1
|
Tilg H, Ianiro G, Gasbarrini A, Adolph TE. Adipokines: masterminds of metabolic inflammation. Nat Rev Immunol 2024:10.1038/s41577-024-01103-8. [PMID: 39511425 DOI: 10.1038/s41577-024-01103-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Adipose tissue is an immunologically active organ that controls host physiology, partly through the release of mediators termed adipokines. In obesity, adipocytes and infiltrating leukocytes produce adipokines, which include the hormones adiponectin and leptin and cytokines such as tumour necrosis factor and IL-1β. These adipokines orchestrate immune responses that are collectively referred to as metabolic inflammation. Consequently, metabolic inflammation characterizes metabolic disorders and promotes distinct disease aspects, such as insulin resistance, metabolic dysfunction-associated liver disease and cardiovascular complications. In this unifying concept, adipokines participate in the immunological cross-talk that occurs between metabolically active organs in metabolic diseases, highlighting the fundamental role of adipokines in obesity and their potential for therapeutic intervention. Here, we summarize how adipokines shape metabolic inflammation in mice and humans, focusing on their contribution to metabolic disorders in the setting of obesity and discussing their value as therapeutic targets.
Collapse
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| | - Gianluca Ianiro
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Antonio Gasbarrini
- Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
2
|
Di Stasi V, Contaldo A, Birtolo LI, Shahini E. Interplay of Cardiometabolic Syndrome and Biliary Tract Cancer: A Comprehensive Analysis with Gender-Specific Insights. Cancers (Basel) 2024; 16:3432. [PMID: 39410050 PMCID: PMC11476000 DOI: 10.3390/cancers16193432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/05/2024] [Accepted: 10/09/2024] [Indexed: 10/20/2024] Open
Abstract
BTC overall incidence is globally increasing. CCA, including its subtypes, is a form of BTC. MetS, obesity, MASLD, and diabetes are all linked to CCA in interconnected ways. The link between obesity and CCA is less well-defined in Eastern countries as compared to Western. Although more research is needed to determine the relationship between MASLD and extrahepatic CCA (eCCA), MASLD may be a concurrent risk factor for intrahepatic CCA, particularly in populations with established or unidentified underlying liver disease. Interestingly, the risk of biliary tract cancer (BTC) seemed to be higher in patients with shorter diabetes durations who were not treated with insulin. Therefore, early detection and prevention of chronic liver disease, as well as additional intervention studies, will undoubtedly be required to determine whether improvements to MetS, weight loss, and diabetes therapy can reduce the risk and progression of BTC. However, further studies are needed to understand how reproductive hormones are involved in causing BTC and to develop consistent treatment for patients. Finally, it is critical to carefully assess the cardiological risk in BTC patients due to their increased intrinsic cardiovascular risk, putting them at risk for thrombotic complications, cardiovascular death, cardiac metastasis, and nonbacterial thrombotic endocarditis. This review aimed to provide an updated summary of the relation between the abovementioned cardio-metabolic conditions and BTC.
Collapse
Affiliation(s)
- Vincenza Di Stasi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Antonella Contaldo
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Umberto I Hospital, Sapienza University of Rome, 00185 Rome, Italy;
| | - Endrit Shahini
- Gastroenterology Unit, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
3
|
Ungvari Z, Fekete M, Varga P, Lehoczki A, Fekete JT, Ungvari A, Győrffy B. Overweight and obesity significantly increase colorectal cancer risk: a meta-analysis of 66 studies revealing a 25-57% elevation in risk. GeroScience 2024:10.1007/s11357-024-01375-x. [PMID: 39379738 DOI: 10.1007/s11357-024-01375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 10/10/2024] Open
Abstract
The incidence of colorectal cancer (CRC) has been steadily rising, and obesity has been identified as a significant risk factor. Numerous studies suggest a strong correlation between excess body weight and increased risk of CRC, but comprehensive quantification through pooled analysis remains limited. This study aims to systematically review and meta-analyze the existing literature to evaluate the association between obesity and CRC risk, considering variations across sex and study designs. A systematic literature search was conducted in PubMed, Cochrane Central Register of Controlled Trials (CENTRAL), and Web of Science to identify randomized controlled trials and human clinical trials from 1992 to 2024. Statistical analysis was performed using the https://metaanalysisonline.com web application using a random effects model to estimate the pooled hazard rates (HR). Forest plots, funnel plots, and Z-score plots were utilized to visualize results. We identified 52 clinical trials and 14 case-control studies, encompassing a total of 83,251,050 and 236,877 subjects, respectively. The pooled analysis indicated that obesity significantly increased the prevalence of CRC (HR = 1.36, 95% CI = 1.24-1.48, p < 0.01). This effect was consistent across sexes, with HRs of 1.57 (95% CI = 1.38-1.78, p = 0.01) for males and 1.25 (95% CI = 1.14-1.38, p < 0.01) for females. Case-control studies specifically showed an effect, but with marginal significance only (HR = 1.27, 95% CI = 0.98-1.65, p = 0.07). The Z-score plot indicated the need for additional analysis in the case-control group. A significant heterogeneity was observed across studies in all four settings. This meta-analysis provides robust evidence that obesity is a significant risk factor for colorectal cancer, with an overall hazard rate indicating a 36% increased risk. The effect is pronounced across both sexes, with males showing a slightly higher risk compared to females. Although case-control studies showed a weaker association, the overall trend supports the link between obesity and CRC. These results underscore the importance of public health interventions aimed at reducing obesity to potentially lower the risk of colorectal cancer.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral College/Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Institute of Preventive Medicine and Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Peter Varga
- Institute of Preventive Medicine and Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - Andrea Lehoczki
- Institute of Preventive Medicine and Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary
| | - János Tibor Fekete
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
| | - Anna Ungvari
- Institute of Preventive Medicine and Public Health, Semmelweis University, Semmelweis University, Budapest, Hungary.
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, 1094, Budapest, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, 1117, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, 7624, Pecs, Hungary
| |
Collapse
|
4
|
Fusco W, Bricca L, Kaitsas F, Tartaglia MF, Venturini I, Rugge M, Gasbarrini A, Cammarota G, Ianiro G. Gut microbiota in colorectal cancer: From pathogenesis to clinic. Best Pract Res Clin Gastroenterol 2024; 72:101941. [PMID: 39645279 DOI: 10.1016/j.bpg.2024.101941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/04/2024] [Indexed: 12/09/2024]
Abstract
Colorectal cancer is the third most common type of cancer, with a significant burden on healthcare and social systems. Its incidence is constantly rising, due to the spread of unhealthy lifestyle, i.e. Western diet. Increasing evidence suggests that westernization-driven microbiome alterations may play a critical role in colorectal tumorigenesis. The current screening strategies for this neoplasm, mainly fecal immunochemical tests, are burdened by unsatisfactory accuracy. Novel, non-invasive biomarkers are rising as the new frontier of colorectal cancer screening, and the microbiome-based ones are showing positive and optimistic results. This Review describes our current knowledge on the role of gut microbiota in colorectal cancer, from its pathogenetic action to its clinical potential as diagnostic biomarker.
Collapse
Affiliation(s)
- William Fusco
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy.
| | - Ludovica Bricca
- Department of Medicine - DIMED, Surgical Pathology and Cytopathology Unit, Università degli Studi di Padova, Padova, Italy
| | - Francesco Kaitsas
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Irene Venturini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Massimo Rugge
- Department of Medicine - DIMED, Surgical Pathology and Cytopathology Unit, Università degli Studi di Padova, Padova, Italy
| | - Antonio Gasbarrini
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Giovanni Cammarota
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
5
|
Anazco D, Acosta A, Cathcart-Rake EJ, D'Andre SD, Hurtado MD. Weight-centric prevention of cancer. OBESITY PILLARS 2024; 10:100106. [PMID: 38495815 PMCID: PMC10943063 DOI: 10.1016/j.obpill.2024.100106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 02/29/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024]
Abstract
Background The link between excess adiposity and carcinogenesis has been well established for multiple malignancies, and cancer is one of the main contributors to obesity-related mortality. The potential role of different weight-loss interventions on cancer risk modification has been assessed, however, its clinical implications remain to be determined. In this clinical review, we present the data assessing the effect of weight loss interventions on cancer risk. Methods In this clinical review, we conducted a comprehensive search of relevant literature using MEDLINE, Embase, Web of Science, and Google Scholar databases for relevant studies from inception to January 20, 2024. In this clinical review, we present systematic reviews and meta-analysis, randomized clinical trials, and prospective and retrospective observational studies that address the effect of different treatment modalities for obesity in cancer risk. In addition, we incorporate the opinions from experts in the field of obesity medicine and oncology regarding the potential of weight loss as a preventative intervention for cancer. Results Intentional weight loss achieved through different modalities has been associated with a reduced cancer incidence. To date, the effect of weight loss on the postmenopausal women population has been more widely studied, with multiple reports indicating a protective effect of weight loss on hormone-dependent malignancies. The effect of bariatric interventions as a protective intervention for cancer has been studied extensively, showing a significant reduction in cancer incidence and mortality, however, data for the effect of bariatric surgery on certain specific types of cancer is conflicting or limited. Conclusion Medical nutrition therapy, exercise, antiobesity medication, and bariatric interventions, might lead to a reduction in cancer risk through weight loss-dependent and independent factors. Further evidence is needed to better determine which population might benefit the most, and the amount of weight loss required to provide a clinically significant preventative effect.
Collapse
Affiliation(s)
- Diego Anazco
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Maria D. Hurtado
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
6
|
Yuan S, Ruan X, Sun Y, Fu T, Zhao J, Deng M, Chen J, Li X, Larsson SC. Birth weight, childhood obesity, adulthood obesity and body composition, and gastrointestinal diseases: a Mendelian randomization study. Obesity (Silver Spring) 2023; 31:2603-2614. [PMID: 37664887 DOI: 10.1002/oby.23857] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/15/2023] [Accepted: 05/02/2023] [Indexed: 09/05/2023]
Abstract
OBJECTIVE This Mendelian randomization study aimed to investigate the associations of birth weight, childhood BMI, and adulthood BMI, waist-hip ratio, and body composition with the risk of 24 gastrointestinal diseases. METHODS Independent genetic instruments associated with the exposures at the genome-wide significance level (p < 5 × 10-8 ) were selected from corresponding large-scale genome-wide association studies. Summary-level data for gastrointestinal diseases were obtained from the UK Biobank, the FinnGen study, and large consortia of European ancestry. RESULTS Genetically predicted higher levels of birth weight were associated with a lower risk of gastroesophageal reflux. Genetically predicted higher childhood BMI was associated with an increased risk of duodenal ulcer, nonalcoholic fatty liver disease, and cholelithiasis. However, the associations did not persist after adjusting for genetically predicted adulthood BMI. Genetically predicted higher adulthood BMI and waist-hip ratio were associated with 19 and 17 gastrointestinal diseases, respectively. Genetically predicted greater visceral adiposity was associated with an increased risk of 17 gastrointestinal diseases. There were no strong associations among genetically predicted whole-body fat and fat-free mass indices with gastrointestinal diseases. CONCLUSIONS This study suggests that greater adulthood adiposity, measured as either BMI, waist-hip ratio, or visceral adipose tissue, is causally associated with an increased risk of a broad range of gastrointestinal diseases in the European population.
Collapse
Affiliation(s)
- Shuai Yuan
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xixian Ruan
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yuhao Sun
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian Fu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianhui Zhao
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Minzi Deng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jie Chen
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xue Li
- Department of Big Data in Health Science, Center of Clinical Big Data and Analytics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Centre for Global Health, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Unit of Medical Epidemiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
7
|
Chavez-Tapia NC, Murúa-Beltrán Gall S, Ordoñez-Vázquez AL, Nuño-Lambarri N, Vidal-Cevallos P, Uribe M. Understanding the Role of Metabolic Syndrome as a Risk Factor for Hepatocellular Carcinoma. J Hepatocell Carcinoma 2022; 9:583-593. [PMID: 35818404 PMCID: PMC9270896 DOI: 10.2147/jhc.s283840] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/23/2022] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) and metabolic syndrome (MetS) have a rising prevalence worldwide. The relationship between these two entities has long been studied and understanding it has become a public health and clinical priority. This association follows, in most patients, the path through non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), cirrhosis and finally HCC. Nonetheless, increasing evidence has been found, that shows MetS as an independent risk factor for the development of HCC. This review brings together the clinical evidence of the relationship between these highly prevalent diseases, with a particular interest in the impact of each component of MetS on HCC; It aims to summarize the complex physiopathological pathways that explain this relationship, and to shed light on the different clinical scenarios of this association, the impact of treating the different components of MetS on the risk of HCC and what is known about screening for HCC in patients with MetS. By doing so, it hopes to improve awareness on this topic.
