Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Genome Res ; 31(5): 747-761, 2021 05.
Article in English | MEDLINE | ID: mdl-33707228

ABSTRACT

Acute myeloid leukemia (AML) is a molecularly complex disease characterized by heterogeneous tumor genetic profiles and involving numerous pathogenic mechanisms and pathways. Integration of molecular data types across multiple patient cohorts may advance current genetic approaches for improved subclassification and understanding of the biology of the disease. Here, we analyzed genome-wide DNA methylation in 649 AML patients using Illumina arrays and identified a configuration of 13 subtypes (termed "epitypes") using unbiased clustering. Integration of genetic data revealed that most epitypes were associated with a certain recurrent mutation (or combination) in a majority of patients, yet other epitypes were largely independent. Epitypes showed developmental blockage at discrete stages of myeloid differentiation, revealing epitypes that retain arrested hematopoietic stem-cell-like phenotypes. Detailed analyses of DNA methylation patterns identified unique patterns of aberrant hyper- and hypomethylation among epitypes, with variable involvement of transcription factors influencing promoter, enhancer, and repressed regions. Patients in epitypes with stem-cell-like methylation features showed inferior overall survival along with up-regulated stem cell gene expression signatures. We further identified a DNA methylation signature involving STAT motifs associated with FLT3-ITD mutations. Finally, DNA methylation signatures were stable at relapse for the large majority of patients, and rare epitype switching accompanied loss of the dominant epitype mutations and reversion to stem-cell-like methylation patterns. These results show that DNA methylation-based classification integrates important molecular features of AML to reveal the diverse pathogenic and biological aspects of the disease.


Subject(s)
DNA Methylation , Leukemia, Myeloid, Acute , Humans , Leukemia, Myeloid, Acute/metabolism , Mutation , Promoter Regions, Genetic
2.
Nature ; 562(7728): 526-531, 2018 10.
Article in English | MEDLINE | ID: mdl-30333627

ABSTRACT

The implementation of targeted therapies for acute myeloid leukaemia (AML) has been challenging because of the complex mutational patterns within and across patients as well as a dearth of pharmacologic agents for most mutational events. Here we report initial findings from the Beat AML programme on a cohort of 672 tumour specimens collected from 562 patients. We assessed these specimens using whole-exome sequencing, RNA sequencing and analyses of ex vivo drug sensitivity. Our data reveal mutational events that have not previously been detected in AML. We show that the response to drugs is associated with mutational status, including instances of drug sensitivity that are specific to combinatorial mutational events. Integration with RNA sequencing also revealed gene expression signatures, which predict a role for specific gene networks in the drug response. Collectively, we have generated a dataset-accessible through the Beat AML data viewer (Vizome)-that can be leveraged to address clinical, genomic, transcriptomic and functional analyses of the biology of AML.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Genomics , Leukemia, Myeloid, Acute/genetics , Core Binding Factor Alpha 2 Subunit/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , Datasets as Topic , Exome/genetics , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Male , Molecular Targeted Therapy , Nuclear Proteins/genetics , Nucleophosmin , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Sequence Analysis, RNA , Serine-Arginine Splicing Factors/genetics
3.
Cancer Cell ; 40(8): 850-864.e9, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35868306

ABSTRACT

Acute myeloid leukemia (AML) is a cancer of myeloid-lineage cells with limited therapeutic options. We previously combined ex vivo drug sensitivity with genomic, transcriptomic, and clinical annotations for a large cohort of AML patients, which facilitated discovery of functional genomic correlates. Here, we present a dataset that has been harmonized with our initial report to yield a cumulative cohort of 805 patients (942 specimens). We show strong cross-cohort concordance and identify features of drug response. Further, deconvoluting transcriptomic data shows that drug sensitivity is governed broadly by AML cell differentiation state, sometimes conditionally affecting other correlates of response. Finally, modeling of clinical outcome reveals a single gene, PEAR1, to be among the strongest predictors of patient survival, especially for young patients. Collectively, this report expands a large functional genomic resource, offers avenues for mechanistic exploration and drug development, and reveals tools for predicting outcome in AML.


