Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 129(4): 1626-1640, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30720463

ABSTRACT

The discovery of recurrent mutations in subunits of the vacuolar-type H+-translocating ATPase (v-ATPase) in follicular lymphoma (FL) highlights a role for the amino acid- and energy-sensing pathway to mTOR in the pathogenesis of this disease. Here, through the use of complementary experimental approaches involving mammalian cells and Saccharomyces cerevisiae, we have demonstrated that mutations in the human v-ATPase subunit ATP6V1B2 (also known as Vma2 in yeast) activate autophagic flux and maintain mTOR/TOR in an active state. Engineered lymphoma cell lines and primary FL B cells carrying mutated ATP6V1B2 demonstrated a remarkable ability to survive low leucine concentrations. The treatment of primary FL B cells with inhibitors of autophagy uncovered an addiction for survival for FL B cells harboring ATP6V1B2 mutations. These data support the idea of mutational activation of autophagic flux by recurrent hotspot mutations in ATP6V1B2 as an adaptive mechanism in FL pathogenesis and as a possible new therapeutically targetable pathway.


Subject(s)
Autophagic Cell Death , Lymphoma, Follicular/enzymology , Mutation , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Cell Line, Tumor , Cell Survival , Humans , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , TOR Serine-Threonine Kinases/genetics , Vacuolar Proton-Translocating ATPases/genetics
2.
Clin Cancer Res ; 23(4): 1049-1059, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27535981

ABSTRACT

Purpose: Ibrutinib, a Bruton tyrosine kinase (BTK) inhibitor, is approved for the treatment of relapsed chronic lymphocytic leukemia (CLL) and CLL with del17p. Mechanistically, ibrutinib interferes with B-cell receptor (BCR) signaling as well as multiple CLL cell-to-microenvironment interactions. Given the importance of ibrutinib in the management of CLL, a deeper understanding of factors governing sensitivity and resistance is warranted.Experimental Design: We studied 48 longitudinally sampled paired CLL samples, 42 of which were procured before and after standard CLL chemotherapies, and characterized them for well-studied CLL molecular traits as well as by whole-exome sequencing and SNP 6.0 array profiling. We exposed these samples to 0.25 to 5 µmol/L of ibrutinib ex vivo and measured apoptosis fractions as well as BCR signaling by immunoblotting. We disrupted TP53 in HG3, PGA1, and PG-EBV cell lines and measured BCR signaling and ibrutinib responses.Results: CLL samples demonstrated a surprisingly wide range of ex vivo sensitivities to ibrutinib, with IC50 values ranging from 0.4 to 9.7 µmol/L. Unmutated IGVH status, elevated ZAP70 expression, and trisomy 12 were associated with heightened sensitivity to ibrutinib treatment. Five CLL samples were substantially more resistant to ibrutinib following relapse from chemotherapy; of these, three had acquired a del17p/TP53-mutated status. A validation sample of 15 CLL carrying TP53 mutations, of which 13 carried both del17p and a TP53 mutation, confirmed substantially less sensitivity to ibrutinib-induced apoptosis.Conclusions: This study identifies that CLL harboring del17p/TP53-mutated cells are substantially less sensitive to ibrutinib-induced apoptosis than del17p/TP53 wild-type cells. Clin Cancer Res; 23(4); 1049-59. ©2016 AACR.


Subject(s)
Biomarkers, Pharmacological , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pyrazoles/administration & dosage , Pyrimidines/administration & dosage , Tumor Suppressor Protein p53/genetics , Adenine/analogs & derivatives , Apoptosis/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Piperidines , Polymorphism, Single Nucleotide , Pyrazoles/adverse effects , Pyrimidines/adverse effects , Exome Sequencing
3.
Clin Cancer Res ; 22(21): 5383-5393, 2016 Nov 01.
Article in English | MEDLINE | ID: mdl-27267853

