Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Signal ; 13(619)2020 02 18.
Article in English | MEDLINE | ID: mdl-32071169

ABSTRACT

High-grade serous ovarian carcinoma (HGSOC) is the most lethal gynecological cancer with few effective, targeted therapies. HGSOC tumors exhibit genomic instability with frequent alterations in the protein kinome; however, only a small fraction of the kinome has been therapeutically targeted in HGSOC. Using multiplexed inhibitor beads and mass spectrometry, we mapped the kinome landscape of HGSOC tumors from patients and patient-derived xenograft models. The data revealed a prevalent signature consisting of established HGSOC driver kinases, as well as several kinases previously unexplored in HGSOC. Loss-of-function analysis of these kinases in HGSOC cells indicated MRCKA (also known as CDC42BPA) as a putative therapeutic target. Characterization of the effects of MRCKA knockdown in established HGSOC cell lines demonstrated that MRCKA was integral to signaling that regulated the cell cycle checkpoint, focal adhesion, and actin remodeling, as well as cell migration, proliferation, and survival. Moreover, inhibition of MRCKA using the small-molecule BDP9066 decreased cell proliferation and spheroid formation and induced apoptosis in HGSOC cells, suggesting that MRCKA may be a promising therapeutic target for the treatment of HGSOC.


Subject(s)
Biomarkers, Tumor/antagonists & inhibitors , Cystadenocarcinoma, Serous/drug therapy , Myotonin-Protein Kinase/antagonists & inhibitors , Ovarian Neoplasms/drug therapy , Proteomics/methods , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Mass Spectrometry/methods , Molecular Targeted Therapy/methods , Myotonin-Protein Kinase/genetics , Myotonin-Protein Kinase/metabolism , Neoplasm Grading , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , RNA Interference , Signal Transduction/drug effects , Signal Transduction/genetics
2.
Cancer Discov ; 9(8): 1050-1063, 2019 08.
Article in English | MEDLINE | ID: mdl-31088841

ABSTRACT

Gilteritinib is a potent and selective FLT3 kinase inhibitor with single-agent clinical efficacy in relapsed/refractory FLT3-mutated acute myeloid leukemia (AML). In this context, however, gilteritinib is not curative, and response duration is limited by the development of secondary resistance. To evaluate resistance mechanisms, we analyzed baseline and progression samples from patients treated on clinical trials of gilteritinib. Targeted next-generation sequencing at the time of AML progression on gilteritinib identified treatment-emergent mutations that activate RAS/MAPK pathway signaling, most commonly in NRAS or KRAS. Less frequently, secondary FLT3-F691L gatekeeper mutations or BCR-ABL1 fusions were identified at progression. Single-cell targeted DNA sequencing revealed diverse patterns of clonal selection and evolution in response to FLT3 inhibition, including the emergence of RAS mutations in FLT3-mutated subclones, the expansion of alternative wild-type FLT3 subclones, or both patterns simultaneously. These data illustrate dynamic and complex changes in clonal architecture underlying response and resistance to mutation-selective tyrosine kinase inhibitor therapy in AML. SIGNIFICANCE: Comprehensive serial genotyping of AML specimens from patients treated with the selective FLT3 inhibitor gilteritinib demonstrates that complex, heterogeneous patterns of clonal selection and evolution mediate clinical resistance to tyrosine kinase inhibition in FLT3-mutated AML. Our data support the development of combinatorial targeted therapeutic approaches for advanced AML.See related commentary by Wei and Roberts, p. 998.This article is highlighted in the In This Issue feature, p. 983.


Subject(s)
Clonal Evolution/genetics , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , ras Proteins/metabolism , Adult , Aged , Aged, 80 and over , Aniline Compounds/pharmacology , Aniline Compounds/therapeutic use , Drug Resistance, Neoplasm/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Protein Kinase Inhibitors/therapeutic use , Pyrazines/pharmacology , Pyrazines/therapeutic use , Single-Cell Analysis , Young Adult , fms-Like Tyrosine Kinase 3/genetics , fms-Like Tyrosine Kinase 3/metabolism
3.
Cancer Biomark ; 22(4): 733-745, 2018.
Article in English | MEDLINE | ID: mdl-29945345

ABSTRACT

To find out if the T cell repertoire is efficiently and specifically provoked in patients with breast cancer, we have investigated the clonotypes of main T cell subsets (based on Vß-Chain) in tumor draining lymph nodes. CD4+ helper, CD8+ cytotoxic and (CD4+CD127dimCD25+) regulatory T cells, were negatively selected, and isolated, from lymph node mononuclear cells of 14 untreated patients with breast cancer. Four non-malignant patients, who underwent surgical operation, were also recruited as the control group. Based on sequences and new nomenclature of the T cell Receptor ß Variables (TRBVs) available in the international ImMunoGeneTics (IMGT) database, 28 TRBV specific forward primers and two TRB Constant region (TRBC) specific reverse primers were developed to amplify all functional alleles. Fluorescent-labeled PCR products were then run on an ABI PRISM 310 Genetic-Analyzer. The data was analyzed by GeneMapper software version 3.1. Clonotype analysis suggested that activated T cells are present in breast cancer. More TRBV usage were detected among CD4+ helper and regulatory subsets, with Gaussian-like pattern in the majority of functional TRBV families; whereas CD8+ cytotoxic T cells showed oligoclonality in almost all TRBV families with one or two dominant peaks in each family. Similar pattern in some of these TRBVs were also observed among controls. Having no expression or polyclonality in the controls, the oligoclonal pattern observed in the TRBV18, however, appears to be specific to breast cancer patients. This phenomenon may reflect the existence of new antigenic stimulation(s) in BC patients, preferentially activating those clones of T cells that express TRBV18.


