Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Nat Commun ; 10(1): 1856, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015447

ABSTRACT

Bronchial premalignant lesions (PMLs) are precursors of lung squamous cell carcinoma, but have variable outcome, and we lack tools to identify and treat PMLs at risk for progression to cancer. Here we report the identification of four molecular subtypes of PMLs with distinct differences in epithelial and immune processes based on RNA-Seq profiling of endobronchial biopsies from high-risk smokers. The Proliferative subtype is enriched with bronchial dysplasia and exhibits up-regulation of metabolic and cell cycle pathways. A Proliferative subtype-associated gene signature identifies subjects with Proliferative PMLs from normal-appearing uninvolved large airway brushings with high specificity. In progressive/persistent Proliferative lesions expression of interferon signaling and antigen processing/presentation pathways decrease and immunofluorescence indicates a depletion of innate and adaptive immune cells compared with regressive lesions. Molecular biomarkers measured in PMLs or the uninvolved airway can enhance histopathological grading and suggest immunoprevention strategies for intercepting the progression of PMLs to lung cancer.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Bronchogenic/pathology , Gene Expression Regulation, Neoplastic/immunology , Lung Neoplasms/pathology , Precancerous Conditions/pathology , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/immunology , Biopsy , Bronchi/diagnostic imaging , Bronchi/immunology , Bronchi/pathology , Bronchoscopy , Carcinoma, Bronchogenic/genetics , Carcinoma, Bronchogenic/immunology , Carcinoma, Bronchogenic/prevention & control , Cohort Studies , Datasets as Topic , Disease Progression , Early Detection of Cancer/methods , Gene Expression Profiling , Gene Regulatory Networks/genetics , Gene Regulatory Networks/immunology , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/genetics , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Mass Screening/methods , Middle Aged , Precancerous Conditions/diagnostic imaging , Precancerous Conditions/genetics , Precancerous Conditions/immunology , RNA, Messenger/genetics , Respiratory Mucosa/cytology , Respiratory Mucosa/diagnostic imaging , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Sequence Analysis, RNA , T-Lymphocytes/immunology , Tomography, X-Ray Computed , Up-Regulation
2.
Clin Cancer Res ; 23(17): 5091-5100, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28533227

ABSTRACT

Purpose: Lung cancer is the leading cause of cancer-related death in the United States. The molecular events preceding the onset of disease are poorly understood, and no effective tools exist to identify smokers with premalignant lesions (PMLs) that will progress to invasive cancer. Prior work identified molecular alterations in the smoke-exposed airway field of injury associated with lung cancer. Here, we focus on an earlier stage in the disease process leveraging the airway field of injury to study PMLs and its utility in lung cancer chemoprevention.Experimental Design: Bronchial epithelial cells from normal appearing bronchial mucosa were profiled by mRNA-Seq from subjects with (n = 50) and without (n = 25) PMLs. Using surrogate variable and gene set enrichment analysis, we identified genes, pathways, and lung cancer-related gene sets differentially expressed between subjects with and without PMLs. A computational pipeline was developed to build and test a chemoprevention-relevant biomarker.Results: We identified 280 genes in the airway field associated with the presence of PMLs. Among the upregulated genes, oxidative phosphorylation was strongly enriched, and IHC and bioenergetics studies confirmed pathway findings in PMLs. The relationship between PMLs and squamous cell carcinomas (SCC) was also confirmed using published lung cancer datasets. The biomarker performed well predicting the presence of PMLs (AUC = 0.92, n = 17), and changes in the biomarker score associated with progression/stability versus regression of PMLs (AUC = 0.75, n = 51).Conclusions: Transcriptomic alterations in the airway field of smokers with PMLs reflect metabolic and early lung SCC alterations and may be leveraged to stratify smokers at high risk for PML progression and monitor outcome in chemoprevention trials. Clin Cancer Res; 23(17); 5091-100. ©2017 AACR.


Subject(s)
Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Precancerous Conditions/genetics , RNA, Messenger/genetics , Adult , Aged , Bronchi/metabolism , Bronchi/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/classification , Lung Neoplasms/pathology , Male , Middle Aged , Precancerous Conditions/pathology , Smokers , Smoking/genetics , Transcriptome/genetics
3.
Mol Cancer Ther ; 16(8): 1717-1726, 2017 08.
Article in English | MEDLINE | ID: mdl-28416604

