Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Future Oncol ; 18(27): 3085-3100, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36004638

ABSTRACT

Inflammation in the tumor microenvironment is a complicit and known carcinogenesis driver. Inhibition of IL-1ß, one of the most abundant and influential cytokines in the tumor microenvironment, may enhance the efficacy of PD-1. In a post-hoc analysis of phase III cardiovascular CANTOS trial, canakinumab, a monoclonal anti-IL-1ß antibody, significantly reduced lung cancer incidence. Immune checkpoint inhibition (ICI) is the standard of care in non-small-cell lung cancer. However, ICI efficacy is heavily impacted by programmed death ligand-1 (PD-L1) status. Most patients with non-small-cell lung cancer have low PD-L1 expression levels. Thus, combinational strategies are needed to improve ICI efficacy and expand its use. Here, we describe the preclinical and clinical evidence to support the combination of IL-1ß and PD-1 under investigation in the CANOPY program. The perioperative use of canakinumab with or without PD-1 inhibition in the CANOPY-N trial is described as a potential chemotherapy-free immunotherapy strategy.


IL-1ß is a small molecule involved in the spreading of cancer cells and scouting for cells that work against the body's protective inflammatory response. In a follow-up analysis of the CANTOS study, people with atherosclerosis who received canakinumab, a drug which limits the activity of IL-1ß in the body, were diagnosed with lung cancer less often than people who received an inactive substance. Immunotherapy is a treatment that can boost the natural defenses of the immune system, but how well it works varies from patient to patient. Recent efforts aim to understand whether blocking unhealthy inflammation with canakinumab and stimulating the body's protective system with immunotherapy at the same time could be an efficacious treatment for patients with lung cancer. Currently there are limited data from experiments in cell and animal models; however, data from the ongoing CANOPY-N clinical trial, which is investigating this treatment combination prior to surgery for patients with lung cancer, are expected by the first half of this year.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung/drug therapy , Clinical Trials, Phase III as Topic , Humans , Immune Checkpoint Inhibitors , Immunosuppression Therapy , Immunotherapy , Inflammation/drug therapy , Lung Neoplasms/epidemiology , Programmed Cell Death 1 Receptor , Tumor Microenvironment
2.
J Immunother Cancer ; 10(2)2022 02.
Article in English | MEDLINE | ID: mdl-35190375

ABSTRACT

BACKGROUND: The efficacy of atezolizumab (A) and/or bevacizumab (B) with carboplatin/paclitaxel (CP) chemotherapy was explored in the phase III, randomized IMpower150 study in patients with non-squamous non-small cell lung cancer (NSCLC) according to KRAS mutations (mKRAS) and co-occurring STK11, KEAP1, or TP53 mutations. METHODS: Mutation status was determined by circulating tumor DNA next-generation sequencing. Overall survival (OS) and progression-free survival (PFS) were analyzed in a mutation-evaluable intention-to-treat population (MEP; n=920) and SP263 (programmed cell death ligand 1 (PD-L1)) biomarker-evaluable population (n=774). RESULTS: Within the mKRAS population (24.5% of MEP), ABCP showed numerical improvements vs BCP in median OS (19.8 vs 9.9 months; HR 0.50; 95% CI 0.34 to 0.72) and PFS (8.1 vs 5.8 months; HR 0.42; 95% CI 0.29 to 0.61)-greater than with ACP (OS: 11.7 vs 9.9 months; HR 0.63; 95% CI 0.43 to 0.91; PFS: 4.8 vs 5.8 months; HR 0.80; 95% CI 0.56 to 1.13) vs BCP. Across PD-L1 subgroups in mKRAS patients, OS and PFS were longer with ABCP vs BCP, but OS with ACP was similar to BCP in PD-L1-low and PD-L1-negative subgroups. Conversely, in KRAS-WT patients, OS was longer with ACP than with ABCP or BCP across PD-L1 subgroups. KRAS was frequently comutated with STK11, KEAP1, and TP53; these subgroups conferred different prognostic outcomes. Within the mKRAS population, STK11 and/or KEAP1 mutations were associated with inferior OS and PFS across treatments compared with STK11-WT and/or KEAP1-WT. In mKRAS patients with co-occurring mSTK11 and/or mKEAP1 (44.9%) or mTP53 (49.3%), survival was longer with ABCP than with ACP or BCP. CONCLUSIONS: These analyses support previous findings of mutation of STK11 and/or KEAP1 as poor prognostic indicators. While clinical efficacy favored ABCP and ACP vs BCP in these mutational subgroups, survival benefits were greater in the mKRAS and KEAP1-WT and STK11-WT population vs mKRAS and mKEAP1 and mSTK11 population, suggesting both prognostic and predictive effects. Overall, these results suggest that atezolizumab combined with bevacizumab and chemotherapy is an efficacious first-line treatment in metastatic NSCLC subgroups with mKRAS and co-occurring STK11 and/or KEAP1 or TP53 mutations and/or high PD-L1 expression.


