1
|
Wei C, Sun H, Liu S, Hu J, Cao B. A nomogram for predicting survival based on hemoglobin A1c and circulating tumor cells in advanced gastric cancer patients receiving immunotherapy. Int Immunopharmacol 2024; 142:113239. [PMID: 39306892 DOI: 10.1016/j.intimp.2024.113239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/22/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND Our study aimed to investigate the correlation between hemoglobin A1c (HbA1c), circulating tumor cells (CTCs) and prognosis in advanced gastric cancer (GC) patients who received immunotherapy and explore the potential prognostic predictors to develop a nomogram. METHODS We retrospectively enrolled 259 patients with advanced GC treated at Beijing Friendship Hospital between September 2014 and March 2024. Patients were divided into the immunochemotherapy cohort (ICT) and the chemotherapy (CT) cohort. Survival rate was calculated by Kaplan-Meier survival curve, and the differences were evaluated by log-rank test. The univariate and multivariate Cox proportional hazards regression model was used to identify factors independently associated with survival. A nomogram was developed to estimate 6-, 12-, and 18-month progression-free survival (PFS) probability based on the ICT cohort. RESULTS Patients achieved higher PFS in the ICT cohort than the CT cohort. We focused on the ICT cohort and constructed a nomogram based on the multivariate analysis, including five variables: age, PD-L1 expression, HbA1c, CTCs and CEA*. The concordance index value was 0.82 in the training cohort and 0.75 in the validation cohort. Furthermore, we proved the nomogram was clinically useful and performed better than PD-L1 expression staging system. Notably, we found high HbA1c level but not diabetes mellitus significantly affected the efficacy of ICT. CONCLUSION ICT showed better PFS than CT. In addition, HbA1c and CTCs were novel biomarkers to predict PFS in patients treated with ICT. The nomogram could predict PFS of advanced GC patients receiving ICT with increased accuracy and favorable clinical utility.
Collapse
Affiliation(s)
- Chenyu Wei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Haolin Sun
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shujing Liu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jiexuan Hu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Bangwei Cao
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
2
|
De la Torre K, Song M, Abe SK, Rahman MS, Islam MR, Saito E, Min S, Huang D, Chen Y, Gupta PC, Sawada N, Tamakoshi A, Shu X, Wen W, Sakata R, Kim J, Nagata C, Ito H, Park SK, Shin M, Pednekar MS, Tsugane S, Kimura T, Gao Y, Cai H, Wada K, Oze I, Shin A, Ahn Y, Ahsan H, Boffetta P, Chia KS, Matsuo K, Qiao Y, Rothman N, Zheng W, Inoue M, Kang D. Diabetes and gastric cancer incidence and mortality in the Asia Cohort Consortium: A pooled analysis of more than a half million participants. J Diabetes 2024; 16:e13561. [PMID: 38751364 PMCID: PMC11096812 DOI: 10.1111/1753-0407.13561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 02/19/2024] [Accepted: 03/14/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Evidence suggests a possible link between diabetes and gastric cancer risk, but the findings remain inconclusive, with limited studies in the Asian population. We aimed to assess the impact of diabetes and diabetes duration on the development of gastric cancer overall, by anatomical and histological subtypes. METHODS A pooled analysis was conducted using 12 prospective studies included in the Asia Cohort Consortium. Among 558 981 participants (median age 52), after a median follow-up of 14.9 years and 10.5 years, 8556 incident primary gastric cancers and 8058 gastric cancer deaths occurred, respectively. Cox proportional hazard regression models were used to estimate study-specific hazard ratios (HRs) and 95% confidence intervals (CIs) and pooled using random-effects meta-analyses. RESULTS Diabetes was associated with an increased incidence of overall gastric cancer (HR 1.15, 95% CI 1.06-1.25). The risk association did not differ significantly by sex (women vs men: HR 1.31, 95% CI 1.07-1.60 vs 1.12, 1.01-1.23), anatomical subsites (noncardia vs cardia: 1.14, 1.02-1.28 vs 1.17, 0.77-1.78) and histological subtypes (intestinal vs diffuse: 1.22, 1.02-1.46 vs 1.00, 0.62-1.61). Gastric cancer risk increased significantly during the first decade following diabetes diagnosis (HR 4.70, 95% CI 3.77-5.86), and decreased with time (nonlinear p < .01). Positive associations between diabetes and gastric cancer mortality were observed (HR 1.15, 95% CI 1.03-1.28) but attenuated after a 2-year time lag. CONCLUSION Diabetes was associated with an increased gastric cancer incidence regardless of sex, anatomical subsite, or subtypes of gastric cancer. The risk of gastric cancer was particularly high during the first decade following diabetes diagnosis.
Collapse
Grants
- R37 CA070867 NCI NIH HHS
- 30-A-15,23-A-31(toku),26-A-2,29-A-4 National Cancer Center Japan Research and Development Fund
- 24H1080 National Cancer Center Korea Research Grant
- 0520160-1 National R&D Program for Cancer Control, Ministry of Health & Welfare, Republic of Korea
- 23-A-31 Ministry of Health, Labour and Welfare of Japan
- NRF-2016R1A2B4014552 National Research Foundation of Korea
- The Japanese Ministry of the Environment
- Ministry of Education, Culture, Sports, Science and Technology, Japan
- International Agency for Research on Cancer, Lyon, France
- 2210990 National Cancer Center Korea Research Grant
- US Department of Energy
- UM1 CA173640 NCI NIH HHS
- UM1 CA182910 NCI NIH HHS
- Ministry of Education, Culture, Sports, Science and Technology, Japan
- Ministry of Health, Labour and Welfare of Japan
- National Research Foundation of Korea
- Division of Cancer Prevention, National Cancer Institute
- The Japanese Ministry of the Environment
- International Agency for Research on Cancer, Lyon, France
- US Department of Energy
Collapse
Affiliation(s)
- Katherine De la Torre
- Department of Biomedical SciencesSeoul National University Graduate SchoolSeoulKorea
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Minkyo Song
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Laboratory of Epidemiology and Population SciencesNational Institute on Aging, National Institute of HealthBaltimoreMarylandUSA
| | - Sarah Krull Abe
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Md. Shafiur Rahman
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
- Research Center for Child Mental Development, Hamamatsu University School of MedicineTokyoJapan
| | - Md. Rashedul Islam
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
- Hitotsubashi Institute for Advanced Study, Hitotsubashi UniversityTokyoJapan
| | - Eiko Saito
- Institute for Global Health Policy Research, National Center for Global Health and MedicineTokyoJapan
| | - Sukhong Min
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Dan Huang
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate SchoolSeoulKorea
| | - Yu Chen
- Department of Population Health and Environmental MedicineNYU Grossman School of MedicineNew YorkNew YorkUSA
| | | | - Norie Sawada
- Division of Cohort ResearchNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Akiko Tamakoshi
- Department of Public HealthHokkaido University Faculty of MedicineSapporoJapan
| | - Xiao‐Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ritsu Sakata
- Radiation Effects Research FoundationHiroshimaJapan
| | - Jeongseon Kim
- Graduate School of Cancer Science and Policy, National Cancer CenterGoyangKorea
| | - Chisato Nagata
- Department of Epidemiology and Preventive MedicineGifu University Graduate School of MedicineGifuJapan
| | - Hidemi Ito
- Department of Preventive Medicine, Division of Cancer Information and ControlAichi Cancer Center Research InstituteNagoyaJapan
- Division of Descriptive Cancer EpidemiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Sue K. Park
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Myung‐Hee Shin
- Department of Social and Preventive MedicineSungkyunkwan University School of MedicineSeoulKorea
| | | | - Shoichiro Tsugane
- Division of Cohort ResearchNational Cancer Center Institute for Cancer ControlTokyoJapan
- International University of Health and Welfare Graduate SchoolTokyoJapan
| | - Takashi Kimura
- Department of Public HealthHokkaido University Faculty of MedicineSapporoJapan
| | - Yu‐Tang Gao
- Department of EpidemiologyShanghai Cancer InstituteShanghaiChina
- Renji Hospital, Shanghai Jiaotong University School of MedicineShanghaiChina
| | - Hui Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Keiko Wada
- Department of Epidemiology and Preventive MedicineGifu University Graduate School of MedicineGifuJapan
| | - Isao Oze
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
| | - Aesun Shin
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Cancer Research Institute, Seoul National UniversitySeoulKorea
| | - Yoon‐Ok Ahn
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
| | - Habibul Ahsan
- Department of Public Health SciencesUniversity of ChicagoChicagoIllinoisUSA
| | - Paolo Boffetta
- Stony Brook Cancer Center, Stony Brook UniversityStony BrookNew YorkUSA
- Department of Medical and Surgical SciencesUniversity of BolognaBolognaItaly
| | - Kee Seng Chia
- Saw Swee Hock School of Public Health, National University of SingaporeSingaporeSingapore
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and PreventionAichi Cancer Center Research InstituteNagoyaJapan
- Department of Cancer EpidemiologyNagoya University Graduate School of Medicine NagoyaNagoyaJapan
| | - You‐Lin Qiao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nathaniel Rothman
- Occupational and Environmental Epidemiology Branch, Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center Vanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Manami Inoue
- Division of PreventionNational Cancer Center Institute for Cancer ControlTokyoJapan
| | - Daehee Kang
- Department of Preventive MedicineSeoul National University College of MedicineSeoulKorea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate SchoolSeoulKorea
| |
Collapse
|
3
|
Liu J, Wang R, Tan S, Zhao X, Hou A. Association between insulin resistance, metabolic syndrome and its components and lung cancer: a systematic review and meta-analysis. Diabetol Metab Syndr 2024; 16:63. [PMID: 38468310 PMCID: PMC10926619 DOI: 10.1186/s13098-024-01308-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 03/05/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND A growing body of evidence points to the association between insulin resistance (IR), metabolic syndrome (MetS) and its components and lung cancer incidence, but remains controversial and unknown. METHODS A systematic search was conducted through PubMed, Embase, Cochrane Library, the China National Knowledge Infrastructure (CNKI) and Wanfang databases for the corresponding studies. Each study reported the risk estimate and 95% confidence intervals (CI) for lung cancer, and a fixed effects model or random effects model was used for outcome. RESULTS We included 31 publications involving 6,589,383 people with 62,246 cases of lung cancer. Diabetes mellitus (DM) (RR = 1.11, 95% CI 1.06-1.16, P = 0.000) and IR (RR = 2.35, 95% CI 1.55-3.58, P = 0.000) showed a positive association with lung cancer risk. BMI (RR = 0.66, 95% CI 0.54-0.81, P = 0.000) and HDL-C (RR = 0.88, 95% CI 0.79-0.97, P = 0.010) were negatively correlated with lung cancer. MetS(RR = 0.99, 95% CI 0.90-1.09, P = 0.801), TC (RR = 0.93, 95% CI 0.81-1.06, P = 0.274), TG (RR = 0.99, 95% CI 0.88-1.12,P = 0.884), LDL-C (RR = 1.01, 95% CI 0.87-1.16, P = 0.928), hypertension (RR = 1.01, 95% CI 0.88-1.15, P = 0.928), FBG (RR = 1.02, 95% CI 0.92-1.13, P = 0.677) and obesity (RR = 1.11, 95% CI 0.92-1.35, P = 0.280) were not associated with lung cancer. CONCLUSION Our study showed that the risk of lung cancer is correlated with DM, IR, BMI, and HDL-C. Timely control of these metabolic disorders may have a positive effect on preventing lung cancer. Trial registration Our study has been registered in the Prospective Register of Systematic Reviews (PROSPERO), ID: CRD42023390710.
Collapse
Affiliation(s)
- Jingxuan Liu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rui Wang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Song Tan
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, China
| | - Xiaohu Zhao
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Aihua Hou
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, China.