Subject(s)
Leukemia, Myeloid, Acute , Cell Differentiation , Cohort Studies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Receptors, Cell Surface/genetics , Transcriptome
4.
Cancer Res ; 79(18): 4744-4753, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31311809

ABSTRACT

Although the use of ATP-competitive tyrosine kinase inhibitors of oncoprotein BCR-ABL1 has enabled durable responses in patients with chronic myeloid leukemia (CML), issues of drug resistance and residual leukemic stem cells remain. To test whether the degradation of BCR-ABL1 kinase could offer improved response, we developed a series of proteolysis-targeting chimera (PROTAC) that allosterically target BCR-ABL1 protein and recruit the E3 ligase Von Hippel-Lindau, resulting in ubiquitination and subsequent degradation of the oncogenic fusion protein. In both human CML K562 cells and murine Ba/F3 cells expressing BCR-ABL1, lead compound GMB-475 induced rapid proteasomal degradation and inhibition of downstream biomarkers, such as STAT5, and showed increased sensitivity compared with diastereomeric controls lacking degradation activity. Notably, GMB-475 inhibited the proliferation of certain clinically relevant BCR-ABL1 kinase domain point mutants and further sensitized Ba/F3 BCR-ABL1 cells to inhibition by imatinib, while demonstrating no toxicity toward Ba/F3 parental cells. Reverse phase protein array analysis suggested additional differences in levels of phosphorylated SHP2, GAB2, and SHC associated with BCR-ABL1 degradation. Importantly, GMB-475 reduced viability and increased apoptosis in primary CML CD34+ cells, with no effect on healthy CD34+ cells at identical concentrations. GMB-475 degraded BCR-ABL1 and reduced cell viability in primary CML stem cells. Together, these findings suggest that combined BCR-ABL1 kinase inhibition and protein degradation may represent a strategy to address BCR-ABL1-dependent drug resistance, and warrant further investigation into the eradication of persistent leukemic stem cells, which rely on neither the presence nor the activity of the BCR-ABL1 protein for survival. SIGNIFICANCE: Small-molecule-induced degradation of BCR-ABL1 in CML provides an advantage over inhibition and provides insights into CML stem cell biology. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/18/4744/F1.large.jpg.


Subject(s)
Fusion Proteins, bcr-abl/antagonists & inhibitors , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Neoplastic Stem Cells/drug effects , Protein Kinase Inhibitors/pharmacology , Proteolysis/drug effects , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Array Analysis , Tumor Cells, Cultured
5.
Cancer Cell ; 36(4): 431-443.e5, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31543464

ABSTRACT

BCR-ABL1 point mutation-mediated resistance to tyrosine kinase inhibitor (TKI) therapy in Philadelphia chromosome-positive (Ph+) leukemia is effectively managed with several approved drugs, including ponatinib for BCR-ABL1T315I-mutant disease. However, therapy options are limited for patients with leukemic clones bearing multiple BCR-ABL1 mutations. Asciminib, an allosteric inhibitor targeting the myristoyl-binding pocket of BCR-ABL1, is active against most single mutants but ineffective against all tested compound mutants. We demonstrate that combining asciminib with ATP site TKIs enhances target inhibition and suppression of resistant outgrowth in Ph+ clinical isolates and cell lines. Inclusion of asciminib restores ponatinib's effectiveness against currently untreatable compound mutants at clinically achievable concentrations. Our findings support combining asciminib with ponatinib as a treatment strategy for this molecularly defined group of patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm/drug effects , Fusion Proteins, bcr-abl/antagonists & inhibitors , Imidazoles/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Niacinamide/analogs & derivatives , Pyrazoles/pharmacology , Pyridazines/pharmacology , Allosteric Regulation/drug effects , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Binding Sites/drug effects , Binding Sites/genetics , Cell Line, Tumor/transplantation , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Female , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Imidazoles/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Molecular Docking Simulation , Molecular Dynamics Simulation , Molecular Targeted Therapy/methods , Mutation , Niacinamide/pharmacology , Niacinamide/therapeutic use , Primary Cell Culture , Pyrazoles/therapeutic use , Pyridazines/therapeutic use
6.
Nat Commun ; 10(1): 244, 2019 01 16.
Article in English | MEDLINE | ID: mdl-30651561