ABSTRACT

PURPOSE: This study was performed to further our understanding of the biological and genetic basis of follicular lymphoma and to identify potential novel therapy targets. EXPERIMENTAL DESIGN: We analyzed previously generated whole exome sequencing data of 23 follicular lymphoma cases and one transformed follicular lymphoma case and expanded findings to a combined total of 125 follicular lymphoma/3 transformed follicular lymphoma. We modeled the three-dimensional location of RRAGC-associated hotspot mutations. We performed functional studies on novel RRAGC mutants in stable retrovirally transduced HEK293T cells, stable lentivirally transduced lymphoma cell lines, and in Saccharomyces cerevisiae RESULTS: We report recurrent mutations, including multiple amino acid hotspots, in the small G-protein RRAGC, which is part of a protein complex that signals intracellular amino acid concentrations to MTOR, in 9.4% of follicular lymphoma cases. Mutations in RRAGC distinctly clustered on one protein surface area surrounding the GTP/GDP-binding sites. Mutated RRAGC proteins demonstrated increased binding to RPTOR (raptor) and substantially decreased interactions with the product of the tumor suppressor gene FLCN (folliculin). In stable retrovirally transfected 293T cells, cultured in the presence or absence of leucine, multiple RRAGC mutations demonstrated elevated MTOR activation as evidenced by increased RPS6KB/S6-kinase phosphorylation. Similar activation phenotypes were uncovered in yeast engineered to express mutations in the RRAGC homolog Gtr2 and in multiple lymphoma cell lines expressing HA-tagged RRAGC-mutant proteins. CONCLUSIONS: Our discovery of activating mutations in RRAGC in approximately 10% of follicular lymphoma provides the mechanistic rationale to study mutational MTOR activation and MTOR inhibition as a potential novel actionable therapeutic target in follicular lymphoma. Clin Cancer Res; 22(21); 5383-93. ©2016 AACR.


Subject(s)
Lymphoma, Follicular/genetics , Monomeric GTP-Binding Proteins/genetics , Mutation/genetics , Neoplasm Recurrence, Local/genetics , TOR Serine-Threonine Kinases/genetics , Amino Acids/genetics , Binding Sites/genetics , Cell Line , Genes, Tumor Suppressor/physiology , Guanosine Diphosphate/genetics , Guanosine Triphosphate/genetics , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phosphorylation/genetics , Regulatory-Associated Protein of mTOR/genetics , Signal Transduction/genetics
4.
Clin Cancer Res ; 22(17): 4525-35, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27060156

ABSTRACT

PURPOSE: Chronic lymphocytic leukemia (CLL)-associated gene mutations that influence CLL cell fitness and chemotherapy resistance should increase in clonal representation when measured before therapy and at relapse. EXPERIMENTAL DESIGN: To uncover mutations associated with CLL relapse, we have performed whole-exome sequencing in a discovery cohort of 61 relapsed CLL patients identifying 86 recurrently mutated genes. The variant allele fractions (VAF) of 19 genes with mutations in ≥3 of 61 cases were measured in 53 paired pre- and posttreatment CLL samples sorted to purity using panel-based deep resequencing or by droplet digital PCR. RESULTS: We identify mutations in TP53 as the dominant subclonal gene driver of relapsed CLL often demonstrating substantial increases in VAFs. Subclonal mutations in SAMHD1 also recurrently demonstrated increased VAFs at relapse. Mutations in ATP10A, FAT3, FAM50A, and MGA, although infrequent, demonstrated enrichment in ≥2 cases each. In contrast, mutations in NOTCH1, SF3B1, POT1, FBXW7, MYD88, NXF1, XPO1, ZMYM3, or CHD2 were predominantly already clonal prior to therapy indicative of a pretreatment pathogenetic driver role in CLL. Quantitative analyses of clonal dynamics uncover rising, stable, and falling clones and subclones without clear evidence that gene mutations other than in TP53 and possibly SAMHD1 are frequently selected for at CLL relapse. CONCLUSIONS: Data in aggregate support a provisional categorization of CLL-associated recurrently mutated genes into three classes (i) often subclonal before therapy and strongly enriched after therapy, or, (ii) mostly clonal before therapy or without further enrichments at relapse, or, (iii) subclonal before and after therapy and enriching only in sporadic cases. Clin Cancer Res; 22(17); 4525-35. ©2016 AACR.


Subject(s)
Clonal Evolution/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Adult , Aged , Aged, 80 and over , Alleles , Biomarkers, Tumor , Disease Progression , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Exome Sequencing
5.
Clin Cancer Res ; 21(9): 2045-56, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25652455

ABSTRACT

PURPOSE: Recurrent gene mutations, chromosomal translocations, and acquired genomic copy number aberrations (aCNA) have been variously associated with acute myelogenous leukemia (AML) patient outcome. However, knowledge of the co-occurrence of such lesions and the relative influence of different types of genomic alterations on clinical outcomes in AML is still evolving. EXPERIMENTAL DESIGN: We performed SNP 6.0 array-based genomic profiling of aCNA/copy neutral loss-of-heterozygosity (cnLOH) along with sequence analysis of 13 commonly mutated genes on purified leukemic blast DNA from 156 prospectively enrolled non-FAB-M3 AML patients across the clinical spectrum of de novo, secondary, and therapy-related AML. RESULTS: TP53 and RUNX1 mutations are strongly associated with the presence of SNP-A-based aCNA/cnLOH, while FLT3 and NPM1 mutations are strongly associated with the absence of aCNA/cnLOH. The presence of mutations in RUNX1, ASXL1, and TP53, elevated SNP-A-based genomic complexity, and specific recurrent aCNAs predicted failure to achieve a complete response to induction chemotherapy. The presence of ≥1 aCNA/cnLOH and higher thresholds predicted for poor long-term survival irrespective of TP53 status, and the presence of ≥1 aCNA/cnLOH added negative prognostic information to knowledge of mutations in TET2, IDH1, NPM1, DNMT3A, and RUNX1. Results of multivariate analyses support a dominant role for TP53 mutations and a role for elevated genomic complexity as predictors of short survival in AML. CONCLUSIONS: Integrated genomic profiling of a clinically relevant adult AML cohort identified genomic aberrations most associated with SNP-A-based genomic complexity, resistance to intensive induction therapies, and shortened overall survival. Identifying SNP-A-based lesions adds prognostic value to the status of several recurrently mutated genes.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/therapy , Adult , DNA Mutational Analysis , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Loss of Heterozygosity , Male , Middle Aged , Nucleophosmin , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Transcriptome
6.
Blood ; 125(4): 668-79, 2015 Jan 22.
Article in English | MEDLINE | ID: mdl-25428220

ABSTRACT

Follicular lymphoma (FL) is the second most common non-Hodgkin lymphoma in the Western world. FL cell-intrinsic and cell-extrinsic factors influence FL biology and clinical outcome. To further our understanding of the genetic basis of FL, we performed whole-exome sequencing of 23 highly purified FL cases and 1 transformed FL case and expanded findings to a combined total of 114 FLs. We report recurrent mutations in the transcription factor STAT6 in 11% of FLs and identified the STAT6 amino acid residue 419 as a novel STAT6 mutation hotspot (p.419D/G, p.419D/A, and p.419D/H). FL-associated STAT6 mutations were activating, as evidenced by increased transactivation in HEK293T cell-based transfection/luciferase reporter assays, heightened interleukin-4 (IL-4) -induced activation of target genes in stable STAT6 transfected lymphoma cell lines, and elevated baseline expression levels of STAT6 target genes in primary FL B cells harboring mutant STAT6. Mechanistically, FL-associated STAT6 mutations facilitated nuclear residency of STAT6, independent of IL-4-induced STAT6-Y641 phosphorylation. Structural modeling of STAT6 based on the structure of the STAT1-DNA complex revealed that most FL-associated STAT6 mutants locate to the STAT6-DNA interface, potentially facilitating heightened interactions. The genetic and functional data combined strengthen the recognition of the IL-4/JAK/STAT6 axis as a driver of FL pathogenesis.


Subject(s)
Cell Nucleus/metabolism , Gene Expression Regulation, Neoplastic , Lymphoma, Follicular/metabolism , Mutation, Missense , Neoplasm Proteins/metabolism , STAT6 Transcription Factor/metabolism , Active Transport, Cell Nucleus/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Genome-Wide Association Study , HEK293 Cells , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Janus Kinases/genetics , Janus Kinases/metabolism , Lymphoma, Follicular/genetics , Lymphoma, Follicular/pathology , Neoplasm Proteins/genetics , Phosphorylation/genetics , STAT6 Transcription Factor/genetics , Transcriptional Activation/genetics
7.
Blood ; 123(10): 1487-98, 2014 Mar 06.
Article in English | MEDLINE | ID: mdl-24435047