| |
Collapse
|
4
|
Wang T, Chen S, Wang Z, Li S, Fei X, Wang T, Zhang M. KIRREL promotes the proliferation of gastric cancer cells and angiogenesis through the PI3K/AKT/mTOR pathway. J Cell Mol Med 2024; 28:e18020. [PMID: 37909722 PMCID: PMC10805501 DOI: 10.1111/jcmm.18020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 11/03/2023] Open
Abstract
Anti-angiogenesis is a promising therapeutic strategy for delaying tumour progression that offers, new hope for gastric cancer targeted therapy. The purpose of this study was to investigate the precise mechanism by which Kin of IRRE-like protein 1 (KIRREL) contributes to the development of gastric cancer, particularly in terms of tumour angiogenesis. Differential expression of KIRREL in tissues and cells was detected using quantitative real-time polymerase chain reaction, western blotting and immunohistochemistry. A bioinformatics analysis was conducted to screen for the function and pathway enrichment of KIRREL in gastric cancer. Lentivirus-induced KIRREL silencing in SNU-5 cells and lentivirus-induced KIRREL overexpression in AGS cells were used to study the effect of KIRREL on the proliferation, cell cycle and angiogenesis of gastric cancer cells. Moreover, the expressions of PI3K, P-PI3K, AKT, P-AKT, mTOR, P-mTOR, HIF-1α and VEGF were also detected. Gastric cancer tissues and cells had high levels of KIRREL expression, which is associated with the proliferation, cell cycle and angiogenesis of gastric cancer cells. After silencing and overexpressing KIRREL in SNU-5 and AGS cells, respectively, the proliferation and angiogenesis of SNU-5 cells were inhibited, while the proliferation and angiogenesis of AGS cells were promoted. According to a bioinformatics analysis of the KIRREL gene, angiogenesis regulation and the PI3K/AKT pathway were highly connected. The PI3K/AKT/mTOR pathway was repressed and stimulated by KIRREL silencing and overexpression, respectively. IGF-1, an AKT agonist, and LY294002, an inhibitor, reversed the effects of KIRREL silencing and overexpression on the PI3K/AKT/mTOR pathway and on gastric cancer cell proliferation and angiogenesis. KIRREL may mediate the proliferation and angiogenesis of gastric cancer cells through the PI3K/AKT/mTOR signalling pathway. These findings could help in the further development of potential anti-angiogenesis targets.
Collapse
Affiliation(s)
- Tao Wang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Shuo Chen
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Ziliang Wang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Siyu Li
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Xichang Fei
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Tong Wang
- Department of General PracticeThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| | - Mingjun Zhang
- Department of OncologyThe Second Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
| |
Collapse
|
5
|
Wang L, Zhang Z. Diabetes Mellitus and Gastric Cancer: Correlation and Potential Mechanisms. J Diabetes Res 2023; 2023:4388437. [PMID: 38020199 PMCID: PMC10653978 DOI: 10.1155/2023/4388437] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023] Open
Abstract
This review summarizes the correlation between diabetes mellitus (DM) and gastric cancer (GC) from the perspectives of epidemiology, drug use, and potential mechanisms. The association between DM and GC is inconclusive, and the positive direction of the association reported in most published meta-analyses suggests that DM may be an independent risk factor for GC. Many clinical investigations have shown that people with DM and GC who undergo gastrectomy may have better glycemic control. The potential link between DM and GC may involve the interaction of multiple common risk factors, such as obesity, hyperglycemia and hyperinsulinemia, H. pylori infection, and the use of metformin. Although in vitro and in vivo data support that H. pylori infection status and metformin can influence GC risk in DM patients, there are conflicting results. Patient survival outcomes are influenced by multiple factors, so further research is needed to identify the patients who may benefit.
Collapse
Affiliation(s)
- Li Wang
- Department of Emergency, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, China
- Zhejiang Provincial Critical Research Center for Emergency Medicine Clinic, Hangzhou 310052, China
- Key Laboratory of Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310052, China
| | - Zhe Zhang
- Department of Emergency Medicine, The First People's Hospital of Linping District, 311100, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Sanikini H, Biessy C, Rinaldi S, Navionis AS, Gicquiau A, Keski-Rahkonen P, Kiss A, Weinstein SJ, Albanes D, Agudo A, Jenab M, Riboli E, Gunter MJ, Murphy G, Cross AJ. Circulating hormones and risk of gastric cancer by subsite in three cohort studies. Gastric Cancer 2023; 26:969-987. [PMID: 37455285 PMCID: PMC10640529 DOI: 10.1007/s10120-023-01414-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 06/20/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Obesity has been positively associated with gastric cancer. Excess fat impacts hormones, which have been implicated in carcinogenesis. We investigated obesity-related hormones and cardia gastric cancer (CGC) and non-cardia gastric cancer (NCGC) risk. METHODS Nested case-control studies were conducted within the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort (61 CGCs, and 172 NCGCs and matched controls) and the Alpha-Tocopherol, Beta-Carotene Cancer Prevention (ATBC) study (100 CGCs and 65 NCGCs and matched controls); serum