Collapse
Affiliation(s)
- Norberto C Chavez-Tapia
- Gastroenterology Department, Medica Sur Clinic & Foundation, Mexico City, Mexico
- Transational Research Department, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | | | - Natalia Nuño-Lambarri
- Transational Research Department, Medica Sur Clinic & Foundation, Mexico City, Mexico
| | | | - Misael Uribe
- Gastroenterology Department, Medica Sur Clinic & Foundation, Mexico City, Mexico
| |
Collapse
|
8
|
Exercise suppresses tumor growth independent of high fat food intake and associated immune dysfunction. Sci Rep 2022; 12:5476. [PMID: 35361802 PMCID: PMC8971502 DOI: 10.1038/s41598-022-08850-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 03/11/2022] [Indexed: 11/24/2022] Open
Abstract
Epidemiological data suggest that exercise training protects from cancer independent of BMI. Here, we aimed to elucidate mechanisms involved in voluntary wheel running-dependent control of tumor growth across chow and high-fat diets. Access to running wheels decreased tumor growth in B16F10 tumor-bearing on chow (− 50%) or high-fat diets (− 75%, p < 0.001), however, tumor growth was augmented in high-fat fed mice (+ 53%, p < 0.001). Tumor growth correlated with serum glucose (p < 0.01), leptin (p < 0.01), and ghrelin levels (p < 0.01), but not with serum insulin levels. Voluntary wheel running increased immune recognition of tumors as determined by microarray analysis and gene expression analysis of markers of macrophages, NK and T cells, but the induction of markers of macrophages and NK cells was attenuated with high-fat feeding. Moreover, we found that the regulator of innate immunity, ZBP1, was induced by wheel running, attenuated by high-fat feeding and associated with innate immune recognition in the B16F10 tumors. We observed no effects of ZBP1 on cell cycle arrest, or exercise-regulated necrosis in the tumors of running mice. Taken together, our data support epidemiological findings showing that exercise suppresses tumor growth independent of BMI, however, our data suggest that high-fat feeding attenuates exercise-mediated immune recognition of tumors.
Collapse
|
9
|
Safiri S, Karamzad N, Kaufman JS, Nejadghaderi SA, Bragazzi NL, Sullman MJM, Almasi-Hashiani A, Mansournia MA, Collins GS, Kolahi AA, Jemal A. Global, regional, and national burden of cancers attributable to excess body weight in 204 countries and territories, 1990 to 2019. Obesity (Silver Spring) 2022; 30:535-545. [PMID: 35041300 DOI: 10.1002/oby.23355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/02/2021] [Accepted: 11/15/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE The aim of this study was to report the level and trends of 13 cancers that are attributable to excess body weight (EBW) for 204 countries and territories from 1990 to 2019. METHODS Using publicly available data, the burden of cancers attributable to EBW was reported from 1990 to 2019 based on the comparative risk assessment approach used in the Global Burden of Disease study 2019. [Correction added on 27 January 2022, after first online publication: 'Using publicly available data,' has been added before the first sentence and 'estimated' was corrected to 'reported'.] RESULTS: In 2019, EBW caused 11.2 million disability-adjusted life-years (DALYs), or 4.4% of all cancer-related DALYs. Between 1990 and 2019, the global EBW-attributable age-standardized cancer DALY rates (per 100,000) increased from 109.9 to 133.9, a relative increase of 21.9%. The age-standardized DALY rates (per 100,000) of cancers attributable to EBW in 2019 were highest and lowest in Mongolia (611.8) and Bangladesh (30.2), respectively. The 60- to 64-year age group had the highest number of DALYs attributable to EBW, whereas there were no large sex differences in the cancer-related burden attributable to EBW. Furthermore, the association between the age-standardized DALY rates and the sociodemographic index was generally positive. CONCLUSIONS Overall, the EBW-attributable burden of cancers has increased in the past three decades. Public health efforts should focus on identifying appropriate preventive interventions at the population and individual levels, especially in the regions and countries with the highest burden.
Collapse
Affiliation(s)
- Saeid Safiri
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nahid Karamzad
- Nutrition Research Center, Department of Biochemistry and Diet Therapy, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jay S Kaufman
- Department of Epidemiology, Biostatistics and Occupational Health, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Seyed Aria Nejadghaderi
- Social Determinants of Health Research Center, Department of Community Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Systematic Review and Meta-analysis Expert Group, Universal Scientific Education and Research Network, Tehran, Iran
| | | | - Mark J M Sullman
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Mohammad Ali Mansournia
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gary S Collins
- Centre for Statistics in Medicine, NDORMS, Botnar Research Centre, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Ali-Asghar Kolahi
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmedin Jemal
- Surveillance and Health Services Research, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
10
|
Zhou J, Sun S, Luan S, Xiao X, Yang Y, Mao C, Chen L, Zeng X, Zhang Y, Yuan Y. Gut Microbiota for Esophageal Cancer: Role in Carcinogenesis and Clinical Implications. Front Oncol 2021; 11:717242. [PMID: 34733778 PMCID: PMC8558403 DOI: 10.3389/fonc.2021.717242] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 09/24/2021] [Indexed: 02/05/2023] Open
Abstract
Esophageal cancer (EC) is a common malignant tumor of the upper digestive tract. The microbiota in the digestive tract epithelium comprises a large number of microorganisms that adapt to the immune defense and interact with the host to form symbiotic networks, which affect many physiological processes such as metabolism, tissue development, and immune response. Reports indicate that there are microbial compositional changes in patients with EC, which provides an important opportunity to advance clinical applications based on findings on the gut microbiota. For example, microbiota detection can be used as a biomarker for screening and prognosis, and microorganism levels can be adjusted to treat cancer and decrease the adverse effects of treatment. This review aims to provide an outline of the gut microbiota in esophageal neoplasia, including the mechanisms involved in microbiota-related carcinogenesis and the prospect of utilizing the microbiota as EC biomarkers and treatment targets. These findings have important implications for translating the use of gut microbiota in clinical applications.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shangwei Sun
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Siyuan Luan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Xiao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yushang Yang
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chengyi Mao
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Longqi Chen
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Zhang
- Department of Periodical Press, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Nursing Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Yuan
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Obesity and Pancreatic Cancer: Insight into Mechanisms. Cancers (Basel) 2021; 13:cancers13205067. [PMID: 34680216 PMCID: PMC8534007 DOI: 10.3390/cancers13205067] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Obesity is recognized as a chronic progressive disease and risk factor for many human diseases. The high and increasing number of obese people may underlie the expected increase in pancreatic cancer cases in the United States. There are several pathways discussed that link obesity with pancreatic cancer. Adipose tissue and adipose tissue-released factors may thereby play an important role. This review discusses selected mechanisms that may accelerate pancreatic cancer development in obesity. Abstract The prevalence of obesity in adults and children has dramatically increased over the past decades. Obesity has been declared a chronic progressive disease and is a risk factor for a number of metabolic, inflammatory, and neoplastic diseases. There is clear epidemiologic and preclinical evidence that obesity is a risk factor for pancreatic cancer. Among various potential mechanisms linking obesity with pancreatic cancer, the adipose tissue and obesity-associated adipose tissue inflammation play a central role. The current review discusses selected topics and mechanisms that attracted recent interest and that may underlie the promoting effects of obesity in pancreatic cancer. These topics include the impact of obesity on KRAS activity, the role of visceral adipose tissue, intrapancreatic fat, adipose tissue inflammation, and adipokines on pancreatic cancer development. Current research on lipocalin-2, fibroblast growth factor 21, and Wnt5a is discussed. Furthermore, the significance of obesity-associated insulin resistance with hyperinsulinemia and obesity-induced gut dysbiosis with metabolic endotoxemia is reviewed. Given the central role that is occupied by the adipose tissue in obesity-promoted pancreatic cancer development, preventive and interceptive strategies should be aimed at attenuating obesity-associated adipose tissue inflammation and/or at targeting specific molecules that mechanistically link adipose tissue with pancreatic cancer in obese patients.
Collapse
|
12
|
Zhang Y, Song M, Chan AT, Schernhammer ES, Wolpin BM, Stampfer MJ, Meyerhardt JA, Fuchs CS, Roberts SB, Willett WC, Hu FB, Giovannucci EL, Ng K. Unrestrained eating behavior and risk of digestive system cancers: a prospective cohort study. Am J Clin Nutr 2021; 114:1612-1624. [PMID: 34293086 PMCID: PMC8588850 DOI: 10.1093/ajcn/nqab235] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Unrestrained eating behavior, as a potential proxy for diet frequency, timing, and caloric intake, has been questioned as a plausible risk factor for digestive system cancers, but epidemiological evidence remains sparse. OBJECTIVES We investigated prospectively the associations between unrestrained eating behavior and digestive system cancer risk. METHODS Participants in the Nurses' Health Study who were free of cancer and reported dietary information in 1994 were followed for ≤18 y. Cox models were used to estimate HRs and 95% CIs for unrestrained eating (eating anything at any time, no concern with figure change, or both) and risk of digestive system cancers. RESULTS During follow-up, 2064 digestive system cancer cases were documented among 70,450 eligible participants in analyses of eating anything at any time, In total, 2081 digestive system cancer cases were documented among 72,468 eligible participants in analyses of no concern with figure change. In fully adjusted analyses, women with the behavior of eating anything at any time had a higher risk of overall digestive system cancer (HR: 1.22; 95% CI: 1.10, 1.35), overall gastrointestinal tract cancer ((HR: 1.33; 95% CI: 1.18, 1.50), buccal cavity and pharynx cancer (HR: 1.50; 95% CI: 1.02, 2.21), esophageal cancer (HR: 1.62; 95% CI: 1.01, 2.62), small intestine cancer (HR: 1.92; 95% CI: 1.02,3. 59), and colorectal cancer (HR: 1.20; 95% CI: 1.04, 1.38), and a non-statistically significant increased risk of stomach cancer (HR: 1.54; 95% CI: 0.96,2.48), compared with women without this behavior. No statistically significant association was observed for pancreatic cancer and liver and gallbladder cancer. The combined effect of eating anything at any time and having no concern with figure change was associated with a significantly increased risk of overall digestive system cancer (HR: 1.27; 95% CI: 1.10, 1.46), overall gastrointestinal tract cancer (HR: 1.45; 95% CI: 1.23, 1.71), and colorectal cancer (HR: 1.34; 95% CI: 1.11, 1.63), compared with women exhibiting the opposite. CONCLUSIONS Unrestrained eating behavior was independently associated with increased risk of gastrointestinal tract cancers. The potential importance of unrestrained eating behavior modification in preventing gastrointestinal tract cancers should be noted.
Collapse
Affiliation(s)
- Yin Zhang
- Address correspondence to YZ (emails: and )
| | - Mingyang Song
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eva S Schernhammer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Center of Public Health, Medical University of Vienna, Vienna, Austria
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Meir J Stampfer
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Smilow Cancer Hospital and Yale Cancer Center, New Haven, CT, USA
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Susan B Roberts
- USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Walter C Willett
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
Akkiz H, Carr BI, Guerra V, Donghia R, Yalçın K, Karaoğullarından U, Altıntaş E, Özakyol A, Şimşek H, Balaban HY, Balkan A, Uyanıkoğlu A, Ekin N, Delik A. Plasma lipids, tumor parameters and survival in HCC patients with HBV and HCV. JOURNAL OF TRANSLATIONAL SCIENCE 2021; 7:10.15761/jts.1000421. [PMID: 34457356 PMCID: PMC8389344 DOI: 10.15761/jts.1000421] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION AND AIMS Hepatocellular carcinoma (HCC) is a consequence of chronic liver disease, particularly from hepatitis B or C and increasingly from obesity and metabolic syndrome. Since lipids are an important component of cell membranes and are involved in cell signaling and tumor cell growth, we wished to evaluate the relationship between HCC patient plasma lipids and maximum tumor diameter and other indices of HCC human biology. METHODS We examined prospectively-collected data from a multi-institutional collaborative Turkish HCC working group, from predominantly HBV-based patients, for plasma lipid profiles, consisting of triglycerides, total cholesterol, LDL-cholesterol (LDL) and HDL-cholesterol (HDL) and compared these with the associated patient maximum tumor diameter (MTD), portal vein thrombosis, alpha-fetoprotein (AFP) and also with patient survival. RESULTS We found that both low HDL (p=0.0002) and high LDL (p=0.003) levels were significantly associated with increased MTD, as well as in a final multiple linear regression model on MTD. The combination of low HDL combined with high HDL levels were significant in a regression model on MTD, PVT and an HCC Aggressiveness Index (Odds Ratio 12.91 compared to an Odds Ratio of 1 for the reference). Furthermore, in a Cox regression model on death, the HDL plus LDL combination had a significantly higher Hazard Ratio than the reference category. CONCLUSIONS Low plasma HDL, high plasma LDL and especially the combination, were significantly related to more aggressive HCC phenotype and the combination was significantly related to a higher Hazard Ratio for death.
Collapse
Affiliation(s)
- H Akkiz
- Çukurova University, Adana, Turkey
| | - BI Carr
- İnonu University, Malatya, Turkey
| | - V Guerra
- National Institute of Gastroenterology, S. de Bellis Research hospital, Castellana Grotte (BA), Italy
| | - R Donghia
- National Institute of Gastroenterology, S. de Bellis Research hospital, Castellana Grotte (BA), Italy
| | - K Yalçın
- Dikle University, Diyarbakır, Turkey
| | | | | | - A Özakyol
- Eskişehir Osmangazi University, Eskişehir, Turkey
| | - H Şimşek
- Hacettepe University, Ankara, Turkey
| | | | - A Balkan
- Gaziantep University, Gazientep, Turkey
| | | | - N Ekin
- Dikle University, Diyarbakır, Turkey
| | - A Delik
- Çukurova University, Adana, Turkey
| |
Collapse
|
14
|
Dimou NL, Papadimitriou N, Mariosa D, Johansson M, Brennan P, Peters U, Chanock SJ, Purdue M, Bishop DT, Gago‐Dominquez M, Giles GG, Moreno V, Platz EA, Tangen CM, Wolk A, Zheng W, Wu X, Campbell PT, Giovannucci E, Lin Y, Gunter MJ, Murphy N. Circulating adipokine concentrations and risk of five obesity-related cancers: A Mendelian randomization study. Int J Cancer 2021; 148:1625-1636. [PMID: 33038280 PMCID: PMC7894468 DOI: 10.1002/ijc.33338] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/27/2020] [Accepted: 09/17/2020] [Indexed: 12/20/2022]
Abstract
Obesity is considered a chronic inflammatory state characterized by continued secretion of adipokines and cytokines. Experimental and epidemiological evidence indicates that circulating adipokines may be associated with the development of obesity-related cancers, but it is unclear if these associations are causal or confounded. We examined potential causal associations of specific adipokines (adiponectin, leptin, soluble leptin receptor [sOB-R] and plasminogen activator inhibitor-1 [PAI-1]) with five obesity-related cancers (colorectal, pancreatic, renal cell carcinoma [RCC], ovarian and endometrial) using Mendelian randomization (MR) methods. We used summary-level data from large genetic consortia for 114 530 cancer cases and 245 284 controls. We constructed genetic instruments using 18 genetic variants for adiponectin, 2 for leptin and 4 for both sOB-R and PAI-1 (P value for inclusion<5 × 10-8 ). Causal estimates were obtained using two-sample MR methods. In the inverse-variance weighted models, we found an inverse association between adiponectin and risk of colorectal cancer (odds ratio per 1 μg/mL increment in adiponectin concentration: 0.90 [95% confidence interval = 0.84-0.97]; P = .01); but, evidence of horizontal pleiotropy was detected and the association was not present when this was taken into consideration. No association was found for adiponectin and risks of pancreatic cancer, RCC, ovarian cancer and endometrial cancer. Leptin, sOB-R and PAI-1 were also similarly unrelated to risk of obesity-related cancers. Despite the large sample size, our MR analyses do not support causal effects of circulating adiponectin, leptin, sOB-R and PAI-1 concentrations on the development of five obesity-related cancers.
Collapse
Affiliation(s)
- Niki L. Dimou
- Section of Nutrition and Metabolism, International Agency for Research on CancerLyonFrance
| | - Nikos Papadimitriou
- Section of Nutrition and Metabolism, International Agency for Research on CancerLyonFrance
| | - Daniela Mariosa
- Section of Genetics, International Agency for Research on CancerLyonFrance
| | - Mattias Johansson
- Section of Genetics, International Agency for Research on CancerLyonFrance
| | - Paul Brennan
- Section of Genetics, International Agency for Research on CancerLyonFrance
| | - Ulrike Peters
- Fred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Stephen J. Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Mark Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | | | - Manuela Gago‐Dominquez
- Fundación Gallega de Medicina Genómica, Grupo de Genéticadel CáncerInstituto de Investigación Sanitaria de Santiago IDISComplejo Hospitalario Univ. Santiago‐CHUS, SERGAS, Santiago de CompostelaSpain
- Moores Cancer CenterUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Graham G. Giles
- Cancer Epidemiology DivisionCancer Council VictoriaMelbourneVictoriaAustralia
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global HealthThe University of MelbourneMelbourneVictoriaAustralia
- Precision MedicineSchool of Clinical Sciences at Monash Health, Monash UniversityClaytonVictoriaAustralia
| | - Victor Moreno
- Oncology Data Analytics ProgramCatalan Institute of Oncology‐IDIBELL, L'Hospitalet de LlobregatBarcelonaSpain
- CIBER Epidemiología y SaludPública (CIBERESP)MadridSpain
- Department of Clinical Sciences, Faculty of MedicineUniversity of BarcelonaBarcelonaSpain
- ONCOBEL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de LlobregatBarcelonaSpain
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public HealthBaltimoreMarylandUSA
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | - Alicja Wolk
- Institute of Environmental Medicine, Karolinska InstitutetStockholmSweden
- Department of Surgical SciencesUppsala UniversityUppsalaSweden
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt‐Ingram Cancer CenterVanderbilt UniversityNashvilleTennesseeUSA
| | - Xifeng Wu
- Department of EpidemiologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
- Department of Precision Health and Data Science, School of Public Health and the Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Peter T. Campbell
- Behavioral and Epidemiology Research Group, American Cancer SocietyAtlantaGeorgiaUSA
| | - Edward Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthHarvard UniversityBostonMassachusettsUSA
- Department of NutritionT.H. Chan School of Public HealthBostonMassachusettsUSA
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research CenterSeattleWashingtonUSA
| | | | - Marc J. Gunter
- Section of Nutrition and Metabolism, International Agency for Research on CancerLyonFrance
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on CancerLyonFrance
| |
Collapse
|
15
|
Malik PRA, Doumouras AG, Malhan RS, Lee Y, Boudreau V, Barlow K, Karpinski M, Anvari M. Obesity, Cancer, and Risk Reduction with Bariatric Surgery. Surg Clin North Am 2021; 101:239-254. [PMID: 33743967 DOI: 10.1016/j.suc.2020.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The prevalence of noncommunicable diseases has increased dramatically in North America and throughout the world and is expected to continue increasing in coming years. Obesity has been linked to several types of cancers and is associated with increased morbidity and mortality following cancer diagnosis. Bariatric surgery has emerged as the prominent model to evaluate the effects of intentional weight loss on cancer incidence and outcomes. Current literature, comprising prospective cohort investigations, indicates site-specific reductions in cancer risk with select bariatric procedures. Future research is required to establish evidence-based indications for bariatric surgery in the context of cancer prevention.
Collapse
Affiliation(s)
- Peter R A Malik
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada; Department of Health Research Methods, Evidence & Impact, Faculty of Health Sciences, McMaster University Medical Centre, 1280 Main Street West, 2C Area, Hamilton, Ontario L8S 4K1, Canada
| | - Aristithes G Doumouras
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada
| | - Roshan S Malhan
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada
| | - Yung Lee
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada
| | - Vanessa Boudreau
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada; Department of Health Research Methods, Evidence & Impact, Faculty of Health Sciences, McMaster University Medical Centre, 1280 Main Street West, 2C Area, Hamilton, Ontario L8S 4K1, Canada
| | - Karen Barlow
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada
| | - Marta Karpinski
- Department of Health Research Methods, Evidence & Impact, Faculty of Health Sciences, McMaster University Medical Centre, 1280 Main Street West, 2C Area, Hamilton, Ontario L8S 4K1, Canada
| | - Mehran Anvari
- Department of Surgery and Centre for Minimal Access Surgery, St. Joseph's Healthcare, 50 Charlton Avenue East, Rm T2141 Hamilton, Ontario L8N 4A6, Canada.
| |
Collapse
|
16
|
Besutti G, Damato A, Venturelli F, Bonelli C, Vicentini M, Monelli F, Mancuso P, Ligabue G, Pattacini P, Pinto C, Giorgi Rossi P. Baseline liver steatosis has no impact on liver metastases and overall survival in rectal cancer patients. BMC Cancer 2021; 21:253. [PMID: 33750342 PMCID: PMC7941741 DOI: 10.1186/s12885-021-07980-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/24/2021] [Indexed: 01/15/2023] Open
Abstract
Background The liver is one of the most frequent sites of metastases in rectal cancer. This study aimed to evaluate how the development of synchronous or metachronous liver metastasis and overall survival are impacted by baseline liver steatosis and chemotherapy-induced liver damage in rectal cancer patients. Methods Patients diagnosed with stage II to IV rectal cancer between 2010 and 2016 in our province with suitable baseline CT scan were included. Data on cancer diagnosis, staging, therapy, outcomes and liver function were collected. CT scans were retrospectively reviewed to assess baseline steatosis (liver density < 48 HU and/or liver-to-spleen ratio < 1.1). Among patients without baseline steatosis and treated with neoadjuvant chemotherapy, chemotherapy-induced liver damage was defined as steatosis appearance, ≥ 10% liver volume increase, or significant increase in liver function tests. Results We included 283 stage II to IV rectal cancer patients with suitable CT scan (41% females; mean age 68 ± 14 years). Steatosis was present at baseline in 90 (31.8%) patients, synchronous liver metastasis in 42 (15%) patients and metachronous liver metastasis in 26 (11%); 152 (54%) deaths were registered. The prevalence of synchronous liver metastasis was higher in patients with steatosis (19% vs 13%), while the incidence of metachronous liver metastasis was similar. After correcting for age, sex, stage, and year of diagnosis, steatosis was not associated with metachronous liver metastasis nor with overall survival. In a small analysis of 63 patients without baseline steatosis and treated with neoadjuvant chemotherapy, chemotherapy-induced liver damage was associated with higher incidence of metachronous liver metastasis and worse survival, results which need to be confirmed by larger studies. Conclusions Our data suggest that rectal cancer patients with steatosis had a similar occurrence of metastases during follow-up, even if the burden of liver metastases at diagnosis was slightly higher, compatible with chance. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07980-9.
Collapse
Affiliation(s)
- Giulia Besutti
- Clinical and Experimental Medicine PhD program, University of Modena and Reggio Emilia, Modena, Italy.,Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Angela Damato
- Medical Oncology Unit, AUSL-IRCCS of Reggio Emilia, Viale Risorgimento 80, 42123, Reggio Emilia, Italy.,Department of Medical Biotechnologies, University of Siena, Strada delle Scotte 4, 53100, Siena, Italy
| | - Francesco Venturelli
- Epidemiology Unit, AUSL-IRCCS of Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Candida Bonelli
- Medical Oncology Unit, AUSL-IRCCS of Reggio Emilia, Viale Risorgimento 80, 42123, Reggio Emilia, Italy
| | - Massimo Vicentini
- Epidemiology Unit, AUSL-IRCCS of Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Filippo Monelli
- Clinical and Experimental Medicine PhD program, University of Modena and Reggio Emilia, Modena, Italy. .,Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Pamela Mancuso
- Epidemiology Unit, AUSL-IRCCS of Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Guido Ligabue
- Department of Radiology, Azienda Ospedaliero-Universitaria Policlinico di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Pierpaolo Pattacini
- Radiology Unit, Department of Diagnostic Imaging and Laboratory Medicine, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carmine Pinto
- Medical Oncology Unit, AUSL-IRCCS of Reggio Emilia, Viale Risorgimento 80, 42123, Reggio Emilia, Italy
| | - Paolo Giorgi Rossi
- Epidemiology Unit, AUSL-IRCCS of Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| |
Collapse
|
17
|
Teper Y, Eibl G. Pancreatic Macrophages: Critical Players in Obesity-Promoted Pancreatic Cancer. Cancers (Basel) 2020; 12:cancers12071946. [PMID: 32709161 PMCID: PMC7409049 DOI: 10.3390/cancers12071946] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 02/07/2023] Open
Abstract
Obesity is a known risk factor for the development of pancreatic cancer, one of the deadliest types of malignancies. In recent years it has become clear that the pancreatic microenvironment is critically involved and a contributing factor in accelerating pancreatic neoplasia. In this context obesity-associated chronic inflammation plays an important role. Among several immune cells, macrophages have been shown to contribute to obesity-induced tissue inflammation. This review article summarizes the current knowledge about the role of pancreatic macrophages in early pancreatic cancer development. It describes the heterogenous origin and mixture of pancreatic macrophages, their role in pancreatic endocrine and exocrine pathology, and the impact of obesity on islet and stromal macrophages. A model is postulated, by which during obesity monocytes are recruited into the pancreas, where they are polarized into pro-inflammatory macrophages that drive early pancreatic neoplasia. This occurs in the presence of local inflammatory, metabolic, and endocrine signals. A stronger appreciation and more detailed knowledge about the role of macrophages in early pancreatic cancer development will lead to innovative preventive or interceptive strategies.
Collapse
|
18
|
Abstract
Obesity is second only to tobacco as a preventable cause of cancer in the US. By multifactorial and often additive mechanisms, obesity leads to the development and promotion of 40% of the cancers diagnosed in this country, including post-menopausal breast, endometrial, colorectal, kidney, liver and pancreatic cancers, among others. Though prevention of obesity should be the ultimate goal of thoughtful and effective healthcare practices, it remains a highly prevalent condition, and morbid obesity (BMI ≥40 Kg/m2) can be refractory to lifestyle interventions in many cases. Currently bariatric surgery is an effective treatment strategy for individuals who suffer from morbid obesity or obesity with associated co-morbidities and fail to lose weight under a medically supervised diet and exercise program. The current review addresses seminal studies that have investigated the potential cancer prevention effects of bariatric surgery, demonstrating a positive impact mostly in post-menopausal breast and endometrial cancers. The controversial association between bariatric surgery and increased colorectal cancer (CRC) risk is also recognized and discussed. Finally, while bariatric surgery should not be routinely recommended as a cancer prevention strategy, it has the potential to decrease the risk for certain types of cancers as a collateral beneficial effect.
Collapse
Affiliation(s)
- Debora S Bruno
- Hematology Oncology Division, Department of Medicine, Genetics & Genome Sciences, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Nathan A Berger
- Hematology Oncology Division, Department of Medicine, Genetics & Genome Sciences, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Department Biochemistry, Genetics & Genome Sciences, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
19
|
Abstract
The preventability estimate for colorectal cancer (CRC) is approximately 50%, highlighting the huge potential for altering modifiable lifestyle factors (including diet and body fatness) in order to reduce risk of this common malignancy. There is strong evidence that dietary factors (including intake of wholegrains, fibre, red and processed meat and alcohol) affect CRC risk. The lack of positive intervention trials and limited mechanistic understanding likely explain limited public health impact of epidemiological observations, to date. An alternative strategy for nutritional prevention of CRC is use of supplements that provide higher individual nutrient exposure than obtained through the diet (chemoprevention). There are positive data for calcium and/or vitamin D and the n-3 fatty acid EPA from polyp prevention trials using colorectal adenoma as a CRC risk biomarker. Although CRC is an obesity-related malignancy, there remains a paucity of observational data supporting intentional weight loss for CRC risk reduction. Some types of obesity surgeries (Roux-en-Y gastric bypass) might actually increase subsequent CRC risk due to alteration of local intestinal factors. There is intense interest in nutritional therapy of patients after diagnosis of CRC, in order to impact on recurrence and overall survival (now often termed cancer interception). In conclusion, nutritional prevention of CRC continues to hold much promise. Increased mechanistic understanding of the role of individual nutrients (linked to intestinal microbiota), as well as a precision medicine approach to CRC chemoprevention and interception based on both tumour and host factors, should enable translation of nutritional interventions into effective CRC risk reduction measures.
Collapse
|
20
|
Wilkins E, Aravani A, Downing A, Drewnowski A, Griffiths C, Zwolinsky S, Birkin M, Alvanides S, Morris MA. Evidence from big data in obesity research: international case studies. Int J Obes (Lond) 2020; 44:1028-1040. [PMID: 31988482 DOI: 10.1038/s41366-020-0532-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND/OBJECTIVE Obesity is thought to be the product of over 100 different factors, interacting as a complex system over multiple levels. Understanding the drivers of obesity requires considerable data, which are challenging, costly and time-consuming to collect through traditional means. Use of 'big data' presents a potential solution to this challenge. Big data is defined by Delphi consensus as: always digital, has a large sample size, and a large volume or variety or velocity of variables that require additional computing power (Vogel et al. Int J Obes. 2019). 'Additional computing power' introduces the concept of big data analytics. The aim of this paper is to showcase international research case studies presented during a seminar series held by the Economic and Social Research Council (ESRC) Strategic Network for Obesity in the UK. These are intended to provide an in-depth view of how big data can be used in obesity research, and the specific benefits, limitations and challenges encountered. METHODS AND RESULTS Three case studies are presented. The first investigated the influence of the built environment on physical activity. It used spatial data on green spaces and exercise facilities alongside individual-level data on physical activity and swipe card entry to leisure centres, collected as part of a local authority exercise class initiative. The second used a variety of linked electronic health datasets to investigate associations between obesity surgery and the risk of developing cancer. The third used data on tax parcel values alongside data from the Seattle Obesity Study to investigate sociodemographic determinants of obesity in Seattle. CONCLUSIONS The case studies demonstrated how big data could be used to augment traditional data to capture a broader range of variables in the obesity system. They also showed that big data can present improvements over traditional data in relation to size, coverage, temporality, and objectivity of measures. However, the case studies also encountered challenges or limitations; particularly in relation to hidden/unforeseen biases and lack of contextual information. Overall, despite challenges, big data presents a relatively untapped resource that shows promise in helping to understand drivers of obesity.
Collapse
Affiliation(s)
- Emma Wilkins
- Leeds Institute for Data Analytics and School of Medicine, University of Leeds, Leeds, UK
| | - Ariadni Aravani
- Leeds Institute for Data Analytics and School of Medicine, University of Leeds, Leeds, UK
| | - Amy Downing
- Leeds Institute for Data Analytics and School of Medicine, University of Leeds, Leeds, UK
| | - Adam Drewnowski
- Center for Public Health Nutrition, University of Washington, Seattle, WA, USA
| | | | | | - Mark Birkin
- Leeds Institute for Data Analytics and School of Geography, University of Leeds, Leeds, UK
| | - Seraphim Alvanides
- Engineering and Environment, Northumbria University, Newcastle, UK.,GESIS-Leibniz Institute for the Social Sciences, Cologne, Germany
| | - Michelle A Morris
- Leeds Institute for Data Analytics and School of Medicine, University of Leeds, Leeds, UK.
| |
Collapse
|
21
|
Almazeedi S, El-Abd R, Al-Khamis A, Albatineh AN, Al-Sabah S. Role of bariatric surgery in reducing the risk of colorectal cancer: a meta-analysis. Br J Surg 2020; 107:348-354. [PMID: 31976551 DOI: 10.1002/bjs.11494] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/25/2019] [Accepted: 11/30/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND Obesity increases the risk of multiple co-morbidities such as type 2 diabetes, cardiovascular disease and most cancers, including colorectal cancer. Currently, the literature presents conflicting results regarding the protective effects of bariatric surgery on the incidence of colorectal cancer. This meta-analysis was conducted to investigate the effect of bariatric surgery on the risk of developing colorectal cancer in obese individuals. METHODS Ovid Embase, Ovid MEDLINE, Cochrane CENTRAL and Web of Science were searched for relevant articles. Articles published by the end of December 2018 were retrieved; data were extracted according to evidence-based PICO (population, intervention, control, outcome) model and analysed using a random-effects model to estimate the pooled relative risk (RR) and its 95 per cent confidence interval. The heterogeneity of studies was tested and quantified using Cochran's Q and I2 statistics. Meta-regression was used to investigate the association of year of study, region, mean length of follow-up and sample size with RR. RESULTS Seven articles, involving a total of 1 213 727 patients, were included in the meta-analysis. The pooled estimate of the RR was 0·64 (95 per cent c.i. 0·42 to 0·98). The test of asymmetry found no significant publication bias. Meta-regression showed that sample size was a statistically significant factor (P = 0·037), but year of publication, region and mean duration of follow-up were not significant. CONCLUSION Patients who underwent bariatric surgery had a greater than 35 per cent reduction in the risk of developing colorectal cancer compared with obese individuals who had no surgery.
Collapse
Affiliation(s)
- S Almazeedi
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
| | - R El-Abd
- Department of Community Medicine and Behavioural Sciences, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - A Al-Khamis
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
| | - A N Albatineh
- Department of Community Medicine and Behavioural Sciences, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - S Al-Sabah
- Department of Surgery, Jaber Al-Ahmed Hospital, Kuwait City, Kuwait
| |
Collapse
|
22
|
Castagneto-Gissey L, Casella-Mariolo J, Casella G, Mingrone G. Obesity Surgery and Cancer: What Are the Unanswered Questions? Front Endocrinol (Lausanne) 2020; 11:213. [PMID: 32351453 PMCID: PMC7174700 DOI: 10.3389/fendo.2020.00213] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 03/25/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity has become a global epidemic with a soaring economic encumbrance due to its related morbidity and mortality. Amongst obesity-related conditions, cancer is indeed the most redoubtable. Bariatric surgery has been proven to be the most effective treatment for obesity and its associated metabolic and cardiovascular disorders. However, the understanding of whether and how bariatric surgery determines a reduction in cancer risk is limited. Obesity-related malignancies primarily include colorectal and hormone-sensitive (endometrium, breast, prostate) cancers. Additionally, esophago-gastric tumors are growing to be recognized as a new category mainly associated with post-bariatric surgery outcomes. In fact, certain types of surgical procedures have been described to induce the development and subsequent progression of pre-cancerous esophageal and gastric lesions. This emerging category is of great concern and further research is required to possibly prevent such risks. Published data has generated conflicting results. In fact, while overall cancer risk reduction was reported particularly in women, some authors showed no improvement or even increased cancer incidence. Although various studies have reported beneficial effects of surgery on risk of specific cancer development, fundamental insights into the pathogenesis of obesity-related cancer are indispensable to fully elucidate its mechanisms.
Collapse
Affiliation(s)
- Lidia Castagneto-Gissey
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
- *Correspondence: Lidia Castagneto-Gissey
| | | | - Giovanni Casella
- Department of Surgical Sciences, Sapienza University of Rome, Rome, Italy
| | - Geltrude Mingrone
- Division of Diabetes & Nutritional Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Division of Diabetes & Nutritional Sciences, Università Cattolica del Sacro Cuore Rome, Rome, Italy
- Division of Diabetes & Nutritional Sciences, Faculty of Life Sciences & Medicine, King's College London, London, United Kingdom
| |
Collapse
|
23
|
Lv Y, Patel N, Zhang HJ. The progress of non-alcoholic fatty liver disease as the risk of liver metastasis in colorectal cancer. Expert Rev Gastroenterol Hepatol 2019; 13:1169-1180. [PMID: 31774328 DOI: 10.1080/17474124.2019.1697231] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: The liver is the most common metastatic site of colorectal cancer (CRC), and the long-term survival rate of CRC patients who cannot resect liver metastatic lesions radically is extremely low. Early identification of risk factors for liver metastasis from CRC may be an effective strategy to reduce the incidence of liver metastasis. The incidence of non-alcoholic fatty liver disease (NAFLD) is increasing in parallel with an increasing prevalence of obesity and metabolic syndrome (MS), which has become one of the main causes of chronic liver disease worldwide.Areas covered: An overview of the related research progress of the association between NAFLD and colorectal liver metastasis (CRLM).Expert opinion: Certain research proves that there is a close relationship between NAFLD and CRC, and the presence of NAFLD can promote the formation and development of CRC. Although the effect of liver diseases on the incidence of liver metastasis in CRC has been noted in recent years, the results are inconsistent and haven't reached a unified conclusion. Therefore, the association between liver metastasis and NAFLD remains the main focal point in the evolution of CRC.
Collapse
Affiliation(s)
- Yan Lv
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, P.R. China
| | - Nishant Patel
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, P.R. China
| | - Hai-Jun Zhang
- Department of Oncology, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, P.R. China.,Precision Medicine Center, Zhongda Hospital, Medical School of Southeast University, Nanjing, Jiangsu, P.R. China
| |
Collapse
|
24
|
Amable G, Martínez-León E, Picco ME, Di Siervi N, Davio C, Rozengurt E, Rey O. Metformin inhibits β-catenin phosphorylation on Ser-552 through an AMPK/PI3K/Akt pathway in colorectal cancer cells. Int J Biochem Cell Biol 2019; 112:88-94. [PMID: 31082618 DOI: 10.1016/j.biocel.2019.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023]
Abstract
Several epidemiologic studies have revealed strong inverse associations between metformin use and risk of colorectal cancer development. Nevertheless, the underlying mechanisms are still uncertain. The Wnt/β-catenin pathway, which plays a central role in intestinal homeostasis and sporadic colorectal cancer development, is regulated by phosphorylation cascades that are dependent and independent of Wnt. Here we report that a non-canonical Ser552 phosphorylation in β-catenin, which promotes its nuclear accumulation and transcriptional activity, is blocked by metformin via AMPK-mediated PI3K/Akt signaling inhibition.
Collapse
Affiliation(s)
- Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Nicolas Di Siervi
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina
| | - Carlos Davio
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina; Departamento de Farmacología, Caba, 1113, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, CA, 90095-1786, USA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina.
| |
Collapse
|
25
|
Murphy N, Jenab M, Gunter MJ. Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol 2018; 15:659-670. [PMID: 29970888 DOI: 10.1038/s41575-018-0038-1] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Excess adiposity is a risk factor for several cancers of the gastrointestinal system, specifically oesophageal adenocarcinoma and colorectal, small intestine, pancreatic, liver, gallbladder and stomach cancers. With the increasing prevalence of obesity in nearly all regions of the world, this relationship could represent a growing source of cancers of the digestive system. Experimental and molecular epidemiological studies indicate important roles for alterations in insulin signalling, adipose tissue-derived inflammation and sex hormone pathways in mediating the association between adiposity and gastrointestinal cancer. The intestinal microbiome, gut hormones and non alcoholic fatty liver disease (NAFLD) also have possible roles. However, important gaps remain in our knowledge. For instance, our understanding of how adiposity throughout the life course is related to the risk of gastrointestinal cancer development and of how obesity influences gastrointestinal cancer prognosis and survival is limited. Nonetheless, the increasing use of state-of-the-art analytical methods (such as omics technologies, Mendelian randomization and MRI) in large-scale epidemiological studies offers exciting opportunities to advance our understanding of the complex relationship between adiposity and gastrointestinal cancers. Here, we examine the epidemiology of associations between obesity and gastrointestinal cancer, explore potential mechanisms underlying these relationships and highlight important unanswered research questions.
