel, simple, broadly applicable, and quantitative multi-color flow cytometry approach to risk stratification in AML., (Copyright© 2019 Ferrata Storti Foundation.)
- Published
- 2019
- Full Text
- View/download PDF
16. Cohesin mutations in myeloid malignancies made simple.
- Author
-
Viny AD and Levine RL
- Subjects
- Animals, Antigens, Nuclear genetics, Antigens, Nuclear metabolism, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone metabolism, Hematologic Neoplasms metabolism, Hematologic Neoplasms pathology, Humans, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Myelodysplastic Syndromes metabolism, Myelodysplastic Syndromes pathology, Neoplasm Proteins metabolism, Cohesins, Cell Cycle Proteins genetics, Chromosomal Proteins, Non-Histone genetics, Hematologic Neoplasms genetics, Leukemia, Myeloid, Acute genetics, Mutation, Myelodysplastic Syndromes genetics, Neoplasm Proteins genetics
- Abstract
Purpose of Review: Recurrent loss of function mutations within genes of the cohesin complex have been identified in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). STAG2 is the most commonly mutated cohesin member in AML as well as solid tumors. STAG2 is recurrently, mutated in Ewing's Sarcoma, bladder cancer, and glioblastoma, and is one of only ten genes known to be recurrently mutated in over four distinct tissue types of human cancer RECENT FINDINGS: The cohesin complex, a multiprotein ring, is canonically known to align and stabilize replicated chromosomes prior to cell division. Although initially thought to lead to unequal chromosomal separation in dividing cells, data in myeloid malignancies show this is not observed in cohesin mutant MDS/AML, either in large patient cohorts or mouse models. Mounting evidence supports a potential alternate mechanism whereby drivers of cell-type specific gene expression and hematopoietic development are impaired through alteration in three-dimensional nuclear organization and gene structure., Summary: Understanding the functional consequences of cohesin mutations in regulating lineage-specific and signal-dependent defects and in myeloid transformation will identify novel pathophysiologic mechanisms of disease and inform the development of novel therapeutic targets.
- Published
- 2018
- Full Text
- View/download PDF
17. Racial disparities in the survival of American children, adolescents, and young adults with acute lymphoblastic leukemia, acute myelogenous leukemia, and Hodgkin lymphoma.
- Author
-
Kahn JM, Keegan TH, Tao L, Abrahão R, Bleyer A, and Viny AD
- Subjects
- Adolescent, Adult, Black or African American statistics & numerical data, Child, Child, Preschool, Female, Follow-Up Studies, Hispanic or Latino statistics & numerical data, Hodgkin Disease ethnology, Hodgkin Disease therapy, Humans, Infant, Infant, Newborn, Leukemia, Myeloid, Acute ethnology, Leukemia, Myeloid, Acute therapy, Male, Neoplasm Staging, Precursor Cell Lymphoblastic Leukemia-Lymphoma ethnology, Precursor Cell Lymphoblastic Leukemia-Lymphoma therapy, Prognosis, Retrospective Studies, SEER Program, Survival Rate, United States, White People statistics & numerical data, Young Adult, Health Status Disparities, Hodgkin Disease mortality, Leukemia, Myeloid, Acute mortality, Precursor Cell Lymphoblastic Leukemia-Lymphoma mortality
- Abstract
Background: Race-based survival in children and adolescents with hematologic malignancies has been a national challenge for decades. Large-scale investigations of age- and race-based survival trends over time in these patients have not previously been reported. The objective of this study was to investigate whether race- and age-related differences in pediatric and adolescent and young adult (AYA) leukemia and lymphoma survival persist and to what extent these differences have changed over time., Methods: Using the Surveillance, Epidemiology, and End Results program, this study investigated the outcomes of black and white (1975-2012; n = 27,369) and white and Hispanic (1992-2012; n = 20,574) children (0-14 years old) and AYAs (15-39 years old) with acute lymphoblastic leukemia (ALL), acute myelogenous leukemia (AML), and Hodgkin lymphoma (HL). Estimates of 5- and 10-year relative survival were compared over time., Results: Trends showed a convergence of survival for white and black children with ALL but a divergence in survival for AYA patients. Hispanic children and AYAs both suffered inferior outcomes. Trends for AML revealed persistent survival differences between black and white children and suggested worsening disparities for AYAs. Survival trends in HL revealed sustained survival differences between black and white AYA patients, whereas no differences were found in Hispanic and white patient outcomes for AML or HL., Conclusions: Although survival for children and AYAs with ALL, AML, and HL has improved over the past 4 decades, differences persist between black, white, and Hispanic children and AYAs; survival disparities between black and white children with ALL have been nearly eliminated. Strategies aimed at identifying causality and reducing disparities are warranted. Cancer 2016. © 2016 American Cancer Society. Cancer 2016;122:2723-2730. © 2016 American Cancer Society., Competing Interests: There are no conflicts of interest to disclose., (© 2016 American Cancer Society.)