hormones were measured. In UK-Biobank (n = 458,713), we included 137 CGCs and 92 NCGCs. Sex-specific analyses were conducted. For EPIC and ATBC, odds ratios (ORs), and for UK-Biobank hazard ratios (HRs), were estimated using conditional logistic regression and Cox regression, respectively. RESULTS Insulin-like growth-factor-1 was positively associated with CGC and NCGC in EPIC men (ORper 1-SD increase 1.94, 95% CI 1.03-3.63; ORper 1-SD increase 1.63, 95% CI 1.05-2.53, respectively), with similar findings for CGC in UK-Biobank women (HRper 1-SD increase 1.76, 95% CI 1.08-2.88). Leptin in EPIC men and C-peptide in EPIC women were positively associated with NCGC (ORT3 vs. T1 2.72, 95% CI 1.01-7.34 and ORper 1-SD increase 2.17, 95% CI 1.19-3.97, respectively). Sex hormone-binding globulin was positively associated with CGC in UK-Biobank men (HRper 1-SD increase 1.29, 95% CI 1.02-1.64). Conversely, ghrelin was inversely associated with NCGC among EPIC and ATBC men (ORper 1-SD increase 0.53, 95% CI 0.34-0.84; ORper 1-SD increase 0.22, 95% CI 0.10-0.50, respectively). In addition, dehydroepiandrosterone was inversely associated with CGC in EPIC and ATBC men combined. CONCLUSIONS Some obesity-related hormones influence CGC and NCGC risk.
Collapse
Affiliation(s)
- Harinakshi Sanikini
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St. Mary's Campus, Norfolk Place, London, W2 1PG, UK.
| | - Carine Biessy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Sabina Rinaldi
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Anne-Sophie Navionis
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Audrey Gicquiau
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Pekka Keski-Rahkonen
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Agneta Kiss
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Stephanie J Weinstein
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Demetrius Albanes
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD, USA
| | - Antonio Agudo
- Unit of Nutrition and Cancer, Catalan Institute of Oncology-ICO, L'Hospitalet de Llobregat, Spain
- Nutrition and Cancer Group, Epidemiology, Public Health, Cancer Prevention and Palliative Care Program, Bellvitge Biomedical Research Institute-IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Elio Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St. Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Marc J Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St. Mary's Campus, Norfolk Place, London, W2 1PG, UK
| | - Gwen Murphy
- Cancer Screening and Prevention Research Group, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St. Mary's Campus, Norfolk Place, London, W2 1PG, UK
- Cancer Screening and Prevention Research Group, Department of Surgery and Cancer, Imperial College London, London, UK
| |
Collapse
|
7
|
Xu HW, Wang MQ, Zhu SL. Analysis of IGFBP7 expression characteristics in pan-cancer and its clinical relevance to stomach adenocarcinoma. Transl Cancer Res 2023; 12:2596-2612. [PMID: 37969374 PMCID: PMC10643967 DOI: 10.21037/tcr-23-1055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/21/2023] [Indexed: 11/17/2023]
Abstract
Background Insulin-like growth factor (IGF) binding proteins (IGFBPs) are involved in tumorigenesis and cancer progression. IGFBP7 has been shown to act as either a tumor suppressive gene or an oncogene in many tumors, including stomach adenocarcinoma (STAD). To provide a more systematic and comprehensive understanding of IGFBP7 gene, we performed an integrative pan-cancer analysis and explored further with the case of STAD. Methods We compared the expression data of IGFBP7 in various cancer and normal tissues obtained from The Cancer Genome Atlas (TCGA) database and the Genotype-Tissue Expression (GTEx) database. The TISIDB web portal was used to analyze the associations of IGFBP7 with cancer molecular subtypes and immune subtypes. We also analyzed the predictive ability and prognostic values of IGFBP7 in pan-cancer, as well as explored its targeted binding proteins and their biological functions. Additionally, we examined the relationship between IGFBP7 and the clinical characteristics of STAD, investigated the co-expression genes and biological functions of differentially expressed genes (DEGs), and validated the mRNA and protein expression levels of IGFBP7 using gastric cancer (GC) and adjacent normal tissues in a small self-case-control study. Results IGFBP7 was found to be overexpressed in STAD and downregulated in many other cancers. The mRNA and protein expression levels of IGFBP7 were also significantly higher in the collected GC tissues compared with adjacent tissues. Expression of IGFBP7 varied significantly across molecular subtypes of nine different cancer types and immune subtypes of eight types, with the highest expression observed in the genomically stable molecular subtype and C3 inflammatory immune subtype in STAD. IGFBP7 demonstrated an area under the curve (AUC) >0.7 for predicting 16 cancer types, and an AUC >0.9 for seven types. Patients in the higher IGFBP7 expression group showed a poorer prognosis for adrenal cortical carcinoma (ACC) and low-grade glioma (LGG), while demonstrating a more favorable prognosis for kidney renal clear cell carcinoma (KIRC). IGFBP7 expression in STAD was significantly associated with T stage, pathological stage, histologic grade, and Helicobacter pylori infection. Conclusions IGFBP7 showed promise as a biomarker for prediction and prognosis in pan-cancer. IGFBP7 was found to be overexpressed in STAD, and its expression was closely associated with the clinical characteristics of STAD.