ABSTRACT

FLT3 mutations are prevalent in AML patients and confer poor prognosis. Crenolanib, a potent type I pan-FLT3 inhibitor, is effective against both internal tandem duplications and resistance-conferring tyrosine kinase domain mutations. While crenolanib monotherapy has demonstrated clinical benefit in heavily pretreated relapsed/refractory AML patients, responses are transient and relapse eventually occurs. Here, to investigate the mechanisms of crenolanib resistance, we perform whole exome sequencing of AML patient samples before and after crenolanib treatment. Unlike other FLT3 inhibitors, crenolanib does not induce FLT3 secondary mutations, and mutations of the FLT3 gatekeeper residue are infrequent. Instead, mutations of NRAS and IDH2 arise, mostly as FLT3-independent subclones, while TET2 and IDH1 predominantly co-occur with FLT3-mutant clones and are enriched in crenolanib poor-responders. The remaining patients exhibit post-crenolanib expansion of mutations associated with epigenetic regulators, transcription factors, and cohesion factors, suggesting diverse genetic/epigenetic mechanisms of crenolanib resistance. Drug combinations in experimental models restore crenolanib sensitivity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzimidazoles/pharmacology , Drug Resistance, Neoplasm/genetics , Leukemia, Myeloid, Acute/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Epigenesis, Genetic/drug effects , Female , GTP Phosphohydrolases/genetics , HEK293 Cells , Humans , Inhibitory Concentration 50 , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Membrane Proteins/genetics , Mice , Middle Aged , Mutation/drug effects , Mutation/genetics , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Tandem Repeat Sequences/genetics , Treatment Outcome , Exome Sequencing
7.
Cancer Res ; 77(16): 4258-4267, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28652245

ABSTRACT

Exclusive of membrane-proximal mutations seen commonly in chronic neutrophilic leukemia (e.g., T618I), functionally defective mutations in the extracellular domain of the G-CSF receptor (CSF3R) have been reported only in severe congenital and idiopathic neutropenia patients. Here, we describe the first activating mutation in the fibronectin-like type III domain of the extracellular region of CSF3R (W341C) in a leukemia patient. This mutation transformed cells via cysteine-mediated intermolecular disulfide bonds, leading to receptor dimerization. Interestingly, a CSF3R cytoplasmic truncation mutation (W791X) found on the same allele as the extracellular mutation and the expansion of the compound mutation was associated with increased leukocytosis and disease progression of the patient. Notably, the primary patient sample and cells transformed by W341C and W341C/W791X exhibited sensitivity to JAK inhibitors. We further showed that disruption of original cysteine pairs in the CSF3R extracellular domain resulted in either gain- or loss-of-function changes, part of which was attributable to cysteine-mediated dimer formation. This, therefore, represents the first characterization of unpaired cysteines that mediate both gain- and loss-of-function phenotypes. Overall, our results show the structural and functional importance of conserved extracellular cysteine pairs in CSF3R and suggest the necessity for broader screening of CSF3R extracellular domain in leukemia patients. Cancer Res; 77(16); 4258-67. ©2017 AACR.


Subject(s)
Cysteine/genetics , Leukemia, Myeloid, Acute/genetics , Mutation, Missense , Receptors, Colony-Stimulating Factor/genetics , Aged , Animals , Exome , Female , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL