Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Thorac Cancer ; 12(9): 1366-1372, 2021 05.
Article in English | MEDLINE | ID: mdl-33710780

ABSTRACT

BACKGROUND: The geriatric nutritional risk index (GNRI) is a simple and useful marker for predicting prognosis and treatment efficacy among patients with various cancers. However, to the best of our knowledge, there are no previous reports regarding the prognostic value of GNRI among patients with non-small cell lung cancer (NSCLC) who were treated with immune checkpoint inhibitors (ICIs). METHODS: We retrospectively evaluated 85 patients with previously treated advanced NSCLC who were administered ICIs at Shinshu University Hospital between February 2016 and October 2020. Progression-free survival (PFS) and overall survival (OS) were compared between groups with high (≥89.5) and low (<89.5) GNRI values. We used univariate and multivariate Cox regression analyses to identify prognostic factors that were associated with PFS and OS. RESULTS: The high and low GNRI groups included 61 and 24 patients, respectively. Relative to the low GNRI group, the high GNRI group had significantly longer median PFS (3.7 vs. 2.4 months, p = 0.041) and significantly longer median OS (14.2 vs. 6.1 months, p = 0.008). Multivariate analyses revealed that independent predictors of favorable OS were high GNRI, performance status of 0-1, and age of ≥70 years. The high GNRI group was significantly more likely to undergo subsequent therapy after immunotherapy (68.6 vs. 33.3%, p = 0.008). CONCLUSIONS: The present study revealed that high GNRI was associated with good outcomes among patients with previously treated NSCLC who were treated with ICIs.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Immunotherapy/methods , Lung Neoplasms/diet therapy , Lung Neoplasms/drug therapy , Nutritional Status/physiology , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Female , Humans , Lung Neoplasms/mortality , Male , Prognosis , Risk Factors
2.
Lung Cancer ; 151: 8-15, 2021 01.
Article in English | MEDLINE | ID: mdl-33278671

ABSTRACT

OBJECTIVES: Accumulating evidence indicates anti-diabetic drug metformin has anti-cancer effect by controlling cancer metabolism. We evaluated whether addition of metformin to chemotherapy improved survival of lung cancer patients. MATERIALS AND METHODS: This randomized phase II study enrolled 164 patients with chemo-native, EGFR-ALK wild-type, stage IIIB/IV non-small-cell lung cancer (NSCLC). Patients were randomized to receive chemotherapy either with metformin (1000 mg twice daily) or alone every 3 weeks for six cycles. The patients received gemcitabine (1000 mg/m2) on days 1 and 8 and carboplatin (5 area under the curve) on day 1. Exploratory studies included serum metabolic panels, positron-emission tomography (PET) imaging, and genetic mutation tests for metabolism-related genes. RESULTS: Metformin group showed no significant difference in the risk of progression and death compared to control group (progression: hazard ratio [HR] = 1.01 [95% confidence interval (CI) = 0.72 - 1.42], P = 0.935; death: HR = 0.95 [95% CI = 0.67-1.34], P = 0.757). Squamous cell carcinoma (SqCC) had significantly higher fluorodeoxyglucose (FDG) uptake on baseline PET image than non-SqCC NSCLC (P = 0.004). In the SqCC with high FDG uptake, the addition of metformin significantly decreased the risk of progression and death (progression: HR = 0.31 [95% CI = 0.12-0.78], P = 0.013; death: HR = 0.42 [95% CI = 0.18-0.94], P = 0.035). The HDL-cholesterol level was significantly increased after the treatment in metformin group compared to control group (P = 0.011). The metformin group showed no survival benefit in the patients with hyperinsulinemia or patients whose insulin level was decreased after treatment. CONCLUSIONS: Addition of metformin to chemotherapy provided no survival benefit in unselected NSCLC patients. However, it significantly improved the survival of the selected patients with SqCC showing high FDG uptake. It suggests metformin shows the synergistic anti-tumor effect in the tumor which are highly dependent on glucose metabolism.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Metformin , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Diet , Disease-Free Survival , Humans , Lung Neoplasms/diet therapy , Lung Neoplasms/drug therapy , Metformin/therapeutic use
3.
Support Care Cancer ; 29(6): 3357-3365, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33128137

ABSTRACT

PURPOSE: The study aimed to determine the poor nutritional status, related factors, and its effect on the prognosis of patients with locally advanced and advanced stage lung cancer. METHODS: The study consisted of 539 patients, 412 (76.4%) of whom were non-small cell lung cancer (NSCLC), and 127 (23.6%) were small cell lung cancer (SCLC). The nutritional status of the patients was evaluated by the Controlling Nutritional Status (CONUT) and Prognostic Nutritional Index (PNI). Poor nutritional status was diagnosed with the CONUT score of ≥ 2 and PNI of ≥the median value. The factors related to nutritional status were determined using a multivariate logistic regression model. The effect of poor nutritional status on survival was calculated by Cox regression analysis. RESULTS: The median age was 64 years (29-87). Poor nutritional status was found in 56.4% (57.8% for NSCLC and 52.0% for SCLC) and 49.2% (51.5% for NSCLC and 41.7% for SCLC) of patients according to CONUT and PNI, respectively. The factors associated with poor nutritional status according to CONUT were age, gender, KPS < 80, and BMI < 18.5 for NSCLC and KPS for SCLC. According to PNI, only KPS < 80 was associated with poor nutritional status by the multivariate logistic regression model. The median overall survival significantly decreased with poor nutritional status according to CONUT and PNI in NSCLC (p < 0.001 and p < 0.001, respectively) and in SCLC (p = 0.05 and p = 0.007, respectively). CONCLUSION: Poor nutritional status is a common factor associated with poor prognosis in patients with locally advanced and advanced stage lung cancer. Patients should be screened for nutritional status and supported.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Lung Neoplasms/diet therapy , Nutritional Status/physiology , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/mortality , Female , Humans , Lung Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Prognosis , Retrospective Studies , Survival Analysis
4.
Aging (Albany NY) ; 12(9): 8001-8015, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32375124

ABSTRACT

Acquired resistance of osimertinib is encountered in clinic treatment of non-small cell lung cancer (NSCLC). However, the molecular mechanisms of osimertinib resistance are not fully revealed. This study aimed to investigate the roles of exosomes in delivering osimertinib resistance in NSCLC. Exosomes were successfully isolated. LncRNA sequencing identified a total of 123 differentially expressed lncRNAs, including 45 upregulated lncRNAs and 78 downregulated lncRNAs. The relative expression level of lncRNA MSTRG.292666.16 was significantly upregulated in osimertinib-resistant plasma, osimertinib-resistant H1975R cells and their derived exosomes, compared with those in osimertinib- sensitive plasma, H1975 cells and exosomes (P < 0.05). Besides, osimertinib-resistant exosomes could regulate gene expressions induced by osimertinib, including miRNA-21, miRNA-125b, TGFß, ARF6 and c-Kit. Osimertinib-resistant exosomes could be taken up by osimertinib-sensitive H1975 cells and resulting in osimertinib-resistance in vivo. Knockdown of lncRNA MSTRG.292666.16 decreased osimertinib resistance of H1975R cells. Our results suggest that exosomal lncRNA MSTRG.292666.16 might be associated with osimertinib resistance in NSCLC.


Subject(s)
Acrylamides/pharmacology , Aniline Compounds/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Drug Resistance, Neoplasm/genetics , Exosomes/genetics , Gene Expression Regulation, Neoplastic , RNA, Long Noncoding/genetics , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Humans , Lung Neoplasms/genetics , MicroRNAs/genetics , RNA, Long Noncoding/biosynthesis
5.
ACS Chem Biol ; 15(1): 158-170, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31874028

ABSTRACT

Selective toxicity among cancer cells of the same lineage is a hallmark of targeted therapies. As such, identifying compounds that impair proliferation of a subset of non-small-cell lung cancer (NSCLC) cell lines represents one strategy to discover new drugs for lung cancer. Previously, phenotypic screens of 202 103 compounds led to the identification of 208 selective NSCLC toxins ( McMillan , E. A. , Cell , 2018 , 173 , 864 ). The mechanism of action for the majority of these compounds remains unknown. Here, we discovered the target for a series of quinazoline diones (QDC) that demonstrate selective toxicity among 96 NSCLC lines. Using photoreactive probes, we found that the QDC binds to both mitochondrial complex I of the electron transport chain and hydroxyacyl CoA dehydrogenase subunit alpha (HADHA), which catalyzes long-chain fatty acid oxidation. Inhibition of complex I is the on-target activity for QDC, while binding to HADHA is off-target. The sensitivity profile of the QDC across NSCLC lines correlated with the sensitivity profiles of six additional structurally distinct compounds. The antiproliferative activity of these compounds is also the consequence of binding to mitochondrial complex I, reflecting significant structural diversity among complex I inhibitors. Small molecules targeting complex I are currently in clinical development for the treatment of cancer. Our results highlight complex I as a target in NSCLC and report structurally diverse scaffolds that inhibit complex I.