Subject(s)
AMP-Activated Protein Kinase Kinases/metabolism , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/drug therapy , Tumor Suppressor Protein p53/metabolism , Adult , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bevacizumab/pharmacology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Middle Aged , Mutation , Prognosis , Progression-Free Survival , Retrospective Studies
3.
Future Oncol ; 17(12): 1459-1472, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33648347

ABSTRACT

Canakinumab is a human IgGκ monoclonal antibody, with high affinity and specificity for IL-1ß. The Canakinumab Anti-Inflammatory Thrombosis Outcome Study (CANTOS) trial, evaluating canakinumab for cardiovascular disease, provided the first signal of the potential of IL-1ß inhibition on lung cancer incidence reduction. Here, we describe the rationale and design for CANOPY-N, a randomized Phase II trial evaluating IL-1ß inhibition with or without immune checkpoint inhibition as neoadjuvant treatment in patients with non-small-cell lung cancer. Patients with stage IB to IIIA non-small-cell lung cancer eligible for complete resection will receive canakinumab or pembrolizumab as monotherapy, or in combination. The primary end point is major pathological response by central review; secondary end points include overall response rate, major pathological response (local review), surgical feasibility rate and pharmacokinetics. Clinical trial registration: NCT03968419 (ClinicalTrials.gov).


Lay abstract A previous study showed that canakinumab reduced the risk of lung cancer in patients with heart disease. Canakinumab blocks an inflammatory protein called IL-1ß that is involved in cancer. Anti-cancer drugs used before surgery ('neo-adjuvant') can improve the success rate of surgery and may help prevent the cancer from returning. Neo-adjuvant trials help us understand how the drugs work and how they affect cancer. CANOPY-N (NCT03968419) is an ongoing randomized, exploratory, Phase II clinical trial testing canakinumab and pembrolizumab (a different cancer immunotherapy), alone or combined, for patients with early non-small-cell lung cancer. The study will test whether treatment can kill most cancer cells in the surgery sample ('major pathological response'). It will also investigate other effects on cancer biology, levels of molecules that measure possible clinical benefit ('biomarkers') and side effects.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Adolescent , Adult , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/immunology , Clinical Trials, Phase II as Topic , Female , Humans , Interleukin-1beta/antagonists & inhibitors , Lung/drug effects , Lung/pathology , Lung/surgery , Lung Neoplasms/diagnosis , Lung Neoplasms/immunology , Male , Neoadjuvant Therapy/methods , Neoplasm Staging , Pneumonectomy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Randomized Controlled Trials as Topic , Young Adult
5.
Sci Rep ; 6: 26591, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27225590