- Published
- 2016
- Full Text
- View/download PDF
18. Roads Diverge--A Robert Frost View of Leukemia Development.
- Author
-
Viny AD and Levine RL
- Subjects
- Humans, Leukemia, Myeloid, Acute genetics, Mutation
- Published
- 2016
- Full Text
- View/download PDF
19. Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms.
- Author
-
Molenaar RJ, Thota S, Nagata Y, Patel B, Clemente M, Przychodzen B, Hirsh C, Viny AD, Hosano N, Bleeker FE, Meggendorfer M, Alpermann T, Shiraishi Y, Chiba K, Tanaka H, van Noorden CJ, Radivoyevitch T, Carraway HE, Makishima H, Miyano S, Sekeres MA, Ogawa S, Haferlach T, and Maciejewski JP
- Subjects
- Aged, DNA-Binding Proteins antagonists & inhibitors, DNA-Binding Proteins genetics, Dioxygenases, Female, Humans, Leukemia, Myeloid, Acute mortality, Male, Middle Aged, Myelodysplastic Syndromes mortality, Nucleophosmin, Prognosis, Proto-Oncogene Proteins antagonists & inhibitors, Proto-Oncogene Proteins genetics, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Mutation, Myelodysplastic Syndromes genetics
- Abstract
Mutations in isocitrate dehydrogenase 1/2 (IDH1/2(MT)) are drivers of a variety of myeloid neoplasms. As they yield the same oncometabolite, D-2-hydroxyglutarate, they are often treated as equivalent, and pooled. We studied the validity of this approach and found IDH1/2 mutations in 179 of 2119 myeloid neoplasms (8%). Cross-sectionally, the frequencies of these mutations increased from lower- to higher risk disease, thus suggesting a role in clinical progression. Variant allelic frequencies indicated that IDH1(MT) and IDH2(MT) are ancestral in up to 14/74 (19%) vs 34/99 (34%; P=0.027) of cases, respectively, illustrating the pathogenic role of these lesions in myeloid neoplasms. IDH1/2(MT) was associated with poor overall survival, particularly in lower risk myelodysplastic syndromes. Ancestral IDH1(MT) cases were associated with a worse prognosis than subclonal IDH1(MT) cases, whereas the position of IDH2(MT) within clonal hierarchy did not impact survival. This may relate to distinct mutational spectra with more DNMT3A and NPM1 mutations associated with IDH1(MT) cases, and more ASXL1, SRSF2, RUNX1, STAG2 mutations associated with IDH2(MT) cases. Our data demonstrate important clinical and biological differences between IDH1(MT) and IDH2(MT) myeloid neoplasms. These mutations should be considered separately as their differences could have implications for diagnosis, prognosis and treatment with IDH1/2(MT) inhibitors of IDH1/2(MT) patients.