Subject(s)
Antineoplastic Agents/chemistry , Carcinoma, Non-Small-Cell Lung/diet therapy , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Lung Neoplasms/diet therapy , Quinazolinones/chemistry , 3-Hydroxyacyl CoA Dehydrogenases/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Fatty Acids/metabolism , Gene Knockout Techniques , Humans , Mitochondrial Trifunctional Protein, alpha Subunit/genetics , Mitochondrial Trifunctional Protein, alpha Subunit/metabolism , Molecular Structure , Molecular Targeted Therapy , Oxidation-Reduction , Oxygen Consumption , Protein Binding , Protein Conformation , Proteomics , Quinazolinones/pharmacology , Structure-Activity Relationship , Substrate Specificity
6.
BMC Cancer ; 19(1): 528, 2019 May 31.
Article in English | MEDLINE | ID: mdl-31151425

ABSTRACT

BACKGROUND: Most advanced elderly cancer patients experience fatigue, anorexia, and declining physical function due to cancer cachexia, for which effective interventions have not been established. We performed a phase I study of a new nonpharmacological multimodal intervention called the nutritional and exercise treatment for advanced cancer (NEXTAC) program and reported the excellent feasibility of and compliance with this program in elderly patients with advanced cancer who were at risk for cancer cachexia. We report here the background, hypothesis, and design of the next-step multicenter, randomized phase II study to evaluate the efficacy of the program, the NEXTAC-TWO study. METHODS: Patients with chemo-naïve advanced non-small cell lung cancer or pancreatic cancer, age ≥ 70 years, performance status ≤2, with adequate organ function and without disability according to the modified Katz index will be eligible. In total, 130 participants will be recruited from 15 Japanese institutions and will be randomized into either the intervention group or a control group. Computer-generated random numbers are allocated to each participant. Stratification factors include performance status (0 to 1 vs. 2), site of primary cancer (lung vs. pancreas), stage (III vs. IV), and type of chemotherapy (cytotoxic vs. others). Interventions and assessment will be performed 4 times every 4 ± 2 weeks from the date of randomization. Interventions will consist of nutritional counseling, nutritional supplements (rich in branched-chain amino acids), and a home-based exercise program. The exercise program will include low-intensity daily muscle training and lifestyle education to promote physical activity. The primary endpoint is disability-free survival. It is defined as the period from the date of randomization to the date of developing disability or death due to any cause. This trial also plans to evaluate the improvements in nutritional status, physical condition, quality of life, activities of daily living, overall survival, and safety as secondary endpoints. Enrollment began in August 2017. The study results will demonstrate the efficacy of multimodal interventions for elderly cancer patients and their application for the maintenance of physical and nutritional conditions in patients with cancer cachexia. This work is supported by a grant-in-aid from the Japan Agency for Medical Research and Development. DISCUSSION: This is the first randomized trial to evaluate the efficacy and safety of a multimodal intervention specific for elderly patients with advanced cancer. TRIAL REGISTRATION: Registered at August 23, 2017. Registry number: UMIN000028801 .


Subject(s)
Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Pancreatic Neoplasms/therapy , Aged , Aged, 80 and over , Cachexia/epidemiology , Cachexia/physiopathology , Cachexia/prevention & control , Cachexia/therapy , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Protocols , Clinical Trials, Phase II as Topic , Combined Modality Therapy , Exercise Therapy , Humans , Japan , Lung Neoplasms/diet therapy , Lung Neoplasms/pathology , Pancreatic Neoplasms/diet therapy , Pancreatic Neoplasms/pathology , Quality of Life , Randomized Controlled Trials as Topic
7.
Biomed Res Int ; 2019: 2585743, 2019.
Article in English | MEDLINE | ID: mdl-31119158

ABSTRACT

PURPOSE: Leptin is a nutritional cytokine encoded by the obesity gene whose concentration in the tumor microenvironment is closely related to the occurrence and progression of cancer. However, previous evidence has suggested that there is no clear relationship between serum leptin concentrations and lung cancer progression. Cancer-associated fibroblasts (CAFs), the most abundant component of the tumor microenvironment in a variety of solid tumors, were recently reported to produce leptin. Therefore, it was inferred that leptin is most likely to affect non-small-cell lung cancer (NSCLC) through an autocrine and paracrine mechanism. In the current study, we investigated the paracrine effect and mechanism of leptin produced by CAFs on NSCLC by establishing a novel in vitro cell coculture system. METHODS: A noncontact coculture device was designed and made by 3D printing. CAFs and paired normal lung fibroblasts (NLFs) from 5 patients were successfully isolated and cocultured with two NSCLC cell lines in a coculture system. The background expression of leptin was detected by western blot. The in situ expression of leptin and its receptor (Ob-R) in NSCLC tissues and paired normal lung tissues was analyzed by immunohistochemistry. Furthermore, we downregulated the expression of leptin in CAFs and assessed changes in its promotion on NSCLC cells in the coculture system. Finally, changes in the phosphorylation of ERK1/2 and AKT were examined to investigate the molecular mechanisms responsible for the paracrine promotion of NSCLC cells by leptin. RESULTS: Leptin was overexpressed in nearly all five primary CAF lines compared with its expression in paired NLFs. IHC staining showed that the expression of leptin was high in NSCLC cells, slightly lower in CAF, and negative in normal lung tissue. Ob-R was strongly expressed in NSCLC cells. The ability of A549 and H1299 cells to proliferate and migrate was enhanced by high leptin levels in both the cocultured fibroblasts and the culture medium. Furthermore, western blot assays suggested that the MAPK/ERK1/2 and PI3K/AKT signaling pathways were activated by leptin produced by CAFs, which demonstrated that the functions of paracrine leptin in NSCLC are as those of the serum leptin to other cancers. CONCLUSION: Leptin produced by CAF promotes proliferation and migration of NSCLC cells probably via PI3K/AKT and MAPK/ERK1/2 signaling pathways in a paracrine manner.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Cell Proliferation/drug effects , Leptin/pharmacology , Paracrine Communication/drug effects , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Coculture Techniques , Humans , Leptin/genetics , Leptin/metabolism , MAP Kinase Signaling System/drug effects , Paracrine Communication/genetics , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Tumor Microenvironment/drug effects
8.
Clin Cancer Res ; 24(17): 4089-4097, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30018118