Collapse
|
8
|
Chen PD, Liao YY, Cheng YC, Wu HY, Wu YM, Huang MC. Decreased B4GALT1 promotes hepatocellular carcinoma cell invasiveness by regulating the laminin-integrin pathway. Oncogenesis 2023; 12:49. [PMID: 37907465 PMCID: PMC10618527 DOI: 10.1038/s41389-023-00494-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 09/26/2023] [Accepted: 10/11/2023] [Indexed: 11/02/2023] Open
Abstract
Beta1,4-galactosyltransferases (B4GALTs) play a crucial role in several diseases, including cancer. B4GALT1 is highly expressed in the liver, and patients with mutations in B4GALT1 exhibit hepatopathy. However, the role of B4GALT1 in liver cancer remains unclear. Here, we found that B4GALT1 was significantly downregulated in hepatocellular carcinoma (HCC) tissue compared with the adjacent liver tissue, and low B4GALT1 expression was associated with vascular invasion and poor overall survival in patients with HCC. Additionally, silencing or loss of B4GALT1 enhanced HCC cell migration and invasion in vitro and promoted lung metastasis of HCC in NOD/SCID mice. Moreover, B4GALT1 knockdown or knockout increased cell adhesion to laminin, whereas B4GALT1 overexpression decreased the adhesion. Through a mass spectrometry-based approach and Griffonia simplicifolia lectin II (GSL-II) pull-down assays, we identified integrins α6 and β1 as the main protein substrates of B4GALT1 and their N-glycans were modified by B4GALT1. Further, the increased cell migration and invasion induced by B4GALT1 knockdown or knockout were significantly reversed using a blocking antibody against integrin α6 or integrin β1. These results suggest that B4GALT1 downregulation alters N-glycosylation and enhances the laminin-binding activity of integrin α6 and integrin β1 to promote invasiveness of HCC cells. Our findings provide novel insights into the role of B4GALT1 in HCC metastasis and highlight targeting the laminin-integrin axis as a potential therapeutic strategy for HCC with low B4GALT1 expression.
Collapse
Affiliation(s)
- Po-Da Chen
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
- Department of Surgical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Ying-Yu Liao
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chia Cheng
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Wu
- Instrumentation center, National Taiwan University, Taipei, Taiwan
| | - Yao-Ming Wu
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan.
- Department of Surgical Oncology, National Taiwan University Cancer Center, Taipei, Taiwan.
| | - Min-Chuan Huang
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
9
|
Rojas A, Schneider I, Lindner C, Gonzalez I, Morales MA. Association between diabetes and cancer. Current mechanistic insights into the association and future challenges. Mol Cell Biochem 2023; 478:1743-1758. [PMID: 36565361 DOI: 10.1007/s11010-022-04630-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 11/30/2022] [Indexed: 12/25/2022]
Abstract
Compelling pieces of epidemiological, clinical, and experimental research have demonstrated that Diabetes mellitus (DM) is a major risk factor associated with increased cancer incidence and mortality in many human neoplasms. In the pathophysiology context of DM, many of the main classical actors are relevant elements that can fuel the different steps of the carcinogenesis process. Hyperglycemia, hyperinsulinemia, metabolic inflammation, and dyslipidemia are among the classic contributors to this association. Furthermore, new emerging actors have received particular attention in the last few years, and compelling data support that the microbiome, the epigenetic changes, the reticulum endoplasmic stress, and the increased glycolytic influx also play important roles in promoting the development of many cancer types. The arsenal of glucose-lowering therapeutic agents used for treating diabetes is wide and diverse, and a growing body of data raised during the last two decades has tried to clarify the contribution of therapeutic agents to this association. However, this research area remains controversial, because some anti-diabetic drugs are now considered as either promotors or protecting elements. In the present review, we intend to highlight the compelling epidemiological shreds of evidence that support this association, as well as the mechanistic contributions of many of these potential pathological mechanisms, some controversial points as well as future challenges.