- Published
- 2015
- Full Text
- View/download PDF
20. Dose-dependent role of the cohesin complex in normal and malignant hematopoiesis.
- Author
-
Viny AD, Ott CJ, Spitzer B, Rivas M, Meydan C, Papalexi E, Yelin D, Shank K, Reyes J, Chiu A, Romin Y, Boyko V, Thota S, Maciejewski JP, Melnick A, Bradner JE, and Levine RL
- Subjects
- Animals, Cell Cycle Proteins genetics, Chondroitin Sulfate Proteoglycans genetics, Chromatin chemistry, Chromosomal Proteins, Non-Histone genetics, Haploinsufficiency, Hematopoietic Stem Cells physiology, Leukemia, Myeloid, Acute genetics, Mice, STAT5 Transcription Factor physiology, fms-Like Tyrosine Kinase 3 genetics, Cohesins, Cell Cycle Proteins physiology, Chromosomal Proteins, Non-Histone physiology, Hematopoiesis, Leukemia, Myeloid, Acute etiology
- Abstract
Cohesin complex members have recently been identified as putative tumor suppressors in hematologic and epithelial malignancies. The cohesin complex guides chromosome segregation; however, cohesin mutant leukemias do not show genomic instability. We hypothesized that reduced cohesin function alters chromatin structure and disrupts cis-regulatory architecture of hematopoietic progenitors. We investigated the consequences of Smc3 deletion in normal and malignant hematopoiesis. Biallelic Smc3 loss induced bone marrow aplasia with premature sister chromatid separation and revealed an absolute requirement for cohesin in hematopoietic stem cell (HSC) function. In contrast, Smc3 haploinsufficiency increased self-renewal in vitro and in vivo, including competitive transplantation. Smc3 haploinsufficiency reduced coordinated transcriptional output, including reduced expression of transcription factors and other genes associated with lineage commitment. Smc3 haploinsufficiency cooperated with Flt3-ITD to induce acute leukemia in vivo, with potentiated Stat5 signaling and altered nucleolar topology. These data establish a dose dependency for cohesin in regulating chromatin structure and HSC function., (© 2015 Viny et al.)
- Published
- 2015
- Full Text
- View/download PDF
21. Genetic alterations of the cohesin complex genes in myeloid malignancies.
- Author
-
Thota S, Viny AD, Makishima H, Spitzer B, Radivoyevitch T, Przychodzen B, Sekeres MA, Levine RL, and Maciejewski JP
- Subjects
- Aged, Core Binding Factor Alpha 2 Subunit genetics, Core Binding Factor Alpha 2 Subunit metabolism, Cross-Sectional Studies, Disease-Free Survival, Female, Humans, Male, Proto-Oncogene Proteins genetics, Proto-Oncogene Proteins metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Survival Rate, Cohesins, Cell Cycle Proteins genetics, Cell Cycle Proteins metabolism, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Genes, Neoplasm, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute mortality, Mutation, Myeloproliferative Disorders genetics, Myeloproliferative Disorders metabolism, Myeloproliferative Disorders mortality
- Abstract
Somatic cohesin mutations have been reported in myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML). To account for the morphologic and cytogenetic diversity of these neoplasms, a well-annotated cohort of 1060 patients with myeloid malignancies including MDS (n = 386), myeloproliferative neoplasms (MPNs) (n = 55), MDS/MPNs (n = 169), and AML (n = 450) were analyzed for cohesin gene mutational status, gene expression, and therapeutic and survival outcomes. Somatic cohesin defects were detected in 12% of patients with myeloid malignancies, whereas low expression of these genes was present in an additional 15% of patients. Mutations of cohesin genes were mutually exclusive and mostly resulted in predicted loss of function. Patients with low cohesin gene expression showed similar expression signatures as those with somatic cohesin mutations. Cross-sectional deep-sequencing analysis for clonal hierarchy demonstrated STAG2, SMC3, and RAD21 mutations to be ancestral in 18%, 18%, and 47% of cases, respectively, and each expanded to clonal dominance concordant with disease transformation. Cohesin mutations were significantly associated with RUNX1, Ras-family oncogenes, and BCOR and ASXL1 mutations and were most prevalent in high-risk MDS and secondary AML. Cohesin defects were associated with poor overall survival (27.2 vs 40 months; P = .023), especially in STAG2 mutant MDS patients surviving >12 months (median survival 35 vs 50 months; P = .017)., (© 2014 by The American Society of Hematology.)
- Published
- 2014
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.