ABSTRACT

Purpose: Higher serum 25-hydroxyvitamin D (25(OH)D) levels are reportedly associated with better survival in early-stage non-small cell lung cancer (NSCLC). Therefore, whether vitamin D supplementation can improve the prognosis of patients with NSCLC was examined (UMIN000001869).Patients and Methods: A randomized, double-blind trial comparing vitamin D supplements (1,200 IU/day) with placebo for 1 year after operation was conducted. The primary and secondary outcomes were relapse-free survival (RFS) and overall survival (OS), respectively. Prespecified subgroup analyses were performed with stratification by stage (early vs. advanced), pathology (adenocarcinoma vs. others), and 25(OH)D levels (low, <20 ng/mL vs. high, ≥20 ng/mL). Polymorphisms of vitamin D receptor (VDR) and vitamin D-binding protein (DBP) and survival were also examined.Results: Patients with NSCLC (n = 155) were randomly assigned to receive vitamin D (n = 77) or placebo (n = 78) and followed for a median of 3.3 years. Relapse and death occurred in 40 (28%) and 24 (17%) patients, respectively. In the total study population, no significant difference in either RFS or OS was seen with vitamin D compared with the placebo group. However, by restricting the analysis to the subgroup with early-stage adenocarcinoma with low 25(OH)D, the vitamin D group showed significantly better 5-year RFS (86% vs. 50%, P = 0.04) and OS (91% vs. 48%, P = 0.02) than the placebo group. Among the examined polymorphisms, DBP1 (rs7041) TT and CDX2 (rs11568820) AA/AG genotypes were markers of better prognosis, even with multivariate adjustment.Conclusions: In patients with NSCLC, vitamin D supplementation may improve survival of patients with early-stage lung adenocarcinoma with lower 25(OH)D levels. Clin Cancer Res; 24(17); 4089-97. ©2018 AACR.


Subject(s)
Adenocarcinoma of Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/diet therapy , Neoplasm Recurrence, Local/diet therapy , Vitamin D/analogs & derivatives , Vitamin D/administration & dosage , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Aged , CDX2 Transcription Factor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , DNA-Binding Proteins/genetics , Dietary Supplements , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Polymorphism, Genetic , Prognosis , Receptors, Calcitriol/genetics , Transcription Factors/genetics , Vitamin D/adverse effects
9.
Semin Ultrasound CT MR ; 39(3): 308-313, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29807641

ABSTRACT

The identification of genetic mutations known as oncogenic driver mutations that lead to the growth and survival of cancer cells has been an important advance in the field of oncology. Treatment in advanced non-small-cell lung cancer (NSCLC) has transitioned from a more general approach to a more personalized approach based on genetic mutations of the cancer itself. Common mutations detected in patients with advanced NSCLC include mutations of epidermal growth factor receptor and anaplastic lymphoma kinase (ALK). Targeted therapies are aimed at the products of these gene mutations and include erlotinib (used in epidermal growth factor receptor mutant NSCLC) and crizotinib (used in anaplastic lymphoma kinase positive NSCLC). In this review, we discuss common genetic mutations in advanced NSCLC, the role of targeted therapies, and imaging findings that can be associated with various genetic mutations.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/diet therapy , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/drug therapy , Tomography, X-Ray Computed/methods , Anaplastic Lymphoma Kinase , Antineoplastic Agents, Immunological/therapeutic use , Arginine Vasopressin/analogs & derivatives , Bevacizumab/therapeutic use , Crizotinib , ErbB Receptors/genetics , Erlotinib Hydrochloride/therapeutic use , Humans , Lung/diagnostic imaging , Mutation/genetics
10.
Anticancer Res ; 37(9): 5141-5145, 2017 09.
Article in English | MEDLINE | ID: mdl-28870946

ABSTRACT

BACKGROUND: The acidic tumor microenvironment is associated with progression of cancers. The purpose of this study was to investigate the association between an alkaline diet and the effect of epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor (TKI) in non-small cell lung cancer (NSCLC) patients. PATIENTS AND METHODS: Eleven advanced or recurrent NSCLC patients with EGFR mutations treated with EGFR-TKI after being instructed to follow an alkaline diet were retrospectively evaluated. RESULTS: The median progression-free survival (PFS) and overall survival (OS) were 19.5 (range=3.1-33.8) and 28.5 (range=15.4-46.6) months. The average dosage of EGFR-TKI was 56±22% of the standard dosage. Urine pH was significantly increased after the alkaline diet (6.00±0.38 vs. 6.95±0.55; p<0.05). CONCLUSION: An alkaline diet may enhance the effect of EGFR-TKI treatment in NSCLC patients with EGFR mutations.