Collapse
Affiliation(s)
- Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Mazda Jenab
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
26
|
Han R, Ma J, Li H. Mechanistic and therapeutic advances in non-alcoholic fatty liver disease by targeting the gut microbiota. Front Med 2018; 12:645-657. [PMID: 30178233 DOI: 10.1007/s11684-018-0645-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common metabolic diseases currently in the context of obesity worldwide, which contains a spectrum of chronic liver diseases, including hepatic steatosis, non-alcoholic steatohepatitis and hepatic carcinoma. In addition to the classical "Two-hit" theory, NAFLD has been recognized as a typical gut microbiota-related disease because of the intricate role of gut microbiota in maintaining human health and disease formation. Moreover, gut microbiota is even regarded as a "metabolic organ" that play complementary roles to that of liver in many aspects. The mechanisms underlying gut microbiota-mediated development of NAFLD include modulation of host energy metabolism, insulin sensitivity, and bile acid and choline metabolism. As a result, gut microbiota have been emerging as a novel therapeutic target for NAFLD by manipulating it in various ways, including probiotics, prebiotics, synbiotics, antibiotics, fecal microbiota transplantation, and herbal components. In this review, we summarized the most recent advances in gut microbiota-mediated mechanisms, as well as gut microbiota-targeted therapies on NAFLD.
Collapse
Affiliation(s)
- Ruiting Han
- Functional Metabolomic and Gut Microbiome Laboratory, Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Junli Ma
- Functional Metabolomic and Gut Microbiome Laboratory, Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Houkai Li
- Functional Metabolomic and Gut Microbiome Laboratory, Institute of Interdisciplinary Integrative Biomedical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
27
|
Carr BI, Giannelli G, Guerra V, Giannini EG, Farinati F, Rapaccini GL, Marco MD, Zoli M, Caturelli E, Masotto A, Virdone R, Sacco R, Trevisani F. Plasma cholesterol and lipoprotein levels in relation to tumor aggressiveness and survival in HCC patients. Int J Biol Markers 2018; 33:423-431. [PMID: 29874983 DOI: 10.1177/1724600818776838] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS: Hepatocellular carcinoma is associated with several chronic liver diseases, especially chronic hepatitis B virus, hepatitis C virus, and alcoholism. It is increasingly appreciated that obesity/metabolic syndrome is also associated with chronic liver disease and subsequent hepatocellular carcinoma. METHODS: We retrospectively investigated the serum lipid profiles in a large hepatocellular carcinoma cohort, associated predominantly with the hepatitis B virus, hepatitis C virus, alcohol or nonalcoholic steatohepatitis. The cohort was examined both as a whole, as well as stratified by etiology. RESULTS: We found significant associations between parameters of hepatocellular carcinoma biology such as maximum tumor diameter, portal vein thrombosis, tumor multifocality or alpha-fetoprotein levels and individual lipid components, including total cholesterol, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, triglycerides and body mass index. In a final multiple linear regression model considering all lipid variables together, only high-density lipoprotein cholesterol was significantly associated with the tumor Tumor Aggressiveness Index. High-density lipoprotein cholesterol was found to have a statistically higher hazard ratio for death than low high-density lipoprotein cholesterol levels (Cox). On examination by etiological group, alpha-fetoprotein levels were significantly higher in patients with hepatitis C virus compared to those with alcohol or nonalcoholic steatohepatitis, but maximum tumor diameter, tumor multifocality and portal vein thrombosis were similar across etiological groups. Nonalcoholic steatohepatitis patients had significantly less cirrhosis than other groups and hepatitis B virus patients had significantly higher cholesterol and low-density lipoprotein cholesterol levels than hepatitis C virus patients. CONCLUSIONS: This is the first report, to our knowledge, of a relationship between serum lipid parameters and indices of hepatocellular carcinoma growth, invasion and aggressiveness, as well as with survival.
Collapse
Affiliation(s)
- Brian I Carr
- 1 Izmir Biomedicine and Genome Institute (iBG), Dokuz Eylul University, Izmir, Turkey
| | - Gianluigi Giannelli
- 2 National Institute of Digestive Diseases, IRCCS Saverio de Bellis, Castellana Grotte, Italy
| | - Vito Guerra
- 2 National Institute of Digestive Diseases, IRCCS Saverio de Bellis, Castellana Grotte, Italy
| | - Edoardo G Giannini
- 3 Department of Internal Medicine, Gastroenterology Unit, University of Genoa, Italy
| | - Fabio Farinati
- 4 Department of Surgical Science and Gastroenterology, Gastroenterology Unit, University of Padua, Italy
| | | | - Maria Di Marco
- 6 Division of Medicine, Azienda Ospedaliera Bolognini, Seriate, Italy
| | - Marco Zoli
- 7 Department of Medical and Surgical Science, Internal Medicine Unit, Alma Mater Studiorum, University of Bologna, Italy
| | | | - Alberto Masotto
- 9 Gastroenterology Unit, Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - Roberto Virdone
- 10 Division of Internal Medicine 2, Ospedali Riuniti Villa Sofia Cervello, Palermo, Italy
| | - Rodolfo Sacco
- 11 Unita Operativa Medicina interna 2, Ospedali Riuniti, Cervello, Palermo, Italy and Metabolic Diseases, Pisa University Hospital, Pisa, Italy
| | - Franco Trevisani
- 12 Department of Medical Surgical Sciences, Medical Semiotics Unit, Alma Mater Studiorum, University of Bologna, Italy
| |
Collapse
|
28
|
Tian Y, Lyu H, He Y, Xia Y, Li J, Shen F. Comparison of Hepatectomy for Patients with Metabolic Syndrome-Related HCC and HBV-Related HCC. J Gastrointest Surg 2018; 22:615-623. [PMID: 29139083 DOI: 10.1007/s11605-017-3629-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 10/31/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Metabolic syndrome (MetS) is a group of clinicopathological manifestations. The outcome of liver surgery in metabolic syndrome-related hepatocellular carcinoma (MetS-HCC) still needs to be evaluated. We aim to clarify the outcomes following liver resection in patients with MetS-HCC compared those with hepatitis B virus-related HCC (HBV-HCC). METHODS All the consecutive patients undergoing hepatectomy for HCC between January 2009 and December 2012 were retrospectively considered. Patients were divided into three groups: MetS-HCC, MetS-HBV-HCC, and HBV-HCC. Data on clinical characteristics, postoperative complications, and long-term outcome were collected and analyzed. RESULTS A total of 1352 patients were included in this study. In MetS-HCC group, the severe morbidity rate was 33.33%, which was higher than that of HBV-HCC group (15.68%). In subgroup analysis, patients with MetS-HCC in American Joint Committee on Cancer (AJCC) stage I had superior DFS and OS when compared with those of the other two groups. CONCLUSIONS We should pay more attention to patients with MetS-HCC perioperatively due to the high rate of surgical complications. Nevertheless, curative treatment should be provided to patients with MetS.
Collapse
Affiliation(s)
- Yunhong Tian
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, 200438, China.,Graduate School of Soochow University, Suzhou, China.,Department of General Surgery, Nanchong Central Hospital, Nanchong, China
| | - Huan Lyu
- Department of General Surgery, Nanchong Central Hospital, Nanchong, China
| | - Yunhong He
- Department of General Surgery, Nanchong Central Hospital, Nanchong, China
| | - Yong Xia
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, 200438, China
| | - Jun Li
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, 200438, China
| | - Feng Shen
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Second Military Medical University, Shanghai, 200438, China.
| |
Collapse
|
29
|
Gingold JA, Zhu D, Lee DF, Kaseb A, Chen J. Genomic Profiling and Metabolic Homeostasis in Primary Liver Cancers. Trends Mol Med 2018. [PMID: 29530485 DOI: 10.1016/j.molmed.2018.02.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (iCCA), the two most common primary liver cancers, represent the second most common cancer-related cause of death worldwide, with most cases being diagnosed at an advanced stage. Recent genome-wide studies have helped to elucidate the molecular pathogenesis and genetic heterogeneity of liver cancers. This review of the genetic landscape of HCC and iCCA discusses the most recent findings from genomic profiling and the current understanding of the pathways involved in the initiation and progression of liver cancer. We highlight recent insights gained from metabolic profiling of HCC and iCCA. This knowledge will be key to developing clinically useful diagnostic/prognostic profiles, building targeted molecular and immunologic therapies, and ultimately curing these complex and heterogeneous diseases.
Collapse
Affiliation(s)
- Julian A Gingold
- Women's Health Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Dandan Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center, Houston, TX 77030, USA
| | - Ahmed Kaseb
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jian Chen
- Department of Gastroenterology, Hepatology, and Nutrition, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
30
|
Aravani A, Downing A, Thomas JD, Lagergren J, Morris EJA, Hull MA. Obesity surgery and risk of colorectal and other obesity-related cancers: An English population-based cohort study. Cancer Epidemiol 2018; 53:99-104. [PMID: 29414638 PMCID: PMC5865073 DOI: 10.1016/j.canep.2018.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/18/2017] [Accepted: 01/02/2018] [Indexed: 12/13/2022]
Abstract
The association between obesity surgery (OS) and cancer risk remains unclear. Colorectal cancer (CRC) risk is increased in obese individuals. No evidence of increased CRC risk after OS in this English population study. Reduced breast cancer risk was apparent after OS. Renal and endometrial cancer risk is elevated in obese patients.
Background The association between obesity surgery (OS) and cancer risk remains unclear. We investigated this association across the English National Health Service. A population-based Swedish study has previously suggested that OS may increase the risk of developing colorectal cancer (CRC). Methods A retrospective observational study of individuals who underwent OS (surgery cohort) or diagnosed with obesity, but had no OS (no-surgery cohort) (1997–2013) were identified using Hospital Episode Statistics. Subsequent diagnosis of CRC, breast, endometrial, kidney and lung cancer, as well as time ‘at risk’, were determined by linkage to National Cancer Registration & Analysis Service and Office of National Statistics data, respectively. Standardised incidence ratios (SIR) in relation to OS were calculated. Results 1 002 607 obese patients were identified, of whom 3.9% (n = 39 747) underwent OS. In the no-surgery obese population, 3 237 developed CRC (SIR 1.12 [95% CI 1.08–1.16]). In those who underwent OS, 43 developed CRC (SIR 1.26 [95% CI 0.92–1.71]). The OS cohort demonstrated decreased breast cancer risk (SIR 0.76 [95% CI 0.62–0.92]), unlike the no surgery cohort (SIR 1.08 [95% CI 1.04–1.11]). Increased risk of endometrial and kidney cancer was observed in surgery and no-surgery cohorts. Conclusions CRC risk is increased in individuals diagnosed as obese. Prior obesity surgery was not associated with an increased CRC risk. However, the OS population was small, with limited follow-up. Risk of breast cancer after OS is reduced compared with the obese no-surgery population, while the risk of endometrial and kidney cancers remained elevated after OS.
Collapse
Affiliation(s)
- Ariadni Aravani
- Leeds Institute of Cancer & Pathology, University of Leeds, Worsley Building, Leeds, LS2 9NL, United Kingdom.
| | - Amy Downing
- Leeds Institute of Cancer & Pathology, University of Leeds, Worsley Building, Leeds, LS2 9NL, United Kingdom.
| | - James D Thomas
- Public Health England, Blenheim House, Duncombe St., Leeds, West Yorkshire, LS1 4PL, United Kingdom.
| | - Jesper Lagergren
- Upper Gastrointestinal Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Karolinska University Hospital, 17176 Stockholm, Sweden; Division of Cancer Studies, King's College London, WC2R 2LS, United Kingdom.
| | - Eva J A Morris
- Leeds Institute of Cancer & Pathology, University of Leeds, Worsley Building, Leeds, LS2 9NL, United Kingdom.
| | - Mark A Hull
- Leeds Institute of Biomedical & Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, United Kingdom.
| |
Collapse
|
31
|
Ortega LS, Bradbury KE, Cross AJ, Morris JS, Gunter MJ, Murphy N. A Prospective Investigation of Body Size, Body Fat Composition and Colorectal Cancer Risk in the UK Biobank. Sci Rep 2017; 7:17807. [PMID: 29259258 PMCID: PMC5736687 DOI: 10.1038/s41598-017-17997-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/04/2017] [Indexed: 12/26/2022] Open
Abstract
Obesity has been consistently associated with a greater colorectal cancer risk, but this relationship is weaker among women. In the UK Biobank, we investigated the associations between body size (body mass index [BMI], height, waist circumference, and waist-to-hip ratio) and body fat composition (total body fat percentage and trunk fat percentage) measurements with colorectal cancer risk among 472,526 men and women followed for 5.6 years on average. Multivariable hazard ratios (HRs) and 95% confidence intervals (95%CI) for developing colorectal cancer (2,636 incident cases) were estimated using Cox proportional hazards models. Among men, when the highest and lowest fifths were compared, BMI (HR = 1.35, 95%CI: 1.13-1.61; Ptrend < 0.0001), waist circumference (HR = 1.66, 95%CI: 1.39-1.99; Ptrend < 0.0001), waist-to-hip ratio (HR = 1.58, 95%CI: 1.31-1.91; Ptrend < 0.0001), total body fat percentage (HR = 1.27, 95%CI: 1.06-1.53; Ptrend = 0.002), and trunk fat percentage (HR = 1.31, 95%CI: 1.09-1.58; Ptrend = 0.002) were associated with greater colorectal cancer risk. For women, only waist-to-hip ratio (HR for highest versus lowest fifth = 1.33, 95%CI: 1.08-1.65; Ptrend = 0.005) was positively associated with colorectal cancer risk. Greater body size (overall and abdominal adiposity) was positively associated with colorectal cancer development in men. For women, abdominal adiposity, rather than overall body size, was associated with a greater colorectal cancer risk.