ABSTRACT

Promoter CpG methylation is a fundamental regulatory process of gene expression. TET proteins are active CpG demethylases converting 5-methylcytosine to 5-hydroxymethylcytosine, with loss of 5 hmC as an epigenetic hallmark of cancers, indicating critical roles of TET proteins in epigenetic tumorigenesis. Through analysis of tumor methylomes, we discovered TET1 as a methylated target, and further confirmed its frequent downregulation/methylation in cell lines and primary tumors of multiple carcinomas and lymphomas, including nasopharyngeal, esophageal, gastric, colorectal, renal, breast and cervical carcinomas, as well as non-Hodgkin, Hodgkin and nasal natural killer/T-cell lymphomas, although all three TET family genes are ubiquitously expressed in normal tissues. Ectopic expression of TET1 catalytic domain suppressed colony formation and induced apoptosis of tumor cells of multiple tissue types, supporting its role as a broad bona fide tumor suppressor. Furthermore, TET1 catalytic domain possessed demethylase activity in cancer cells, being able to inhibit the CpG methylation of tumor suppressor gene (TSG) promoters and reactivate their expression, such as SLIT2, ZNF382 and HOXA9. As only infrequent mutations of TET1 have been reported, compared to TET2, epigenetic silencing therefore appears to be the dominant mechanism for TET1 inactivation in cancers, which also forms a feedback loop of CpG methylation during tumorigenesis.


Subject(s)
DNA Methylation , Down-Regulation , Mixed Function Oxygenases/genetics , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Catalytic Domain , Cell Line, Tumor , CpG Islands , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Feedback, Physiological , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mixed Function Oxygenases/chemistry , Nerve Tissue Proteins/genetics , Protein Domains , Proto-Oncogene Proteins/chemistry , Transcription Factors/genetics
6.
Lancet Oncol ; 11(1): 75-84, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20129130

ABSTRACT

Population-based differences in toxicity and clinical outcome following treatment with anticancer drugs have an important effect on oncology practice and drug development. These differences arise from complex interactions between biological and environmental factors, which include genetic diversity affecting drug metabolism and the expression of drug targets, variations in tumour biology and host physiology, socioeconomic disparities, and regional preferences in treatment standards. Some well-known examples include the high prevalence of activating epidermal growth factor receptor (EGFR) mutations in pulmonary adenocarcinoma among northeast (China, Japan, Korea) and parts of southeast Asia (excluding India) non-smokers, which predict sensitivity to EGFR kinase inhibitors, and the sharp contrast between Japan and the west in the management and survival outcome of gastric cancer. This review is a critical overview of population-based differences in the four most prevalent cancers in the world: lung, breast, colorectal, and stomach cancer. Particular attention is given to the clinical relevance of such knowledge in terms of the individualisation of drug therapy and in the design of clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/epidemiology , Pharmacogenetics , Precision Medicine , Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Breast Neoplasms/epidemiology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/epidemiology , Environment , Female , Genetic Predisposition to Disease , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/epidemiology , Male , Patient Selection , Racial Groups , Risk Assessment , Risk Factors , Stomach Neoplasms/drug therapy , Stomach Neoplasms/epidemiology , Treatment Outcome
7.
Cancer Genet Cytogenet ; 153(1): 57-9, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15325095

ABSTRACT

Adenocarcinoma (AdC) of the lung represents a common histologic subtype of non-small cell lung cancer. While there is a rising incidence of AdC in nonsmoking women, information on the cytogenetic changes involved has been minimal to date. In the present study, spectral karyotyping analysis uncovered the genome-wide chromosomal aberrations in two AdC tumors derived from women who were lifelong nonsmokers. Simple and complex structural rearrangements were indicated. A ploidy status of hypertetraploidy was suggested in both cases, with recurring derivative translocations involving chromosome arms 3q, 8q, 12q, 15q, 22q, and Xq.


Subject(s)
Adenocarcinoma/genetics , Chromosome Aberrations , Lung Neoplasms/genetics , Spectral Karyotyping , Adenocarcinoma/pathology , Aged , Aneuploidy , Cell Transformation, Neoplastic/genetics , Female , Humans , Lung Neoplasms/pathology , Middle Aged , Translocation, Genetic , Tumor Cells, Cultured/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL
...