Subject(s)
Adenocarcinoma , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms , Protein Kinase Inhibitors/therapeutic use , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/diet therapy , Adenocarcinoma/drug therapy , Afatinib , Aged , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Disease-Free Survival , ErbB Receptors/genetics , Erlotinib Hydrochloride/therapeutic use , Female , Gefitinib , Humans , Hydrogen-Ion Concentration , Kaplan-Meier Estimate , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/diet therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Male , Middle Aged , Mutation , Positron Emission Tomography Computed Tomography , Quinazolines/therapeutic use , Urine/chemistry
11.
J Cell Biochem ; 118(10): 3462-3471, 2017 10.
Article in English | MEDLINE | ID: mdl-28332225

ABSTRACT

Anaplastic lymphoma kinase (ALK) positive non-small cell lung cancer (NSCLC) patients are mostly treated with ALK tyrosine kinase inhibitors (TKIs). Crizotinib is the first generation ALK inhibitor practiced as a primary chemo to combat cancer cells followed by second generation inhibitor ceritinib which are effective against crizotinib resistant ALK mutations. However, patients treated with these drugs invariably relapsed because of the development of new drug resistance mutations. In this study we explored the crizotinib resistance in the presence of ALK mutations L1196M and G1269A through molecular dynamics simulation studies. Further mutation specific inhibitors CID 71748211 and CID 71728095 were identified to potentially inhibit ALK with mutations L1196M and G1269A, respectively. This computational investigation in-sighted the molecular factors involved in crizotinib resistance which enhanced in the identification of new ALK drugs that brings individualized medicine to treat ALK positive NSCLC patients with specific mutations. J. Cell. Biochem. 118: 3462-3471, 2017. © 2017 Wiley Periodicals, Inc.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Drug Resistance, Neoplasm , Lung Neoplasms/enzymology , Molecular Dynamics Simulation , Mutation, Missense , Protein Kinase Inhibitors/chemistry , Pyrazoles/chemistry , Pyridines/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/chemistry , Amino Acid Substitution , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/genetics , Crizotinib , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics
12.
Clin Nutr ; 36(4): 1022-1028, 2017 08.
Article in English | MEDLINE | ID: mdl-27373496

ABSTRACT

PURPOSE: To assess the efficacy of oral glutamine (Gln) supplementation on clinical and survival outcomes of patients with locally advanced non-small cell lung cancer (LA-NSCLC). MATERIALS/METHODS: Between 2010 and 2014, 122 stage III NSCLC patients were retrospectively analyzed. All patients received curative intent chemoradiotherapy (CRT). Prophylactic oral Gln powder was applied at a dose of 10 g tid. Effect of oral Gln supplementation in the prevention of severe (≥grade 2-3) acute radiation-induced esophagitis (ARE) and weight loss, and their relation with overall survival (OS) and disease-free survival (DFS) was measured. RESULTS: Median follow-up was 13.14 months (range; 1.97-55.36). Fifty-six (46%) patients had received oral Gln. Severe ARE was significantly lower in Gln-supplemented group (30% vs 70%; p = 0.002). Gln-free patients demonstrated a higher weight loss (p = 0.0001). In multivariate analysis hemoglobin (hb) level (<12 g/dL; p = 0.01) and nodal stage (N3; p = 0.01) were poor prognostic factors that affect OS; Weight loss (p = 0.06) and Gln-free (p = 0.05) reached nearly significant levels that poorly affect OS. Similarly, nodal stage (N3, p = 0.014) and Gln-free (p = 0.035) were poor prognostic factors that affect DFS. Weight loss (≥2%, p = 0.06) and hb level (<12 g/dL, p = 0.07) reached borderline significance that poorly affect DFS. Nodal stage (N3) was the only poor prognostic factor that affect OS and DFS in univariate analysis (p = 0.01, p = 0.009; respectively). CONCLUSION: Oral Gln supplementation significantly reduces grade 2-3 esophagitis and weight loss and also no negative impact on tumor control and survival outcomes in patients with LA-NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Chemoradiotherapy/adverse effects , Dietary Supplements , Esophagitis/prevention & control , Glutamine/therapeutic use , Lung Neoplasms/diet therapy , Radiation Injuries/prevention & control , Adult , Aged , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Combined Modality Therapy/adverse effects , Dietary Supplements/adverse effects , Esophagitis/etiology , Esophagitis/physiopathology , Female , Follow-Up Studies , Glutamine/adverse effects , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Radiation Injuries/physiopathology , Retrospective Studies , Severity of Illness Index , Survival Analysis , Tumor Burden/radiation effects , Weight Loss/radiation effects
14.
Mol Med Rep ; 13(3): 2590-6, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26847819