Collapse
Affiliation(s)
- Luisa Saldana Ortega
- Primary Care Clinical Trials Unit, Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Kathryn E Bradbury
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Amanda J Cross
- School of Public Health, Imperial College London, London, UK
| | | | - Marc J Gunter
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France
| | - Neil Murphy
- Section of Nutrition and Metabolism, International Agency for Research on Cancer, Lyon, France.
| |
Collapse
|
32
|
Eibl G, Rozengurt E. KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol 2017; 54:50-62. [PMID: 29079305 DOI: 10.1016/j.semcancer.2017.10.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 10/22/2017] [Indexed: 02/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) continues to be a lethal disease with no efficacious treatment modalities. The incidence of PDAC is expected to increase, at least partially because of the obesity epidemic. Increased efforts to prevent or intercept this disease are clearly needed. Mutations in KRAS are initiating events in pancreatic carcinogenesis supported by genetically engineered mouse models of the disease. However, oncogenic KRAS is not entirely sufficient for the development of fully invasive PDAC. Additional genetic mutations and/or environmental, nutritional, and metabolic stressors, e.g. inflammation and obesity, are required for efficient PDAC formation with activation of KRAS downstream effectors. Multiple factors "upstream" of KRAS associated with obesity, including insulin resistance, inflammation, changes in gut microbiota and GI peptides, can enhance/modulate downstream signals. Multiple signaling networks and feedback loops "downstream" of KRAS have been described that respond to obesogenic diets. We propose that KRAS mutations potentiate a signaling network that is promoted by environmental factors. Specifically, we envisage that KRAS mutations increase the intensity and duration of the growth-promoting signaling network. As the transcriptional activator YAP plays a critical role in the network, we conclude that the rationale for targeting the network (at different points), e.g. with FDA approved drugs such as statins and metformin, is therefore compelling.
Collapse
Affiliation(s)
- Guido Eibl
- Departments of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States.
| | - Enrique Rozengurt
- Departments of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States; CURE: Digestive Diseases Research Center, University of California at Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
33
|
Anhê FF, Varin TV, Schertzer JD, Marette A. The Gut Microbiota as a Mediator of Metabolic Benefits after Bariatric Surgery. Can J Diabetes 2017; 41:439-447. [PMID: 28552651 DOI: 10.1016/j.jcjd.2017.02.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 01/25/2017] [Accepted: 02/07/2017] [Indexed: 02/07/2023]
|
34
|
Huang L, Wei ZJ, Li TJ, Jiang YM, Xu AM. A prospective appraisal of preoperative body mass index in D2-resected patients with non-metastatic gastric carcinoma and Siewert type II/III adenocarcinoma of esophagogastric junction: results from a large-scale cohort. Oncotarget 2017; 8:68165-68179. [PMID: 28978106 PMCID: PMC5620246 DOI: 10.18632/oncotarget.19251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 06/16/2017] [Indexed: 02/06/2023] Open
Abstract
Objective To prospectively investigate associations of presurgical body mass index (BMI) with clinicopathological factors and its prognostic significance in radically D2-resected patients with non-metastasized gastric cancer (GC) and Siewert type II/III adenocarcinoma of esophagogastric junction (AEG). Methods A large prospective cohort consisting of radically-resected GC and AEG patients was analyzed. Follow-up was successful in 671 out of 700 patients, who were categorized into underweight (BMI<18.5), normal-weight (BMI=18.5-22.9), overweight (BMI=23-24.9), and obese (BMI≥25) groups according to Asian standards. BMI-associated factors were explored using multivariable logistic regression with adjustment. Cancer-specific survival analyses were conducted applying both univariable and multivariable Cox regression methods. Results Pre-operation, higher hemoglobin levels and smaller anemia proportions were observed in larger BMI groups. Higher BMI tended to be associated with higher neutrophil-lymphocyte ratios (NLRs). Patients with higher BMI had smaller tumors and more often stage I tumors, but longer surgical time and postsurgical stay. In multivariable analyses, higher hemoglobin levels, upper tumor location, poorer differentiation, and higher NLR were significantly associated with higher BMI. Overall, survival analyses revealed no significant role of BMI. However, in further stratifications after adjustment, compared to patients with normal BMI, obese patients had better survival in women, but worse in those with AEG; underweight was associated with reduced mortality risk in tumors differentiated well to moderately; overweight patients had increased death hazard when having thrombocytopenia. Conclusion Overall, preoperative BMI had limited prognostic significance in operated GC patients. However, under specific conditions (e.g., female, AEG, good differentiation, and thrombocytopenia), BMI might indicate postoperative survival.
Collapse
Affiliation(s)
- Lei Huang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhi-Jian Wei
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tuan-Jie Li
- Department of General Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Yu-Ming Jiang
- Department of General Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - A-Man Xu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of General Surgery, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
35
|
|
36
|
Catalán V, Gómez-Ambrosi J, Rodríguez A, Ramírez B, Izaguirre M, Hernández-Lizoain JL, Baixauli J, Martí P, Valentí V, Moncada R, Silva C, Salvador J, Frühbeck G. Increased Obesity-Associated Circulating Levels of the Extracellular Matrix Proteins Osteopontin, Chitinase-3 Like-1 and Tenascin C Are Associated with Colon Cancer. PLoS One 2016; 11:e0162189. [PMID: 27612200 PMCID: PMC5017763 DOI: 10.1371/journal.pone.0162189] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 08/18/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Excess adipose tissue represents a major risk factor for the development of colon cancer with inflammation and extracellular matrix (ECM) remodeling being proposed as plausible mechanisms. The aim of this study was to investigate whether obesity can influence circulating levels of inflammation-related extracellular matrix proteins in patients with colon cancer (CC), promoting a microenvironment favorable for tumor growth. METHODS Serum samples obtained from 79 subjects [26 lean (LN) and 53 obese (OB)] were used in the study. Enrolled subjects were further subclassified according to the established diagnostic protocol for CC (44 without CC and 35 with CC). Anthropometric measurements as well as circulating metabolites and hormones were determined. Circulating concentrations of the ECM proteins osteopontin (OPN), chitinase-3-like protein 1 (YKL-40), tenascin C (TNC) and lipocalin-2 (LCN-2) were determined by ELISA. RESULTS Significant differences in circulating OPN, YKL-40 and TNC concentrations between the experimental groups were observed, being significantly increased due to obesity (P<0.01) and colon cancer (P<0.05). LCN-2 levels were affected by obesity (P<0.05), but no differences were detected regarding the presence or not of CC. A positive association (P<0.05) with different inflammatory markers was also detected. CONCLUSIONS To our knowledge, we herein show for the first time that obese patients with CC exhibit increased circulating levels of OPN, YKL-40 and TNC providing further evidence for the influence of obesity on CC development via ECM proteins, representing promising diagnostic biomarkers or target molecules for therapeutics.
Collapse
Affiliation(s)
- Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
| | - Maitane Izaguirre
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | | | - Jorge Baixauli
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Pablo Martí
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Víctor Valentí
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Javier Salvador
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
- Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA) Pamplona, Spain
- Department of Endocrinology & Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
- * E-mail:
| |
Collapse
|
37
|
Jarvis D, Mitchell JS, Law PJ, Palin K, Tuupanen S, Gylfe A, Hänninen UA, Cajuso T, Tanskanen T, Kondelin J, Kaasinen E, Sarin AP, Kaprio J, Eriksson JG, Rissanen H, Knekt P, Pukkala E, Jousilahti P, Salomaa V, Ripatti S, Palotie A, Järvinen H, Renkonen-Sinisalo L, Lepistö A, Böhm J, Meklin JP, Al-Tassan NA, Palles C, Martin L, Barclay E, Farrington SM, Timofeeva MN, Meyer BF, Wakil SM, Campbell H, Smith CG, Idziaszczyk S, Maughan TS, Kaplan R, Kerr R, Kerr D, Buchanan DD, Win AK, Hopper JL, Jenkins MA, Lindor NM, Newcomb PA, Gallinger S, Conti D, Schumacher F, Casey G, Taipale J, Aaltonen LA, Cheadle JP, Dunlop MG, Tomlinson IP, Houlston RS. Mendelian randomisation analysis strongly implicates adiposity with risk of developing colorectal cancer. Br J Cancer 2016; 115:266-72. [PMID: 27336604 PMCID: PMC4947703 DOI: 10.1038/bjc.2016.188] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 05/09/2016] [Accepted: 05/14/2016] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Observational studies have associated adiposity with an increased risk of colorectal cancer (CRC). However, such studies do not establish a causal relationship. To minimise bias from confounding we performed a Mendelian randomisation (MR) analysis to examine the relationship between adiposity and CRC. METHODS We used SNPs associated with adult body mass index (BMI), waist-hip ratio (WHR), childhood obesity and birth weight as instrumental variables in a MR analysis of 9254 CRC cases and 18 386 controls. RESULTS In the MR analysis, the odds ratios (ORs) of CRC risk per unit increase in BMI, WHR and childhood obesity were 1.23 (95% CI: 1.02-1.49, P=0.033), 1.59 (95% CI: 1.08-2.34, P=0.019) and 1.07 (95% CI: 1.03-1.13, P=0.018), respectively. There was no evidence for association between birth weight and CRC (OR=1.22, 95% CI: 0.89-1.67, P=0.22). Combining these data with a concurrent MR-based analysis for BMI and WHR with CRC risk (totalling to 18 190 cases, 27 617 controls) provided increased support, ORs for BMI and WHR were 1.26 (95% CI: 1.10-1.44, P=7.7 × 10(-4)) and 1.40 (95% CI: 1.14-1.72, P=1.2 × 10(-3)), respectively. CONCLUSIONS These data provide further evidence for a strong causal relationship between adiposity and the risk of developing CRC highlighting the urgent need for prevention and treatment of adiposity.
Collapse
Affiliation(s)
- David Jarvis
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Jonathan S Mitchell
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Philip J Law
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SW7 3RP, UK
| | - Kimmo Palin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Sari Tuupanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Alexandra Gylfe
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Ulrika A Hänninen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Tatiana Cajuso
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Tomas Tanskanen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Johanna Kondelin
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Eevi Kaasinen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Antti-Pekka Sarin
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Johan G Eriksson
- National Institute for Health and Welfare, Helsinki 00271, Finland
- Folkhälsan Research Centre, Helsinki 00250, Finland
- Unit of General Practice and Primary Health Care, University of Helsinki, Helsinki University Hospital, Helsinki 00014, Finland
| | - Harri Rissanen
- National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Paul Knekt
- National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Eero Pukkala
- Finnish Cancer Registry, Institute for Statistical and Epidemiological Cancer Research, Helsinki 00130, Finland
- School of Health Sciences, University of Tampere, Tampere 33014, Finland
| | - Pekka Jousilahti
- National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Veikko Salomaa
- National Institute for Health and Welfare, Helsinki 00271, Finland
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki 00014, Finland
- Department of Medicine, Analytic and Translational Genetics Unit, Massachusetts General Hospital, Boston, MA 02114, USA
- Program in Medical and Population Genetics, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Heikki Järvinen
- Department of Surgery, Helsinki University Central Hospital, Hospital District of Helsinki and Uusimaa, Helsinki 00029, Finland
| | - Laura Renkonen-Sinisalo
- Department of Surgery, Abdominal Center, Helsinki University Hospital Helsinki 00029, Finland
| | - Anna Lepistö
- Department of Surgery, Abdominal Center, Helsinki University Hospital Helsinki 00029, Finland
| | - Jan Böhm
- Department of Pathology, Central Finland Central Hospital, Jyväskylä 40620, Finland
| | - Jukka-Pekka Meklin
- Department of Surgery, Jyväskylä Central Hospital, University of Eastern Finland, Jyväskylä 40620, Finland
| | - Nada A Al-Tassan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| | - Claire Palles
- Wellcome Trust Centre for Human Genetics, NIHR Comprehensive Biomedical Research Centre, Oxford OX3 7BN, UK
| | - Lynn Martin
- Wellcome Trust Centre for Human Genetics, NIHR Comprehensive Biomedical Research Centre, Oxford OX3 7BN, UK
| | - Ella Barclay
- Wellcome Trust Centre for Human Genetics, NIHR Comprehensive Biomedical Research Centre, Oxford OX3 7BN, UK
| | - Susan M Farrington
- Colon Cancer Genetics Group, MRC Human Genetics Unit, The University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
- The Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Edinburgh EH25 9RG, UK
| | - Maria N Timofeeva
- Colon Cancer Genetics Group, MRC Human Genetics Unit, The University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Brian F Meyer
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| | - Salma M Wakil
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh 12713, Saudi Arabia
| | - Harry Campbell
- Centre for Population Health Sciences, University of Edinburgh, Edinburgh EH8 9AG, UK
| | - Christopher G Smith
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Shelley Idziaszczyk
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Timothy S Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Richard Kaplan
- MRC Clinical Trials Unit, Aviation House, London WC2B 6NH, UK
| | - Rachel Kerr
- Department of Oncology, Oxford Cancer Centre, University of Oxford, Churchill Hospital, Oxford OX3 7LE, UK
| | - David Kerr
- Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Daniel D Buchanan
- Department of Pathology, Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, The University of Melbourne, Melbourne, VIC 3010, Australia
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Aung K Win
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Mark A Jenkins
- Centre for Epidemiology and Biostatistics, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Noralane M Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ 85259, USA
| | - Polly A Newcomb
- Cancer Prevention Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Steve Gallinger
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - David Conti
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Fred Schumacher
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Graham Casey
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Jussi Taipale
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
- Department of Biosciences and Nutrition, SciLife Center, Karolinska Institutet, Solna SE 141 83, Sweden
| | - Lauri A Aaltonen
- Genome-Scale Biology Research Program, Research Programs Unit, University of Helsinki 00014, Helsinki, Finland
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki 00014, Finland
| | - Jeremy P Cheadle
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Malcolm G Dunlop
- Colon Cancer Genetics Group, MRC Human Genetics Unit, The University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Ian P Tomlinson
- Wellcome Trust Centre for Human Genetics, NIHR Comprehensive Biomedical Research Centre, Oxford OX3 7BN, UK
| | - Richard S Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London SW7 3RP, UK
| |
Collapse
|
38
|
Pathak S, Pandanaboyana S, Daniels I, Smart N, Prasad KR. Obesity and colorectal liver metastases: Mechanisms and management. Surg Oncol 2016; 25:246-51. [PMID: 27566030 DOI: 10.1016/j.suronc.2016.05.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 05/19/2016] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Colorectal cancer (CRC) is the third commonest malignancy after lung and breast cancer. The most common cause of mortality from CRC is from distant metastases. Obesity is a known risk factor for primary CRC development. However, its role in metastatic disease progression is not fully understood. The article aims to provide an overview of the role of obesity in colorectal liver metastases (CRLM). Furthermore, possible strategies to minimise this effect are discussed. An electronic search of MedLine, EMBASE, CINAHL and google scholar was performed. Relevant articles were included in the article. Obesity causes localised inflammation within the liver microenvironment which may predispose to metastases development. Furthermore, obesity causes systemic inflammation leading to release of protumourigenic growth factors. Several studies demonstrated the effects of lifestyle modification, medications, bariatric surgery and omega-3 fatty acids on steatosis within the context of liver surgery. It is currently unclear whether obesity directly leads to metastatic disease via chronic systemic inflammation or whether obesity induced steatosis provides a fertile microenvironment for metastases deposition. With a global increase in obesity useful strategies to minimise the effects of obesity on the liver include life-style modification, pre-operative dietary regimes and omega-3 fatty acids intake. Pre-operative optimisation of the patient is a key concept. Further randomised control trials are needed to guide management strategies.
Collapse
Affiliation(s)
- Samir Pathak
- St James's University Hospital, Beckett Street, Leeds, West Yorkshire, LS9 7TF, United Kingdom; Exeter Surgical Health Services Research Unit (HeSRU), Royal Devon & Exeter Hospital, Barrack Road, Exeter, Devon, EX2 5DW, United Kingdom.
| | - Sanjay Pandanaboyana
- St James's University Hospital, Beckett Street, Leeds, West Yorkshire, LS9 7TF, United Kingdom
| | - Ian Daniels
- Exeter Surgical Health Services Research Unit (HeSRU), Royal Devon & Exeter Hospital, Barrack Road, Exeter, Devon, EX2 5DW, United Kingdom
| | - Neil Smart
- Exeter Surgical Health Services Research Unit (HeSRU), Royal Devon & Exeter Hospital, Barrack Road, Exeter, Devon, EX2 5DW, United Kingdom
| | - K R Prasad
- St James's University Hospital, Beckett Street, Leeds, West Yorkshire, LS9 7TF, United Kingdom
| |
Collapse
|
39
|
Babic A, Shah SM, Song M, Wu K, Meyerhardt JA, Ogino S, Yuan C, Giovannucci EL, Chan AT, Stampfer MJ, Fuchs CS, Ng K. Soluble tumour necrosis factor receptor type II and survival in colorectal cancer. Br J Cancer 2016; 114:995-1002. [PMID: 27031855 PMCID: PMC4984918 DOI: 10.1038/bjc.2016.85] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/03/2016] [Accepted: 03/08/2016] [Indexed: 12/21/2022] Open
Abstract
Background: Chronic inflammation may play a role in colorectal cancer (CRC) pathogenesis. The relationship between soluble tumour necrosis factor receptor type II (sTNF-RII) and survival among CRC patients is not well defined. Methods: We prospectively evaluated the association between pre-diagnosis plasma levels of sTNF-RII and mortality in 544 CRC patients from the Nurses' Health Study and Health Professionals Follow-Up Study diagnosed from 1990 to 2010. Primary and secondary end points were overall and CRC-specific mortality, respectively. Cox proportional hazards models were used to calculate multivariate hazard ratios for mortality. Results: Higher sTNF-RII levels were significantly associated with increased overall mortality (multivariate HR=1.48, 95% CI 1.02–2.16, P-trend=0.006), but not with CRC-specific mortality (HR=1.23, 95% CI 0.72–2.08, P-trend=0.34). In subgroup analyses, among regular aspirin users, those with higher sTNF-RII levels had an adjusted HR of 0.52 (95% CI 0.20–1.33) for overall mortality compared with those with lower sTNF-RII levels, whereas among nonregular aspirin users the adjusted HR was 2.26 (95% CI 1.23–4.01, P for interaction=0.53). Conclusions: Among CRC patients, higher sTNF-RII levels are associated with a significant increase in overall mortality, but not CRC-specific mortality. The role of inflammation and anti-inflammatory medications in survival of CRC patients warrants further exploration.
Collapse
Affiliation(s)
- Ana Babic
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Sonali M Shah
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA.,Division of Hematology and Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Mingyang Song
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.,Department of Epidemiology, Harvard T. H. School of Public Health, Boston, MA 02215, USA
| | - Kana Wu
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Shuji Ogino
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA.,Department of Epidemiology, Harvard T. H. School of Public Health, Boston, MA 02215, USA.,Division of MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.,Department of Epidemiology, Harvard T. H. School of Public Health, Boston, MA 02215, USA.,Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Andrew T Chan
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA.,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Meir J Stampfer
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA 02115, USA.,Department of Epidemiology, Harvard T. H. School of Public Health, Boston, MA 02215, USA.,Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Charles S Fuchs
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| | - Kimmie Ng
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA 02215, USA
| |
Collapse
|
40
|
Affiliation(s)
- Kamran B Lankarani
- Health Policy Research Center, Shiraz University of Medical Sciences, Shiraz, Islamic Republic of Iran
| |
Collapse
|
41
|
The tumor microenvironment in esophageal cancer. Oncogene 2016; 35:5337-5349. [PMID: 26923327 PMCID: PMC5003768 DOI: 10.1038/onc.2016.34] [Citation(s) in RCA: 231] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 02/08/2023]
Abstract
Esophageal cancer is a deadly disease, ranking sixth among all cancers in mortality. Despite incremental advances in diagnostics and therapeutics, esophageal cancer still carries a poor prognosis, and thus there remains a need to elucidate the molecular mechanisms underlying this disease. There is accumulating evidence that a comprehensive understanding of the molecular composition of esophageal cancer requires attention to not only tumor cells but also the tumor microenvironment, which contains diverse cell populations, signaling factors, and structural molecules that interact with tumor cells and support all stages of tumorigenesis. In esophageal cancer, environmental exposures can trigger chronic inflammation, which leads to constitutive activation of pro-inflammatory signaling pathways that promote survival and proliferation. Anti-tumor immunity is attenuated by cell populations such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs), as well as immune checkpoints like programmed death-1 (PD-1). Other immune cells such as tumor-associated macrophages can have other pro-tumorigenic functions, including the induction of angiogenesis and tumor cell invasion. Cancer-associated fibroblasts secrete growth factors and alter the extracellular matrix (ECM) to create a tumor niche and enhance tumor cell migration and metastasis. Further study of how these TME components relate to the different stages of tumor progression in each esophageal cancer subtype will lead to development of novel and specific TME-targeting therapeutic strategies, which offer considerable potential especially in the setting of combination therapy.
Collapse
|
42
|
Maeda K, Saigo C, Kito Y, Sakuratani T, Yoshida K, Takeuchi T. Expression of TMEM207 in Colorectal Cancer: Relation between TMEM207 and Intelectin-1. J Cancer 2016; 7:207-13. [PMID: 26819645 PMCID: PMC4716854 DOI: 10.7150/jca.13732] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 11/01/2015] [Indexed: 02/06/2023] Open
Abstract
Recent research advances highlighted an intestinal goblet cell-produced lectin, intelectin-1 (also known as omentin-1), as a tumor suppressor. One study indicated that downregulation of intelectin-1 may be related to the unfavorable prognosis among patients with colorectal carcinoma at an advanced stage. The present study was aimed at analyzing the expression of a hitherto uncharacterized transmembrane protein TMEM207 in colorectal carcinoma, and we found that the TMEM207 function is linked to intelectin-1 processing. With specific antibodies, TMEM207 immunoreactivity was detected in 38 of 216 colorectal cancer tissue samples. TMEM207 immunoreactivity correlated inversely with lymph node metastatic status (p < 0.01). TMEM207 expression significantly correlated with the mucinous phenotype of colorectal carcinoma. A coimmunoprecipitation assay revealed an interaction between intelectin-1 and TMEM207 in colorectal cancer cells. A proximal ligation assay indicated that intelectin-1 and TMEM207 were colocalized to the cytoplasm of the colorectal cancer cells. A small-interfering-RNA-mediated knockdown of TMEM207 increased polyubiquitination and proteasome degradation of intelectin-1 in cultured colorectal cancer cells and decreased intelectin-1 secretion. These findings indicate that a loss of TMEM207 expression leads to insufficient intelectin-1 production thus promoting colorectal carcinogenesis.
Collapse
Affiliation(s)
- Kenichi Maeda
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Chiemi Saigo
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yusuke Kito
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takuji Sakuratani
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazuhiro Yoshida
- 1. Department of Surgical Oncology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tamotsu Takeuchi
- 2. Department of Pathology and Translational Research, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
43
|
Park JM, Lee HJ, Yoo JH, Ko WJ, Cho JY, Hahm KB. Overview of gastrointestinal cancer prevention in Asia. Best Pract Res Clin Gastroenterol 2015; 29:855-67. [PMID: 26651248 DOI: 10.1016/j.bpg.2015.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/11/2015] [Accepted: 09/02/2015] [Indexed: 01/31/2023]
Abstract
"War on cancer" was declared through the National Cancer Act by President Richard Nixon in 1971, but cancer statistics from the American Cancer Society and other sources indicated the failure of this war, suggesting instead focus on the message that a "prevention strategy" might be much more effective than cancer treatment. While cancer statistics notoriously showed sharp increases in incidence as well as in mortality concurrent with economic growth in Asia, fortunately Asian countries benefit from plentiful resources of natural compounds, which can prevent cancer. Just like cancer chemotherapeutics targeted to kill cancer cells in Western countries, natural agents activating molecular mechanisms for cancer prevention, reversion of premalignant tumors, and even ablation of cancer stem cells, are very abundant in Asia. Currently, these natural agents are under very active investigations targeting the hallmarks of cancer prevention, including selective induction of apoptosis in cancer cells, suppression of growth factors or their signaling, suppression of cell proliferation and of cancer-promoting angiogenesis, induction of mesenchymal-epithelial transition, and disruption of the tumor microenvironment, developing promising cancer preventive agents. However, Asia is the most populous continent in the world and some Asian countries do not have the resources to implement cancer screening programs for early detection or treatment. In addition, despite the excellent cancer preventive screening strategies in some Asian countries, well-designed clinical trials for cancer prevention are somewhat delayed compared to Western countries. In this review article, several phytochemicals/phytoceuticals produced and studied in different Asian countries will be introduced, including Korean red ginseng (pride of Korea), curcumin (Indian spice for life), black or green tea (popular in Japan/Sri Lanka), genistein from tofu (famous Chinese food), diallylsulfide or S-allylcysteine (garlic, popularly consumed as a food ingredient in many Asian countries), capsaicin, 6-gingerol, flavopiridol, and silymarin (abundant in various Asian foods). Whereas in Western countries cancer chemotherapeutics involve strategies not only to block the growth of the primary tumor, but also to inhibit its progression to metastatic disease, the endless pursuit of effective agents for cancer prevention may be a unique and featured strategy in Asia. More active efforts for clinical application of these principles should be supported.
Collapse
Affiliation(s)
- Jong-Min Park
- CHA Cancer Prevention Research Center, CHA University, CHA Bio Complex, Seongnam, Republic of Korea.
| | - Ho-Jae Lee
- Laboratory of Cancer Prevention, Gachon University, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
| | - Jun Hwan Yoo
- CHA Cancer Prevention Research Center, CHA University, CHA Bio Complex, Seongnam, Republic of Korea.
| | - Weon Jin Ko
- Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Republic of Korea.
| | - Joo Young Cho
- Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Republic of Korea.
| | - Ki Baik Hahm
- CHA Cancer Prevention Research Center, CHA University, CHA Bio Complex, Seongnam, Republic of Korea; Digestive Disease Center, CHA University Bundang Medical Center, Seongnam, Republic of Korea.
| |
Collapse
|
44
|
Thrift AP, Gong J, Peters U, Chang-Claude J, Rudolph A, Slattery ML, Chan AT, Locke AE, Kahali B, Justice AE, Pers TH, Gallinger S, Hayes RB, Baron JA, Caan BJ, Ogino S, Berndt SI, Chanock SJ, Casey G, Haile RW, Du M, Harrison TA, Thornquist M, Duggan DJ, Le Marchand L, Lindor NM, Seminara D, Song M, Wu K, Thibodeau SN, Cotterchio M, Win AK, Jenkins MA, Hopper JL, Ulrich CM, Potter JD, Newcomb PA, Hoffmeister M, Brenner H, White E, Hsu L, Campbell PT. Mendelian Randomization Study of Body Mass Index and Colorectal Cancer Risk. Cancer Epidemiol Biomarkers Prev 2015; 24:1024-31. [PMID: 25976416 PMCID: PMC4490960 DOI: 10.1158/1055-9965.epi-14-1309] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 05/01/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND High body mass index (BMI) is consistently linked to increased risk of colorectal cancer for men, whereas the association is less clear for women. As risk estimates from observational studies may be biased and/or confounded, we conducted a Mendelian randomization study to estimate the causal association between BMI and colorectal cancer. METHODS We used data from 10,226 colorectal cancer cases and 10,286 controls of European ancestry. The Mendelian randomization analysis used a weighted genetic risk score, derived from 77 genome-wide association study-identified variants associated with higher BMI, as an instrumental variable (IV). We compared the IV odds ratio (IV-OR) with the OR obtained using a conventional covariate-adjusted analysis. RESULTS Individuals carrying greater numbers of BMI-increasing alleles had higher colorectal cancer risk [per weighted allele OR, 1.31; 95% confidence interval (CI), 1.10-1.57]. Our IV estimation results support the hypothesis that genetically influenced BMI is directly associated with risk for colorectal cancer (IV-OR per 5 kg/m(2), 1.50; 95% CI, 1.13-2.01). In the sex-specific IV analyses higher BMI was associated with higher risk of colorectal cancer among women (IV-OR per 5 kg/m(2), 1.82; 95% CI, 1.26-2.61). For men, genetically influenced BMI was not associated with colorectal cancer (IV-OR per 5 kg/m(2), 1.18; 95% CI, 0.73-1.92). CONCLUSIONS High BMI was associated with increased colorectal cancer risk for women. Whether abdominal obesity, rather than overall obesity, is a more important risk factor for men requires further investigation. IMPACT Overall, conventional epidemiologic and Mendelian randomization studies suggest a strong association between obesity and the risk of colorectal cancer.
Collapse
Affiliation(s)
- Aaron P Thrift
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas.
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Anja Rudolph
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts. Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Adam E Locke
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Bratati Kahali
- Department of Internal Medicine, Division of Gastroenterology, and Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Anne E Justice
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Tune H Pers
- Divisions of Endocrinology and Genetics and Center for Basic and Translational Obesity Research, Boston Children's Hospital, Boston, Massachusetts. Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, Massachusetts. Department of Genetics, Harvard Medical School, Boston, Massachusetts. Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Steven Gallinger
- Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, New York
| | - John A Baron
- Department of Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Bette J Caan
- Division of Research, Kaiser Permanente Northern California, Oakland, California
| | - Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, Maryland
| | - Graham Casey
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Robert W Haile
- Department of Medicine, Division of Oncology, Stanford University, Stanford, California
| | - Mengmeng Du
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington. Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark Thornquist
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - David J Duggan
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Noralane M Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, Arizona
| | - Daniela Seminara
- Division of Cancer Control and Population Sciences, National Cancer Institute, NIH, Bethesda, Maryland
| | - Mingyang Song
- Department of Epidemiology, Harvard School of Public Health, Boston, Massachusetts. Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Kana Wu
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts
| | - Stephen N Thibodeau
- Departments of Laboratory Medicine and Pathology and Laboratory Genetics, Mayo Clinic, Rochester, Minnesota
| | | | - Aung Ko Win
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Mark A Jenkins
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - John L Hopper
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | | | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington. Centre for Public Health Research, Massey University, Wellington, New Zealand
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany. German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Public Health, University of Washington, Seattle, Washington
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington
| | - Peter T Campbell
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| |
Collapse
|
45
|
Pawlik TM, Soubrane O. Metabolic syndrome-associated hepatocellular carcinoma: Questions still unanswered. J Hepatol 2015; 63:8-9. [PMID: 25862602 DOI: 10.1016/j.jhep.2015.03.035] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 03/31/2015] [Indexed: 01/07/2023]
Affiliation(s)
- Timothy M Pawlik
- Johns Hopkins Hospital, Division of Surgical Oncology, Blalock 688, Baltimore, MD, USA
| | - Olivier Soubrane
- Beaujon Hospital, HPB Surgery and Liver Transplant, 100 Bd du General Leclerc, Clichy 92100, France.
| |
Collapse
|
46
|
Clinical and Oncological Value of Preoperative BMI in Gastric Cancer Patients: A Single Center Experience. Gastroenterol Res Pract 2015; 2015:810134. [PMID: 25759721 PMCID: PMC4338394 DOI: 10.1155/2015/810134] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/20/2015] [Accepted: 01/20/2015] [Indexed: 12/12/2022] Open
Abstract
Introduction. The impact of preoperative BMI on surgical outcomes and long-term survival of gastric cancer patients was investigated in various reports with contrasting results. Materials & Methods. A total of 378 patients who underwent a surgical resection for primary gastric cancer between 1994 and 2011 were retrospectively studied. Patients were stratified according to BMI into a normal group (<25, group A), an overweight group (25-30, group B), and an obesity group (≥30, group C). These 3 groups were compared according to clinical-pathological characteristics, surgical treatment, and long-term survival. Results. No significant correlations between BMI and TNM (2010), UICC stage (2010), Lauren's histological type, surgical results, lymph node dissection, and postoperative morbidity and mortality were observed. Factors related to higher BMI were male gender (P < 0.05), diabetes (P < 0.001), and serum blood proteins (P < 0.01). A trend to fewer lymph nodes retrieved during gastrectomy with lymphadenectomy in overweight patients (B and C groups) was observed, although not statistically significant. There was no difference in overall survival or disease-specific survival between the three groups. Conclusion. According to our data, BMI should not be considered a significant predictor of postoperative complications or long-term result in gastric cancer patients.
Collapse
|
47
|
Calpe S, Compare D, Mari L, Nardone G, Parikh K. Immune signaling and regulation in esophageal cancer. Ann N Y Acad Sci 2014; 1325:15-22. [DOI: 10.1111/nyas.12518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Silvia Calpe
- Center for Experimental Medicine; Academic Medical Center; Amsterdam the Netherlands
| | - Debora Compare
- Department of Clinical and Experimental Medicine; University of Naples Federico II; Naples Italy
| | - Luigi Mari
- Center for Experimental Medicine; Academic Medical Center; Amsterdam the Netherlands
- Department of Gastroenterology and Hepatology; Academic Medical Center; Amsterdam the Netherlands
| | - Gerardo Nardone
- Department of Clinical and Experimental Medicine; University of Naples Federico II; Naples Italy
| | - Kaushal Parikh
- Center for Experimental Medicine; Academic Medical Center; Amsterdam the Netherlands
- Department of Gastroenterology and Hepatology; Academic Medical Center; Amsterdam the Netherlands
| |
Collapse
|
48
|
Riondino S, Roselli M, Palmirotta R, Della-Morte D, Ferroni P, Guadagni F. Obesity and colorectal cancer: Role of adipokines in tumor initiation and progression. World J Gastroenterol 2014; 20:5177-5190. [PMID: 24833848 PMCID: PMC4017033 DOI: 10.3748/wjg.v20.i18.5177] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 01/20/2014] [Accepted: 03/06/2014] [Indexed: 02/06/2023] Open
Abstract
Obesity-associated diseases account for a large portion of public health challenges. Among obesity-related disorders, a direct and independent relationship has been ascertained for colorectal cancer (CRC). The evidence that adipocyte hypertrophy and excessive adipose tissue accumulation (mainly visceral) can promote pathogenic adipocyte and adipose tissue-related diseases, has led to formulate the concept of “adiposopathy”, defined as adipocyte and adipose tissue dysfunction that contributes to metabolic syndrome. Adipose tissue can, indeed, be regarded as an important and highly active player of the innate immune response, in which cytokine/adipokine secretion is responsible for a paracrine loop between adipocytes and macrophages, thus contributing to the systemic chronic low-grade inflammation associated with visceral obesity, which represents a favorable niche for tumor development. The adipocyte itself participates as a central mediator of this inflammatory response in obese individuals by secreting hormones, growth factors and proinflammatory cytokines, which are of particular relevance for the pathogenesis of CRC. Among adipocyte-secreted hormones, the most relevant to colorectal tumorigenesis are adiponectin, leptin, resistin and ghrelin. All these molecules have been involved in cell growth and proliferation, as well as tumor angiogenesis and it has been demonstrated that their expression changes from normal colonic mucosa to adenoma and adenocarcinoma, suggesting their involvement in multistep colorectal carcinogenesis. These findings have led to the hypothesis that an unfavorable adipokine profile, with a reduction of those with an anti-inflammatory and anti-cancerous activity, might serve as a prognostic factor in CRC patients and that adipokines or their analogues/antagonists might become useful agents in the management or chemoprevention of CRC.
Collapse
|
49
|
Overweight, obesity and endometrial cancer risk: results from a systematic review and meta-analysis. Int J Biol Markers 2014; 29:e21-9. [PMID: 24170556 DOI: 10.5301/jbm.5000047] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/06/2013] [Indexed: 12/21/2022]
Abstract
AIM Findings from recent studies suggest that obesity may be associated with an increased risk of endometrial cancer, but several earlier studies were less conclusive. Here we strive to estimate this relationship in a meta-analysis of published data. METHODS We searched Pubmed and Embase for studies on body mass index and the risk of endometrial cancer, published from 1989 to 2011. Data were independently extracted and analyzed using random or fixed effects meta-analysis depending on the degree of heterogeneity. RESULTS Seven cohort studies and 11 case-control studies were included in the meta-analysis. Overall, the conditions of excess body weight ([EBW] defined as body mass index [BMI] ≥25 kg/m²), obesity (BMI ≥30 kg/m²) and overweight (25< BMI <30 kg/m²) were associated with an increased risk of endometrial cancer (relative risk [RR] for EBW=1.62, 95% confidence interval [CI], 1.39-1.89; for obesity RR=2.54, 95% CI, 2.11-3.06; for overweight RR=1.32, 95% CI, 1.16-1.50). Subgroup analyses showed that the positive associations were independent of study design, geographic locations, self-reported BMI, alcohol use, smoking habit, history of diabetes, hormone therapy, age at menarche, age at menopause, parity, and age at first full term pregnancy. However, there was no statistically significant association between EBW and endometrial cancer risk for measured BMI (for EBW RR=1.29, 95% CI, 0.66-2.53). CONCLUSIONS The findings from this meta-analysis strongly support that the conditions of EBW, overweight, and obesity are all associated with an increased risk of endometrial cancer. Also, the strength of the association increases with increasing BMI.
Collapse
|
50
|
Effect of body mass index in patients undergoing resection for gastric cancer: a single center US experience. J Gastrointest Surg 2014; 18:505-11. [PMID: 24443204 DOI: 10.1007/s11605-014-2455-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Accepted: 01/03/2014] [Indexed: 01/31/2023]
Abstract
INTRODUCTION With rising obesity in the USA, the impact of body mass index (BMI) on survival in gastric cancer remains unclear. METHODS An institutional database of patients undergoing surgical evaluation for gastric cancer was reviewed. Patients were stratified by the following BMI: <18.5, 18.5-25, 25.1-30, and >30. Clinicopathologic factors and overall survival (OS) were analyzed. RESULTS From 1997 to 2012, 222 patients underwent exploration for gastric adenocarcinoma. 186 (84%) had BMI recorded: nine (5%) <18.5, 72 (39%) 18.5-25, 62 (33%) 25.1-30, and 43 (23%) >30. One hundred thirty-five (73%) ultimately underwent resection. Although American Society of Anesthesiology score and blood loss were not associated with increasing BMI, operative time was longer, p = 0.02. Proximal location, perineural invasion, lymphovascular invasion, positive surgical margins, and positive lymph nodes (LN+) were all associated with worse OS but not with increased BMI. Although increased BMI was associated with a lower lymph node count, p = 0.004, the number of LN + and final pathologic stage were not associated with BMI. Additionally, use of neoadjuvant or adjuvant chemotherapy was not associated with BMI. Median OS was 22 months. Median OS was improved with increased BMI: 21 months for <18.5, 13 months 18.5-25, 28 months 25-30, and 34 months >30, p = 0.02. Disease-free survival (DFS) was similar: 2 months for <18.5, 7 months 18.5-25, 15 months 25.1-30, and 15 months >30, p = 0.02. CONCLUSION Although BMI may impact the technical difficulty of resection for gastric cancer, increasing BMI is not associated with more advanced disease. In this experience, increased BMI does not adversely impact OS or DFS.
Collapse
|