Collapse
Affiliation(s)
- Armando Rojas
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile.
| | - Ivan Schneider
- Medicine Faculty, Catholic University of Maule, Talca, Chile
| | | | - Ileana Gonzalez
- Biomedical Research Labs, Medicine Faculty, Catholic University of Maule, Talca, Chile
| | - Miguel A Morales
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|
10
|
Huang Z, Zhou J, Chen L, Zhang Y. Metabolic Syndrome and Clinical Outcomes of Patients with Gastric Cancer: A Meta-Analysis. Horm Metab Res 2023; 55:333-342. [PMID: 37011889 DOI: 10.1055/a-2038-5830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
Metabolic syndrome (MetS) is suggested to participate in the pathogenesis and progress of some cancers via inducing low-grade systemic inflammation. However, the influence of MetS on patients with gastric cancer (GC) remains not fully determined. A systematic review and meta-analysis was therefore performed to evaluate the influence of MetS on clinical outcomes of patients with GC. A search of PubMed, Embase, Web of Science, Wanfang, and CNKI retrieved relevant cohort studies from the inception of the databases to October 11, 2022. We pooled the results using a random-effects model that incorporates heterogeneity. In the meta-analysis, 6649 patients with GC were included, and all of them received gastrectomy. A total of 1248 (18.8%) patients had MetS at baseline. Pooled results showed that MetS was associated with higher risks of postoperative complications [risk ratio (RR): 2.41, 95% confidence interval (CI): 1.85 to 3.14, p<0.001; I2=55%], overall mortality (RR: 1.73, 95% CI: 1.85 to 3.14, p<0.001; I2=77%), and recurrence of GC (RR: 2.00, 95% CI: 1.10 to 3.63, p=0.02; I2=39%). Subgroup analyses showed similar results in prospective and retrospective cohort studies and in studies with MetS diagnosed with the Chinese Diabetes Society criteria and the National Cholesterol Education Program Adult Treatment Panel III criteria (p for subgroup difference all>0.05). In patients with GC after gastrectomy, MetS may be a predictor of high incidence of postoperative complications, cancer recurrence, and overall mortality.
Collapse
Affiliation(s)
- Zhiyang Huang
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Jianwei Zhou
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| | - Long Chen
- Center for Rehabilitation Medicine and Department of Anesthesiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yibing Zhang
- Department of Gastroenterology, The Dingli Clinical College of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
11
|
Correlation Study on the Expression of INSR, IRS-1, and PD-L1 in Nonsmall Cell Lung Cancer. JOURNAL OF ONCOLOGY 2022; 2022:5233222. [PMID: 36245982 PMCID: PMC9553505 DOI: 10.1155/2022/5233222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/05/2022] [Accepted: 08/11/2022] [Indexed: 11/18/2022]
Abstract
Objective To study the expression and correlation of insulin receptor (INSR), insulin receptor substrate-1 (IRS-1), and programmed cell death ligand-1 (PD-L1) in nonsmall cell lung cancer (NSCLC). Methods 45 lung cancer tissues and 30 adjacent normal tissues of NSCLC patients diagnosed in the Second Affiliated Hospital of Shandong First Medical University from June 2019 to August 2020 were selected. The expressions of INSR, IRS-1, and PD-L1 proteins in tumor tissues and adjacent tissues of NSCLC were detected by immunohistochemical staining. Results The expression of INSR and IRS-1 in NSCLC was significantly higher than that in adjacent normal lung tissue (P < 0.05). INSR expression had statistical significance with the degree of pathological differentiation of nonsmall cell carcinoma (P = 0.031), but had no significant association with age, gender, pathological type, TNM stage, and lymph node metastasis status (P > 0.05). There was no significant correlation between IRS-1 positive expression and NSCLC patients' age, gender, pathological typing, degree of differentiation, TNM stage, and lymph node metastasis (P > 0.05). PD-L1 positive expression was correlated with lymph node metastasis of NSCLC (P = 0.028), while there was no significant correlation with gender, age, pathological type, TNM stage, and pathological differentiation degree of NSCLC patients (P > 0.05). Spearman correlation analysis showed that PD-L1 protein expression had a significant positive correlation with IRS-1 protein expression (r = 0.373), but was not correlated with the expression of INSR protein. Conclusion IRS-1 may be involved in the regulation of PD-L1 expression and mediate the occurrence of tumor immune escape, which is expected to become a new target for NSCLC immunotherapy and provide new clinical evidence for immunosuppressive therapy.