ABSTRACT

Metformin, the most widely administered oral anti­diabetic therapeutic agent, exerts its glucose-lowering effect predominantly via liver kinase B1 (LKB1)-dependent activation of adenosine monophosphate-activated protein kinase (AMPK). Accumulating evidence has demonstrated that metformin possesses potential antitumor effects. However, whether the antitumor effect of metformin is via the LKB1/AMPK signaling pathway remains to be determined. In the current study, the effects of metformin on proliferation, cell cycle progression, and apoptosis of human non­small cell lung cancer (NSCLC) H460 (LKB1­null) and H1299 (LKB1­positive) cells were assessed, and the role of LKB1/AMPK signaling in the anti­growth effects of metformin were investigated. Cell viability was determined using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, cell cycle distribution and apoptosis were assessed by flow cytometry, and protein expression levels were measured by western blotting. Metformin inhibited proliferation, induced significant cell cycle arrest at the G0­G1 phase and increased apoptosis in NSCLC cells in a time- and concentration-dependent manner, regardless of the level of LKB1 protein expression. Furthermore, knockdown of LKB1 with short hairpin RNA (shRNA) did not affect the antiproliferative effect of metformin in the H1299 cells. Metformin stimulated AMPK phosphorylation and subsequently suppressed the phosphorylation of mammalian target of rapamycin and its downstream effector, 70­kDa ribosomal protein S6 kinase in the two cell lines. These effects were abrogated by silencing AMPK with small interfering RNA (siRNA). In addition, knockdown of AMPK with siRNA inhibited the effect of metformin on cell proliferation in the two cell lines. These results provide evidence that the growth inhibition of metformin in NSCLC cells is mediated by LKB1­independent activation of AMPK, indicating that metformin may be a potential therapeutic agent for the treatment of human NSCLC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/diet therapy , Metformin/pharmacology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinase Kinases , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Humans , Liver/enzymology , Metformin/therapeutic use , Signal Transduction
15.
Nutr Cancer ; 65(2): 240-6, 2013.
Article in English | MEDLINE | ID: mdl-23441611

ABSTRACT

Although advances in cancer therapies continue to develop, the shortness of the survival of lung cancer patients is still disappointing. Therefore, finding new adjuvant strategies is within the focus of cancer cure. Based on observations that deuterium depletion inhibits the growth of cancer cell lines and suppresses certain proto-oncogenes, we have conducted a clinical study in 129 patients with small cell and nonsmall cell lung cancers who consumed deuterium-depleted drinking water (DDW) as a nontoxic agent in addition to conventional chemotherapy and radiotherapy. Median survival time (MST) was 25.9 mo in males and 74.1 mo in female patients; the difference between genders was statistically significant (p < 0.05). Median survival of subjects with brain metastasis was 27.1 mo. Cumulative 5-yr survival probabilities were 19%, 52%, and 33% in males, females, and all patients with brain metastasis, respectively. Gene expression analysis in mouse lung indicated that DDW attenuates 7,12-dimethylbenz(a)anthracene (DMBA)-induced expression of Bcl2, Kras, and Myc in females. In conclusion, DDW counteracts the DMBA-induced overexpression of Bcl2, Kras and Myc genes in mouse lung, and it may extend survival of lung cancer patients as a nontoxic anticancer dietary supplement, especially for women with tumors overexpressing cancer-related genes, because MST of DDW-consuming group was 2-4 times longer than it is generally observed in lung cancer patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/mortality , Deuterium , Drinking Water , Gene Expression Regulation/drug effects , Lung Neoplasms/diet therapy , Lung Neoplasms/mortality , Lung/drug effects , Small Cell Lung Carcinoma/diet therapy , Small Cell Lung Carcinoma/mortality , 9,10-Dimethyl-1,2-benzanthracene/toxicity , Adult , Aged , Aged, 80 and over , Animals , Brain Neoplasms/diet therapy , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Carcinoma, Non-Small-Cell Lung/pathology , Dietary Supplements , Drinking Water/chemistry , Female , Genes, bcl-2 , Genes, myc , Humans , Lung/physiology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred CBA , Middle Aged , Proto-Oncogene Proteins p21(ras)/genetics , Small Cell Lung Carcinoma/pathology , Water
16.
Lung Cancer ; 79(2): 111-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23199512