Subject(s)
Breast Neoplasms, Male/genetics , Breast Neoplasms/genetics , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocyte Subsets/immunology , Adult , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms, Male/pathology , CD4-Positive T-Lymphocytes/immunology , Clonal Evolution/genetics , Female , Humans , Lymph Nodes/immunology , Male , Receptors, Antigen, T-Cell, alpha-beta/immunology
4.
Cancer Biol Ther ; 18(9): 694-704, 2017 Sep 02.
Article in English | MEDLINE | ID: mdl-28886275

ABSTRACT

Colorectal cancer (CRC) is a leading cause of cancer-related deaths in the United States. We analyzed 26 MSI-High and 558 non-MSI-High CRC tumors. BRCA2 mutations were highly enriched (50%) in MSI-High CRC. Immunohistochemistry showed that BRCA2-mutated MSI-High CRC had high c-MET (64%) expression compared with BRCA-WT (17%). We hypothesized a mechanistic link between BRCA2-deficiency and c-MET overexpression and synergistic interaction between drugs that treat BRCA-deficient tumors (mitomycin C (MMC) or PARP inhibitors) and c-MET inhibitors (crizotinib). We tested CRC cell lines for sensitivity to MMC plus crizotinib or other drug combinations including PARP-inhibitors. Combined treatment of tumor cells with crizotinib and MMC led to increased apoptosis as compared with each drug alone. Additionally, combination treatment with increasing concentrations of both drugs demonstrated a synergistic anti-cancer effect (CI = 0.006-0.74). However, we found no evidence for c-MET upregulation upon effective BRCA2 knockdown in tumor cells -/+DNA damage. Although we found no mechanistic link between BRCA2 deficiency and c-MET overexpression, c-MET is frequently overexpressed in CRC and BRCA2 is mutated especially in MSI-H CRC. The combination of crizotinib with MMC appeared synergistic regardless of MSI or BRCA2 status. Using an in-vivo CRC xenograft model we found reduced tumor growth with combined crizotinib and MMC therapy (p = 0.0088). Our preclinical results support clinical testing of the combination of MMC and crizotinib in advanced CRC. Targeting cell survival mediated by c-MET in combination with targeting DNA repair may be a reasonable strategy for therapy development in CRC or other cancers.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Mitomycin/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyridines/pharmacology , Alleles , Animals , Cell Line, Tumor , Cell Survival/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Crizotinib , Disease Models, Animal , Drug Synergism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Gene Silencing , Genes, BRCA2 , Humans , Immunohistochemistry , Mice , Mutation , Proto-Oncogene Proteins c-met , Xenograft Model Antitumor Assays
5.
Oncotarget ; 8(25): 39945-39962, 2017 Jun 20.
Article in English | MEDLINE | ID: mdl-28591715

ABSTRACT

Deficient mismatch repair (MMR) and microsatellite instability (MSI) contribute to ~15% of colorectal cancer (CRCs). We hypothesized MSI leads to mutations in DNA repair proteins including BRCA2 and cancer drivers including EGFR. We analyzed mutations among a discovery cohort of 26 MSI-High (MSI-H) and 558 non-MSI-H CRCs profiled at Caris Life Sciences. Caris-profiled MSI-H CRCs had high mutation rates (50% vs 14% in non-MSI-H, P < 0.0001) in BRCA2. Of 1104 profiled CRCs from a second cohort (COSMIC), MSH2/MLH1-mutant CRCs showed higher mutation rates in BRCA2 compared to non-MSH2/MLH1-mutant tumors (38% vs 6%, P < 0.0000001). BRCA2 mutations in MSH2/MLH1-mutant CRCs included 75 unique mutations not known to occur in breast or pancreatic cancer per COSMIC v73. Only 5 deleterious BRCA2 mutations in CRC were previously reported in the BIC database as germ-line mutations in breast cancer. Some BRCA2 mutations were predicted to disrupt interactions with partner proteins DSS1 and RAD51. Some CRCs harbored multiple BRCA2 mutations. EGFR was mutated in 45.5% of MSH2/MLH1-mutant and 6.5% of non-MSH2/MLH1-mutant tumors (P < 0.0000001). Approximately 15% of EGFR mutations found may be actionable through TKI therapy, including N700D, G719D, T725M, T790M, and E884K. NTRK gene mutations were identified in MSH2/MLH1-mutant CRC including NTRK1 I699V, NTRK2 P716S, and NTRK3 R745L. Our findings have clinical relevance regarding therapeutic targeting of BRCA2 vulnerabilities, EGFR mutations or other identified oncogenic drivers such as NTRK in MSH2/MLH1-mutant CRCs or other tumors with mismatch repair deficiency.


Subject(s)
BRCA2 Protein/genetics , Colorectal Neoplasms/genetics , ErbB Receptors/genetics , Mutation , Receptor, trkA/genetics , Receptor, trkB/genetics , Receptor, trkC/genetics , BRCA2 Protein/chemistry , Cohort Studies , DNA Mismatch Repair/genetics , ErbB Receptors/chemistry , Gene Frequency , Humans , Microsatellite Instability , Models, Molecular , MutL Protein Homolog 1/genetics , MutS Homolog 2 Protein/genetics , Protein Domains , Receptor, trkA/chemistry , Receptor, trkB/chemistry , Receptor, trkC/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...