ABSTRACT

Fibroblast growth factor receptor (FGFR) genetic alterations are frequently observed in cancer, suggesting that FGFR inhibition may be a promising therapy in patients harboring these lesions. Identification of predictive and pharmacodynamic biomarkers to select and monitor patients most likely to respond to FGFR inhibition will be the key to clinical development of this class of agents. Sensitivity to FGFR inhibition and correlation with FGFR pathway activation status were determined in molecularly annotated panels of cancer cell lines and xenograft models. Pathway inhibition in response to FGFR inhibitor treatment was assessed in cell lines (both in vitro and in vivo) and in samples from patients treated with the FGFR inhibitor JNJ-42756493 (erdafitinib). Frequency of FGFR aberrations was assessed in a panel of NSCLC, breast, prostate, ovarian, colorectal, and melanoma human tumor tissue samples. FGFR translocations and gene amplifications present in clinical specimens were shown to display potent transforming activity associated with constitutive pathway activation. Tumor cells expressing these FGFR activating mutants displayed sensitivity to the selective FGFR inhibitor erdafitinib and resulted in suppression of FGFR phosphorylation and downstream signal transduction. Clinically, patients receiving erdafitinib showed decreased Erk phosphorylation in tumor biopsies and elevation of serum phosphate. In a phase I study, a heavily pretreated bladder cancer patient with an FGFR3-TACC3 translocation experienced a partial response when treated with erdafitinib. This preclinical study confirmed pharmacodynamics and identified new predictive biomarkers to FGFR inhibition with erdafitinib and supports further clinical evaluation of this compound in patients with FGFR genetic alterations. Mol Cancer Ther; 16(8); 1717-26. ©2017 AACR.


Subject(s)
Oncogenes , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/genetics , Animals , Biomarkers, Tumor/metabolism , Male , Oncogene Proteins, Fusion/genetics , Pyrazoles/therapeutic use , Quinoxalines/therapeutic use , Rats, Nude , Tomography, X-Ray Computed , Urinary Bladder Neoplasms/diagnostic imaging , Urinary Bladder Neoplasms/drug therapy , Urinary Bladder Neoplasms/pathology
4.
Mol Cancer Ther ; 16(6): 1010-1020, 2017 06.
Article in English | MEDLINE | ID: mdl-28341788

ABSTRACT

Fibroblast growth factor (FGF) signaling plays critical roles in key biological processes ranging from embryogenesis to wound healing and has strong links to several hallmarks of cancer. Genetic alterations in FGF receptor (FGFR) family members are associated with increased tumor growth, metastasis, angiogenesis, and decreased survival. JNJ-42756493, erdafitinib, is an orally active small molecule with potent tyrosine kinase inhibitory activity against all four FGFR family members and selectivity versus other highly related kinases. JNJ-42756493 shows rapid uptake into the lysosomal compartment of cells in culture, which is associated with prolonged inhibition of FGFR signaling, possibly due to sustained release of the inhibitor. In xenografts from human tumor cell lines or patient-derived tumor tissue with activating FGFR alterations, JNJ-42756493 administration results in potent and dose-dependent antitumor activity accompanied by pharmacodynamic modulation of phospho-FGFR and phospho-ERK in tumors. The results of the current study provide a strong rationale for the clinical investigation of JNJ-42756493 in patients with tumors harboring FGFR pathway alterations. Mol Cancer Ther; 16(6); 1010-20. ©2017 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Discovery , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Lysosomes/metabolism , Male , Mice , Molecular Targeted Therapy , Phosphorylation , Protein Binding , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/administration & dosage , Pyrazoles/pharmacokinetics , Quinoxalines/administration & dosage , Quinoxalines/pharmacokinetics , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
5.
J Clin Oncol ; 33(30): 3401-8, 2015 Oct 20.
Article in English | MEDLINE | ID: mdl-26324363

ABSTRACT

PURPOSE: JNJ-42756493 is an orally administered pan-fibroblast growth factor receptor (FGFR) tyrosine kinase inhibitor. This first-in-human study evaluates the safety, pharmacokinetics, and pharmacodynamics and defines the recommended phase II dose (RP2D) of JNJ-42756493. PATIENTS AND METHODS: Eligible patients with advanced solid tumors received escalating doses of JNJ-42756493 from 0.5 to 12 mg administered continuously daily or JNJ-42756493 10 or 12 mg administered intermittently (7 days on/7 days off). RESULTS: Sixty-five patients were enrolled. The most common treatment-emergent adverse events included hyperphosphatemia (65%), asthenia (55%), dry mouth (45%), nail toxicity (35%), constipation (34%), decreased appetite (32%), and dysgeusia (31%). Twenty-seven patients (42%) experienced grade ≥ 3 treatment-emergent adverse events, and one dose-limiting toxicity of grade 3 ALT elevation was observed at 12 mg daily. Maximum-tolerated dose was not defined. Nine milligrams daily was considered as the initial RP2D; however, tolerability was improved with intermittent schedules, and 10 mg administered on a 7-days-on/7-days-off schedule was considered the final RP2D. Pharmacokinetics were linear, dose proportional, and predictable, with a half-life of 50 to 60 hours. Dose-dependent elevations in serum phosphate, a manifestation of pharmacodynamic effect, occurred in all patients starting at 4 mg daily. Among 23 response-evaluable patients with tumor FGFR pathway alterations, four confirmed responses and one unconfirmed partial response were observed in patients with glioblastoma and urothelial and endometrial cancer (all with FGFR2 or FGFR3 translocations); 16 patients had stable disease. CONCLUSION: JNJ-42756493 administered at 10 mg on a 7-days-on/7-days-off schedule achieved exposures at which clinical responses were observed, demonstrated pharmacodynamic biomarker activity, and had a manageable safety profile.


Subject(s)
Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Pyrazoles/administration & dosage , Quinoxalines/administration & dosage , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Administration, Oral , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Quinoxalines/adverse effects , Quinoxalines/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...