ABSTRACT

Non-small-cell lung cancer (NSCLC) is the leading cause of cancer deaths worldwide; however, only limited therapeutic treatments are available. The aim of present study was to elucidate the therapeutic effect of dietary restriction in human NSCLC xenografts. Adult female nude mice were injected subcutaneously in the right dorsal flank with NSCLC cell line A549 cells. 5 days after tumor implantation, animals were randomly divided into ad libitum-fed group (AL, 95% of average diary intake) or dietary-restriction-fed group (DR, 70% average diary intake). 24 days after implantation, it was found that DR inhibited tumor growth marked by lower tumor volume and weight. DR suppressed tumor proliferation marked by reduced proliferating cell nuclear antigen (PCNA) expression and activated mitochondria-mediated apoptosis. DR decreased microvessel density marked by decreased CD31 immunostaining and promoted vessel maturation marked by increased alpha-smooth muscle actin (α-SMA) and reduced Factor VIII expression. DR reduced intratumoral interstitial fluid pressure and attenuated tumor hypoxia detected by EF5 immunostaining. In addition, DR suppressed NFκB signaling pathway and downregulated its downstream proteins expression including cyclooxygenase 2 (COX-2) and inducible nitric oxide synthase (iNOS). DR suppressed phosphoinositide 3-kinase (PI3K)/AKT signaling pathway. In conclusion, dietary restriction suppresses tumor growth, reduces angiogenesis, and improves tumor microenvironment in human non-small-cell lung cancer xenografts. Dietary restriction could thus be envisaged as a nutritional countermeasure against non-small-cell lung cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood supply , Carcinoma, Non-Small-Cell Lung/diet therapy , Lung Neoplasms/blood supply , Lung Neoplasms/diet therapy , 3-Hydroxybutyric Acid/blood , Actins/metabolism , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Cyclooxygenase 2/metabolism , Extracellular Fluid/physiology , Factor VIII/metabolism , Female , Humans , Hypoxia/diet therapy , Insulin-Like Growth Factor I/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Microvessels/metabolism , Microvessels/pathology , NF-kappa B/metabolism , Neovascularization, Pathologic , Nitric Oxide Synthase Type II/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Pressure , Proliferating Cell Nuclear Antigen/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/blood
17.
Br J Nutr ; 108(2): 327-33, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22114792

ABSTRACT

PUFA from fish oil appear to have anti-inflammatory and anti-oxidative effects and improve nutritional status in cancer patients. With this as background, the aim of the present study was to investigate the effect of EPA plus DHA on inflammatory condition, and oxidative and nutritional status in patients with lung cancer. In our multicentre, randomised, double-blind trial, thirty-three patients with a diagnosis of advanced inoperable non-small-cell lung cancer and undergoing chemotherapy were divided into two groups, receiving four capsules/d containing 510 mg of EPA and 340 mg of DHA, or 850 mg of placebo, for 66 d. At the start of chemotherapy (T0), after 8 d (T1), 22 d (T2) and 66 d (T3), biochemical (inflammatory and oxidative status parameters) and anthropometric parameters were measured in both groups. A significant increase of body weight in the n-3 group at T3 v. T0 was observed. Concerning inflammation, C-reactive protein and IL-6 levels differed significantly between the n-3 and placebo groups at T3, and progressively decreased during chemotherapy in the n-3 group, evidencing n-3 PUFA anti-inflammatory action. Concerning oxidative status, plasma reactive oxygen species levels increased in the placebo group v. the n-3 group at the later treatment times. Hydroxynonenal levels increased in the placebo group during the study, while they stabilised in the n-3 group. Our data confirm that the continual assumption of EPA plus DHA determined an anti-inflammatory and anti-oxidative action which could be considered a preliminary goal in anti-cachectic therapy.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Carcinoma, Non-Small-Cell Lung/diet therapy , Dietary Supplements , Fatty Acids, Omega-3/therapeutic use , Lung Neoplasms/diet therapy , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antioxidants/adverse effects , C-Reactive Protein/analysis , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Combined Modality Therapy/adverse effects , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Dietary Supplements/adverse effects , Docosahexaenoic Acids/administration & dosage , Double-Blind Method , Eicosapentaenoic Acid/administration & dosage , Fatty Acids, Omega-3/adverse effects , Female , Humans , Interleukin-6/blood , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Male , Oxidative Stress , Patient Dropouts , Weight Gain , Gemcitabine
18.
J Hum Nutr Diet ; 24(5): 431-40, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21733143