ABSTRACT

Follicular lymphoma (FL) constitutes the second most common non-Hodgkin lymphoma in the western world. FL carries characteristic recurrent structural genomic aberrations. However, information regarding the coding genome in FL is still evolving. Here, we describe the results of massively parallel exome sequencing and single nucleotide polymorphism 6.0 array genomic profiling of 11 highly purified FL cases, and 1 transformed FL case and the validation of selected mutations in 102 FL cases. We report the identification of 15 novel recurrently mutated genes in FL. These include frequent mutations in the linker histone genes HIST1H1 B-E (27%) and mutations in OCT2 (also known as POU2F2; 8%), IRF8 (6%), and ARID1A (11%). A subset of the mutations in HIST1H1 B-E affected binding to DNMT3B, and mutations in HIST1H1 B-E and in EZH2 or ARID1A were largely mutually exclusive, implicating HIST1H1 B-E in epigenetic deregulation in FL. Mutations in OCT2 (POU2F2) affected its transcriptional and functional properties as measured through luciferase assays, the biological analysis of stably transduced cell lines, and global expression profiling. Finally, multiple novel mutated genes located within regions of acquired uniparental disomy in FL are identified. In aggregate, these data substantially broaden our understanding of the genomic pathogenesis of FL.


Subject(s)
Histones/genetics , Interferon Regulatory Factors/genetics , Lymphoma, Follicular/genetics , Mutation , Nuclear Proteins/genetics , Octamer Transcription Factor-2 , Amino Acid Sequence , CREB-Binding Protein/genetics , DNA Mutational Analysis , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Epigenesis, Genetic , Exome , High-Throughput Nucleotide Sequencing , Histones/chemistry , Humans , Molecular Sequence Data , Neoplasm Proteins/genetics , Polycomb Repressive Complex 2/genetics , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-bcl-2/genetics , Receptors, Tumor Necrosis Factor, Member 14/genetics , Sequence Alignment , Transcription Factors , Transcriptional Activation
8.
Clin Cancer Res ; 19(11): 2893-904, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23620403

ABSTRACT

PURPOSE: The identification of gene mutations and structural genomic aberrations that are critically involved in chronic lymphocytic leukemia (CLL) pathogenesis is still evolving. One may postulate that genomic driver lesions with effects on CLL cell proliferation, apoptosis thresholds, or chemotherapy resistance should increase in frequency over time when measured sequentially in a large CLL cohort. EXPERIMENTAL DESIGN: We sequentially sampled a large well-characterized CLL cohort at a mean of 4 years between samplings and measured acquired copy number aberrations (aCNA) and LOH using single-nucleotide polymorphism (SNP) 6.0 array profiling and the mutational state of TP53, NOTCH1, and SF3B1 using Sanger sequencing. The paired analysis included 156 patients, of whom 114 remained untreated and 42 received intercurrent therapies, predominantly potent chemoimmunotherapy, during the sampling interval. RESULTS: We identify a strong effect of intercurrent therapies on the frequency of acquisition of aCNAs in CLL. Importantly, the spectrum of acquired genomic changes was largely similar in patients who did or did not receive intercurrent therapies; therefore, various genomic changes that become part of the dominant clones are often already present in CLL cell populations before therapy. Furthermore, we provide evidence that therapy of CLL with preexisting TP53 mutations results in outgrowth of genomically very complex clones, which dominate at relapse. CONCLUSIONS: Using complementary technologies directed at the detection of genomic events that are present in substantial proportions of the clinically relevant CLL disease bulk, we capture aspects of genomic evolution in CLL over time, including increases in the frequency of genomic complexity, specific recurrent aCNAs, and TP53 mutations.


