Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Expert Rev Anticancer Ther ; 22(8): 815-822, 2022 08.
Article in English | MEDLINE | ID: mdl-35835722

ABSTRACT

INTRODUCTION: Unresectable pleural mesothelioma is a poor prognosis disease. Improvement in overall survival (OS) has been shown with PEMETREXED combined with CISPLATIN. BEVACIZUMAB combined with chemotherapy is associated with an improvement in OS, compared to chemotherapy alone, but is not supported by health insurance everywhere. AREAS COVERED: Immune Checkpoint Inhibition (ICI) monotherapy seemed to be promising but is controversial. ICI combination showed significant results. NIVOLUMAB, an anti-Programmed-Death-receptor 1, associated with IPILIMUMAB, an anti-Cytotoxic-T-Lymphocyte-Associated-protein 4, was evaluated in two phase II trials and a phase III trial, recently published. This combination led to a significant benefit in survival in first line compared to chemotherapy (OS 18.1 months (95%CI (16.8-21.4)) vs 14.1 (95%CI (12.4-16.2) HR 0.74 (95%CI 0.6-0.91) p = 0.002). EXPERT OPINION: These results represent a big step in unresectable pleural mesothelioma. The benefit in non-epithelioid subtype is impressive (OS 18.1 months (95%CI 12.2-22.8) vs 8.8 months 95%CI (7.4-10.2) HR 0.46 (95%CI (0.31-0.68))). Benefit in epithelioid subtype (OS 18.7 months 95%CI (16.9-22) vs 16.5 95%CI (14.9-20.5) HR 0.86 95%CI (0.69-1.08)) is similar to the benefit of the combination of BEVACIZUMAB and chemotherapy. Identification of predictive biomarkers is needed to identify patients who are most likely to benefit from each therapeutic strategy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Mesothelioma, Malignant , Pleural Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Ipilimumab/therapeutic use , Mesothelioma, Malignant/drug therapy , Nivolumab/therapeutic use , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology
2.
Cancers (Basel) ; 13(14)2021 Jul 09.
Article in English | MEDLINE | ID: mdl-34298655

ABSTRACT

Despite the introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) to treat advanced lung cancer harboring EGFR-activating mutations, the prognosis remains unfavorable because of intrinsic and/or acquired resistance. We generated a new state-of-the-art mouse strain harboring the human EGFRT790M/L858R oncogene and MET overexpression (EGFR/MET strain) that mimics the MET amplification occurring in one out of five patients with EGFR-mutated lung cancer that relapsed after treatment with osimertinib, a third-generation anti-EGFR TKI. We found that survival was reduced in EGFR/MET mice compared with mice harboring only EGFRT790M/L858R (EGFR strain). Moreover, EGFR/MET-driven lung tumors were resistant to osimertinib, recapitulating the phenotype observed in patients. Conversely, as also observed in patients, the crizotinib (anti-MET TKI) and osimertinib combination improved survival and reduced tumor burden in EGFR/MET mice, further validating the model's value for preclinical studies. We also found that in EGFR/MET mice, MET overexpression negatively regulated EGFR activity through MIG6 induction, a compensatory mechanism that allows the coexistence of the two onco-genic events. Our data suggest that single EGFR or MET inhibition might not be a good therapeutic option for EGFR-mutated lung cancer with MET amplification, and that inhibition of both pathways should be the best clinical choice in these patients.

3.
JCI Insight ; 5(15)2020 08 06.
Article in English | MEDLINE | ID: mdl-32759499

ABSTRACT

Platinum-based chemotherapy in combination with immune-checkpoint inhibitors is the current standard of care for patients with advanced lung adenocarcinoma (LUAD). However, tumor progression evolves in most cases. Therefore, predictive biomarkers are needed for better patient stratification and for the identification of new therapeutic strategies, including enhancing the efficacy of chemotoxic agents. Here, we hypothesized that discoidin domain receptor 1 (DDR1) may be both a predictive factor for chemoresistance in patients with LUAD and a potential target positively selected in resistant cells. By using biopsies from patients with LUAD, KRAS-mutant LUAD cell lines, and in vivo genetically engineered KRAS-driven mouse models, we evaluated the role of DDR1 in the context of chemotherapy treatment. We found that DDR1 is upregulated during chemotherapy both in vitro and in vivo. Moreover, analysis of a cohort of patients with LUAD suggested that high DDR1 levels in pretreatment biopsies correlated with poor response to chemotherapy. Additionally, we showed that combining DDR1 inhibition with chemotherapy prompted a synergistic therapeutic effect and enhanced cell death of KRAS-mutant tumors in vivo. Collectively, this study suggests a potential role for DDR1 as both a predictive and prognostic biomarker, potentially improving the chemotherapy response of patients with LUAD.


Subject(s)
Adenocarcinoma of Lung/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Discoidin Domain Receptor 1/antagonists & inhibitors , Drug Resistance, Neoplasm , Lung Neoplasms/drug therapy , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Movement , Cell Proliferation , Cisplatin/administration & dosage , Discoidin Domain Receptor 1/genetics , Discoidin Domain Receptor 1/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Paclitaxel/administration & dosage , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Curr Oncol Rep ; 22(11): 107, 2020 08 15.
Article in English | MEDLINE | ID: mdl-32803433

ABSTRACT

PURPOSE OF REVIEW: Lung cancer is still the first cause of cancer-related deaths worldwide. The development of immune checkpoint inhibitors (ICI) has drastically changed the prognosis of some patients, but the rate of long responders does not exceed 20%. Moreover, ICIs are not adverse events-free and remain expensive. Therefore, predictive biomarkers of long-term benefit to ICI are required. RECENT FINDINGS: The two main fields being evaluated currently are PD-L1 expression and tumor mutational burden (TMB). The first one is the only one used in routine practice, and the second is being evaluated in phase 3 clinical trials. In addition, other biomarkers are being assessed as complex signatures, tumor-infiltrated lymphocytes, T cell receptor repertoire, or molecular profiling. The aim of this review is to summarize the current validated or promising biomarkers in lung cancer which could help to better select patients who will respond to ICI.


Subject(s)
Biomarkers, Tumor , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/diagnosis , B7-H1 Antigen , Clinical Trials, Phase III as Topic , Humans , Immunotherapy , Lung Neoplasms/drug therapy , Lymphocytes, Tumor-Infiltrating , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/drug therapy , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL
...