Collapse
|
12
|
Sun H, Qi X. The role of insulin and incretin-based drugs in biliary tract cancer: epidemiological and experimental evidence. Discov Oncol 2022; 13:70. [PMID: 35933633 PMCID: PMC9357599 DOI: 10.1007/s12672-022-00536-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 07/26/2022] [Indexed: 11/29/2022] Open
Abstract
Insulin and incretin-based drugs are important antidiabetic agents with complex effects on cell growth and metabolism. Emerging evidence shows that insulin and incretin-based drugs are associated with altered risk of biliary tract cancer (BTC). Observational study reveals that insulin is associated with an increased risk of extrahepatic cholangiocarcinoma (ECC), but not intrahepatic cholangiocarcinoma (ICC) or gallbladder cancer (GBC). This type-specific effect can be partly explained by the cell of origin and heterogeneous genome landscape of the three subtypes of BTC. Similar to insulin, incretin-based drugs also exhibit very interesting contradictions and inconsistencies in response to different cancer phenotypes, including BTC. Both epidemiological and experimental evidence suggests that incretin-based drugs can be a promoter of some cancers and an inhibitor of others. It is now more apparent that this type of drugs has a broader range of physiological effects on the body, including regulation of endoplasmic reticulum stress, autophagy, metabolic reprogramming, and gene expression. In particular, dipeptidyl peptidase-4 inhibitors (DPP-4i) have a more complex effect on cancer due to the multi-functional nature of DPP-4. DPP-4 exerts both catalytic and non-enzymatic functions to regulate metabolic homeostasis, immune reaction, cell migration, and proliferation. In this review, we collate the epidemiological and experimental evidence regarding the effect of these two classes of drugs on BTC to provide valuable information.
Collapse
Affiliation(s)
- Hua Sun
- Department of Geriatrics, Zhejiang Hospital of Integrated Traditional Chinese and Western Medicine, No.208 East Huancheng Road, Hangzhou, Zhejiang, China
| | - Xiaohui Qi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.197 Ruijin Er Road, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, No.573 Xujiahui Road, Shanghai, China.
| |
Collapse
|
13
|
Vafaee R, Jahani Sherafat S, Rezaei Tavirani M, Ahmadi N. Down-regulation of TP53 is a highlighted molecular event in gastric ulcer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:249-255. [PMID: 36311969 PMCID: PMC9589131 DOI: 10.22037/ghfbb.v15i3.2544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/02/2022] [Indexed: 01/24/2023]
Abstract
AIM The current study explored the crucial dysregulate proteins and biochemical pathways in gastric ulcer as its main aim. BACKGROUND Gastric ulcer as an acid-related gastrointestinal disease is known as one of the most public gastrointestinal disorders. METHODS A total of 100 proteins from STRING database were analyzed by Cytoscape and its applications to find the central proteins and the related biochemical pathways. Action map analysis was applied to explore regulatory relationships between the critical proteins. RESULTS Network analysis and gene ontology revealed that IL6, ALB, TNF, INS, IL1B, IL10, TP53, CXCL8, and PTGS2 are the highlighted proteins related to gastric ulcer. Six clusters of biochemical pathways, namely "response to external stimulus," "multicellular organismal process," "regulation of biological quality," "cellular response to stimulus," "cellular response to chemical stimulus," and "transport" were identified as the dysregulated pathway in patients. CONCLUSION Down-regulation of TP53 by IL2, PTGS2, and TNF seems to be a main process occurring in gastric ulcer patients.
Collapse
Affiliation(s)
- Reza Vafaee
- Critical Care Quality Improvement Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Jahani Sherafat
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Rezaei Tavirani
- Proteomics research center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nayebali Ahmadi
- Proteomics research center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|