Subject(s)
Clonal Evolution/genetics , Genomics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Adult , Aged , Aged, 80 and over , Chromosome Aberrations , DNA Copy Number Variations , Disease Progression , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Loss of Heterozygosity , Male , Middle Aged , Mutation , Phosphoproteins/genetics , Polymorphism, Single Nucleotide , RNA Splicing Factors , Receptor, Notch1/genetics , Ribonucleoprotein, U2 Small Nuclear/genetics , Tumor Suppressor Protein p53/genetics
9.
Clin Cancer Res ; 17(9): 2679-92, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21307146

ABSTRACT

PURPOSE: The chromosomal deletion 11q affects biology and clinical outcome in chronic lymphocytic leukemia (CLL) but del11q-deregulated genes remain incompletely characterized. EXPERIMENTAL DESIGN: We have employed integrated genomic profiling approaches on CLL cases with and without del11q to identify 11q-relevant genes. RESULTS: We have identified differential expression of the insulin receptor (INSR) in CLL, including high-level INSR expression in the majority of CLL with del11q. High INSR mRNA expression in 11q CLL (∼10-fold higher mean levels than other genomic categories) was confirmed by quantitative PCR in 247 CLL cases. INSR protein measurements in 257 CLL cases through flow cytometry, compared with measurements in normal CD19(+) B cells and monocytes, confirmed that a subset of CLL aberrantly expresses high INSR levels. INSR stimulation by insulin in CLL cells ex vivo resulted in the activation of canonical INSR signaling pathways, including the AKT-mTOR and Ras/Raf/Erk pathways, and INSR activation partially abrogated spontaneous CLL cell apoptosis ex vivo. Higher INSR levels correlated with shorter time to first therapy and shorter overall survival (OS). In bivariate analysis, INSR expression predicted for rapid initial disease progression and shorter OS in ZAP-70-low/negative CLL. Finally, in multivariate analysis (ZAP-70 status, IgV(H) status, and INSR expression), we detected elevated HRs and trends for short OS for CLL cases with high INSR expression (analyzed inclusive or exclusive of cases with del11q). CONCLUSIONS: Our aggregate biochemical and clinical outcome data suggest biologically meaningful elevated INSR expression in a substantial subset of all CLL cases, including many cases with del11q.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Receptor, Insulin/physiology , Adult , Chromosome Deletion , Chromosomes, Human, Pair 11 , Cluster Analysis , Comparative Genomic Hybridization , Cytogenetic Analysis , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Genetic Predisposition to Disease , Humans , Microarray Analysis , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Validation Studies as Topic
10.
Bioorg Med Chem Lett ; 16(4): 1060-4, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16289811

ABSTRACT

The activated factor VII/tissue factor complex (FVIIa/TF) is known to play a key role in the formation of blood clots. Inhibition of this complex may lead to new antithrombotic drugs. A fluoropyridine-based series of FVIIa/TF inhibitors was discovered which utilized a diisopropylamino group for binding in the S2 and S3 binding pockets of the active site of the enzyme complex. In this series, an enhancement in binding affinity was observed by substitution at the 5-position of the hydroxybenzoic acid sidechain. An X-ray crystal structure indicates that amides at this position may increase inhibitor binding affinity through interactions with the S1'/S2' pocket.


Subject(s)
Enzyme Inhibitors/pharmacology , Factor VIIa/antagonists & inhibitors , Pyridines/pharmacology , Thromboplastin/antagonists & inhibitors , Crystallography, X-Ray , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Models, Molecular , Molecular Structure , Pyridines/chemical synthesis , Pyridines/chemistry , Structure-Activity Relationship
11.
Bioorg Med Chem Lett ; 15(21): 4752-6, 2005 Nov 01.
Article in English | MEDLINE | ID: mdl-16125385

ABSTRACT

The activated Factor VII/tissue factor complex (FVIIa/TF) plays a key role in the formation of blood clots. Inhibition of this complex may lead to new antithrombotic drugs. An X-ray crystal structure of a fluoropyridine-based FVIIa/TF inhibitor bound in the active site of the enzyme complex suggested that incorporation of substitution at the 5-position of the hydroxybenzoic acid side chain could lead to the formation of more potent inhibitors through interactions with the S1'/S2' pocket.


Subject(s)
Enzyme Inhibitors/chemical synthesis , Factor VIIa/chemistry , Fibrinolytic Agents/chemical synthesis , Pyridines/chemical synthesis , Thromboplastin/chemistry , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/chemistry , Factor VIIa/antagonists & inhibitors , Factor Xa Inhibitors , Fibrinolytic Agents/chemistry , Fibrinolytic Agents/pharmacology , Humans , Inhibitory Concentration 50 , Protein Binding , Prothrombin Time , Pyridines/chemistry , Structure-Activity Relationship , Thromboplastin/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...