Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
1.
Proc Natl Acad Sci U S A ; 111(3): 1114-9, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395800

ABSTRACT

The underpinnings of STAT3 hyperphosphorylation resulting in enhanced signaling and cancer progression are incompletely understood. Loss-of-function mutations of enzymes that dephosphorylate STAT3, such as receptor protein tyrosine phosphatases, which are encoded by the PTPR gene family, represent a plausible mechanism of STAT3 hyperactivation. We analyzed whole exome sequencing (n = 374) and reverse-phase protein array data (n = 212) from head and neck squamous cell carcinomas (HNSCCs). PTPR mutations are most common and are associated with significantly increased phospho-STAT3 expression in HNSCC tumors. Expression of receptor-like protein tyrosine phosphatase T (PTPRT) mutant proteins induces STAT3 phosphorylation and cell survival, consistent with a "driver" phenotype. Computational modeling reveals functional consequences of PTPRT mutations on phospho-tyrosine-substrate interactions. A high mutation rate (30%) of PTPRs was found in HNSCC and 14 other solid tumors, suggesting that PTPR alterations, in particular PTPRT mutations, may define a subset of patients where STAT3 pathway inhibitors hold particular promise as effective therapeutic agents.


Subject(s)
Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/metabolism , Mutation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , STAT3 Transcription Factor/metabolism , Amino Acid Sequence , Cell Line, Tumor , Cell Survival , Computer Simulation , HEK293 Cells , Humans , Immunohistochemistry , Models, Molecular , Molecular Sequence Data , Phosphorylation , Protein Structure, Tertiary , Proteome , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Transfection
2.
JAMA Oncol ; 1(2): 238-44, 2015 May.
Article in English | MEDLINE | ID: mdl-26181029

ABSTRACT

IMPORTANCE: Randomized clinical trials demonstrate no benefit for epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors in unselected patients with head and neck squamous cell carcinoma (HNSCC). However, a patient with stage IVA HNSCC received 13 days of neoadjuvant erlotinib and experienced a near-complete histologic response. OBJECTIVE: To determine a mechanism of exceptional response to erlotinib therapy in HNSCC. DESIGN, SETTING, AND PARTICIPANTS: Single patient with locally advanced HNSCC who received erlotinib monotherapy in a window-of-opportunity clinical trial (patients scheduled to undergo primary cancer surgery are treated briefly with an investigational agent). Whole-exome sequencing of pretreatment tumor and germline patient samples was performed at a quaternary care academic medical center, and a candidate somatic variant was experimentally investigated for mediating erlotinib response. INTERVENTION: A brief course of erlotinib monotherapy followed by surgical resection. MAIN OUTCOMES AND MEASURES: Identification of pretreatment tumor somatic alterations that may contribute to the exceptional response to erlotinib. Hypotheses were formulated regarding enhanced erlotinib response in preclinical models harboring the patient tumor somatic variant MAPK1 E322K following the identification of tumor somatic variants. RESULTS: No EGFR alterations were observed in the pretreatment tumor DNA. Paradoxically, the tumor harbored an activating MAPK1 E322K mutation (allelic fraction 0.13), which predicts ERK activation and erlotinib resistance in EGFR-mutant lung cancer. The HNSCC cells with MAPK1 E322K exhibited enhanced EGFR phosphorylation and erlotinib sensitivity compared with wild-type MAPK1 cells. CONCLUSIONS AND RELEVANCE: Selective erlotinib use in HNSCC may be informed by precision oncology approaches.


Subject(s)
Antineoplastic Agents/administration & dosage , Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Erlotinib Hydrochloride/administration & dosage , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/genetics , Mitogen-Activated Protein Kinase 1/genetics , Protein Kinase Inhibitors/administration & dosage , Tongue Neoplasms/drug therapy , Tongue Neoplasms/genetics , Adult , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Biopsy , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/pathology , Chemotherapy, Adjuvant , DNA Mutational Analysis , Drug Administration Schedule , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Genetic Predisposition to Disease , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/pathology , Humans , Male , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Targeted Therapy , Mutation , Neoadjuvant Therapy , Neoplasm Staging , Phenotype , Phosphorylation , Predictive Value of Tests , Randomized Controlled Trials as Topic , Remission Induction , Squamous Cell Carcinoma of Head and Neck , Time Factors , Tongue Neoplasms/enzymology , Tongue Neoplasms/pathology , Treatment Outcome
3.
Cancer Discov ; 3(7): 761-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23619167

ABSTRACT

Genomic findings underscore the heterogeneity of head and neck squamous cell carcinoma (HNSCC). Identification of mutations that predict therapeutic response would be a major advance. We determined the mutationally altered, targetable mitogenic pathways in a large HNSCC cohort. Analysis of whole-exome sequencing data from 151 tumors revealed the phosphoinositide 3-kinase (PI3K) pathway to be the most frequently mutated oncogenic pathway (30.5%). PI3K pathway-mutated HNSCC tumors harbored a significantly higher rate of mutations in known cancer genes. In a subset of human papillomavirus-positive tumors, PIK3CA or PIK3R1 was the only mutated cancer gene. Strikingly, all tumors with concurrent mutation of multiple PI3K pathway genes were advanced (stage IV), implicating concerted PI3K pathway aberrations in HNSCC progression. Patient-derived tumorgrafts with canonical and noncanonical PIK3CA mutations were sensitive to an mTOR/PI3K inhibitor (BEZ-235), in contrast to PIK3CA-wild-type tumorgrafts. These results suggest that PI3K pathway mutations may serve as predictive biomarkers for treatment selection.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Squamous Cell/genetics , Head and Neck Neoplasms/genetics , Phosphatidylinositol 3-Kinases/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Head and Neck Neoplasms/pathology , High-Throughput Nucleotide Sequencing , Humans , Imidazoles/administration & dosage , Mutation , Neoplasm Staging , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Quinolines/administration & dosage , Squamous Cell Carcinoma of Head and Neck
SELECTION OF CITATIONS
SEARCH DETAIL