ABSTRACT

BACKGROUND: Weight loss in patients with cancer is common and associated with a poorer survival and quality of life. Benefits from nutritional interventions are unclear. The present study assessed the effect of dietary advice and/or oral nutritional supplements on survival, nutritional endpoints and quality of life in patients with weight loss receiving palliative chemotherapy for gastrointestinal and non-small cell lung cancers or mesothelioma. METHODS: Participants were randomly assigned to receive no intervention, dietary advice, a nutritional supplement or dietary advice plus supplement before the start of chemotherapy. Patients were followed for 1 year. Survival, nutritional status and quality of life were assessed. RESULTS: In total, 256 men and 102 women (median age, 66 years; range 24-88 years) with gastrointestinal (n = 277) and lung (n = 81) cancers were recruited. Median (range) follow-up was 6 (0-49) months. One-year survival was 38.6% (95% confidence interval 33.3-43.9). No differences in survival, weight or quality of life between groups were seen. Patients surviving beyond 26 weeks experienced significant weight gain from baseline to 12 weeks, although this was independent of nutritional intervention. CONCLUSIONS: Simple nutritional interventions did not improve clinical or nutritional outcomes or quality of life. Weight gain predicted a longer survival but occurred independently of nutritional intervention.


Subject(s)
Carcinoma, Non-Small-Cell Lung/diet therapy , Dietary Supplements , Gastrointestinal Neoplasms/diet therapy , Mesothelioma/diet therapy , Nutritional Status/drug effects , Palliative Care/methods , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Confidence Intervals , Dietetics , Endpoint Determination , Female , Follow-Up Studies , Gastrointestinal Neoplasms/drug therapy , Humans , Male , Mesothelioma/drug therapy , Middle Aged , Prospective Studies , Quality of Life , Weight Gain/drug effects , Weight Loss/drug effects , Young Adult
19.
Biochem Pharmacol ; 81(4): 510-7, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21114963

ABSTRACT

Methotrexate (MTX) is a dihydrofolate reductase (DHFR) inhibitor widely used as an anticancer drug in different kinds of human cancers. Here we investigated the anti-tumor mechanism of MTX against non-small cell lung cancer (NSCLC) A549 cells. MTX not only inhibited in vitro cell growth via induction of apoptosis, but also inhibited tumor formation in animal xenograft model. RNase protection assay (RPA) and RT-PCR demonstrated its induction of p53 target genes including DR5, p21, Puma and Noxa. Moreover, MTX promoted p53 phosphorylation at Ser15 and acetylaion at Lys373/382, which increase its stability and expression. The apoptosis and inhibition of cell viability induced by MTX were dependent on p53 and, partially, on p21. In addition, MTX also increased E-cadherin expression through inhibition of histone deacetylase (HDAC) activity and downregulation of polycomb group protein enhancer of zeste homologue 2 (EZH2). Therefore, the anticancer mechanism of MTX acts through initiation of p53-dependent apoptosis and restoration of E-cadherin expression by downregulation of HDAC/EZH2.


Subject(s)
Apoptosis/drug effects , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/drug therapy , DNA-Binding Proteins/genetics , Histone Deacetylases/genetics , Methotrexate/pharmacology , Transcription Factors/genetics , Tumor Suppressor Protein p53/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Down-Regulation/drug effects , Down-Regulation/genetics , Enhancer of Zeste Homolog 2 Protein , Humans , Polycomb Repressive Complex 2
20.
J Biol Chem ; 284(18): 12153-64, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19269967

ABSTRACT

Cell senescence, an irreversible cell cycle arrest, reflects a safeguard program that limits the capacity of uncontrolled cell proliferation. Treatment of tumor cells with certain chemotherapeutic agents activates premature senescence to decrease the tumorigenecity. Here we show that sublethal concentrations of adriamycin could induce premature senescence in lung cancer cells. Adriamycin treatment resulted in the up-regulation of BMP4, which is underexpressed in NSCLC (non-small cell lung cancers). Moreover, the BMP4-Smad pathway played a key role in mediating adriamycin-induced senescence. Overexpression of BMP4 was able to induce premature senescence in lung cancer cells and this process required the participation of cyclin/cyclin-dependent kinase (cdk) inhibitors p16(INK4a) and p21(WAF1/cip1). We also show that increases of p16(INK4a) and p21(WAF1/cip1) expression in response to BMP4 were mediated by the Smad signaling pathway. Furthermore, our data revealed that p300 was recruited to P16(INK4a) and P21(WAF1/cip1) promoters by Smad1/5/8 to induce the hyperacetylation of histones H3 and H4 at the promoters. The present study provides useful clues to the evaluation of the potentiality of BMP4 as a responsive molecular target for cancer chemotherapy.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Bone Morphogenetic Protein 4/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Cellular Senescence/drug effects , Doxorubicin/pharmacology , Lung Neoplasms/metabolism , Signal Transduction/drug effects , Smad Proteins/metabolism , Acetylation/drug effects , Antibiotics, Antineoplastic/therapeutic use , Bone Morphogenetic Protein 4/genetics , Carcinoma, Non-Small-Cell Lung/diet therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Doxorubicin/therapeutic use , Histones/genetics , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Promoter Regions, Genetic/genetics , Smad Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...