1
|
Kunutsor SK, Lehoczki A, Laukkanen JA. Coffee consumption, cancer, and healthy aging: epidemiological evidence and underlying mechanisms. GeroScience 2024:10.1007/s11357-024-01332-8. [PMID: 39266809 DOI: 10.1007/s11357-024-01332-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 08/26/2024] [Indexed: 09/14/2024] Open
Abstract
This comprehensive review examines the role of coffee consumption in promoting healthy aging and its potential impact on cancer prevention. Previous research has shown that moderate coffee intake may contribute to extending healthspan and enhancing longevity through beneficial effects on cardiometabolic health and key biological processes involved in aging. However, the relationship between coffee consumption and cancer risk remains controversial. This review synthesizes longitudinal observational and interventional data on the effects of coffee consumption on overall and site-specific cancers, explores underlying biological mechanisms, and discusses clinical and public health implications. Additionally, the review highlights evidence from Mendelian randomization (MR) studies to assess potential causal relationships. Our findings suggest that coffee consumption is associated with a reduced risk of several cancers, including skin, liver, prostate, and endometrial cancers, and may also lower cancer recurrence rates, particularly in colorectal cancer. These protective associations appear consistent across different demographic groups, with the most significant benefits observed at consumption levels of three or more cups per day. However, evidence is inconclusive for many other cancers, and coffee consumption is consistently linked to an increased risk of lung cancer. MR studies generally do not support a strong causal relationship for most cancers, though some suggest potential protective effects for hepatocellular, colorectal, and possibly prostate cancers, with mixed results for ovarian cancer and an increased risk for esophageal cancer and multiple myeloma. The protective effect of coffee on liver and prostate cancer is supported by both observational and MR studies. The potential anti-cancer benefits of coffee are attributed to its bioactive compounds, such as caffeine, chlorogenic acids, and diterpenes, which possess antioxidant and anti-inflammatory properties. These compounds may reduce oxidative stress, inhibit cancer cell proliferation, induce apoptosis, and modulate hormone levels. The review emphasizes the need for further research to clarify dose-response relationships, causal associations, and the biological mechanisms underlying these associations. While coffee consumption appears to contribute to cancer prevention and healthy aging, caution is warranted due to the increased risk of certain cancers, highlighting the complexity of its health effects.
Collapse
Affiliation(s)
- Setor K Kunutsor
- Section of Cardiology, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, 409 Tache Avenue, St. Boniface Hospital, Winnipeg, MB, R2H 2A6, Canada.
| | - Andrea Lehoczki
- Department of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Jari A Laukkanen
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
- Institute of Clinical Medicine, Department of Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Wellbeing Services County of Central Finland, Jyväskylä, Finland District, Jyväskylä, Finland
| |
Collapse
|
2
|
Chan DSM, Cariolou M, Markozannes G, Balducci K, Vieira R, Kiss S, Becerra-Tomás N, Aune D, Greenwood DC, González-Gil EM, Copson E, Renehan AG, Bours M, Demark-Wahnefried W, Hudson MM, May AM, Odedina FT, Skinner R, Steindorf K, Tjønneland A, Velikova G, Baskin ML, Chowdhury R, Hill L, Lewis SJ, Seidell J, Weijenberg MP, Krebs J, Cross AJ, Tsilidis KK. Post-diagnosis dietary factors, supplement use and colorectal cancer prognosis: A Global Cancer Update Programme (CUP Global) systematic literature review and meta-analysis. Int J Cancer 2024; 155:445-470. [PMID: 38692645 DOI: 10.1002/ijc.34906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 12/15/2023] [Accepted: 01/17/2024] [Indexed: 05/03/2024]
Abstract
The role of diet in colorectal cancer prognosis is not well understood and specific lifestyle recommendations are lacking. We searched for randomised controlled trials (RCTs) and longitudinal observational studies on post-diagnosis dietary factors, supplement use and colorectal cancer survival outcomes in PubMed and Embase from inception until 28th February 2022. Random-effects dose-response meta-analyses were conducted when at least three studies had sufficient information. The evidence was interpreted and graded by the CUP Global independent Expert Committee on Cancer Survivorship and Expert Panel. Five RCTs and 35 observational studies were included (30,242 cases, over 8700 all-cause and 2100 colorectal cancer deaths, 3700 progression, recurrence, or disease-free events). Meta-analyses, including 3-10 observational studies each, were conducted for: whole grains, nuts/peanuts, red and processed meat, dairy products, sugary drinks, artificially sweetened beverages, coffee, alcohol, dietary glycaemic load/index, insulin load/index, marine omega-3 polyunsaturated fatty acids, supplemental calcium, circulating 25-hydroxyvitamin D (25[OH]D) and all-cause mortality; for alcohol, supplemental calcium, circulating 25(OH)D and colorectal cancer-specific mortality; and for circulating 25(OH)D and recurrence/disease-free survival. The overall evidence was graded as 'limited'. The inverse associations between healthy dietary and/or lifestyle patterns (including diets that comprised plant-based foods), whole grains, total, caffeinated, or decaffeinated coffee and all-cause mortality and the positive associations between unhealthy dietary patterns, sugary drinks and all-cause mortality provided 'limited-suggestive' evidence. All other exposure-outcome associations provided 'limited-no conclusion' evidence. Additional, well-conducted cohort studies and carefully designed RCTs are needed to develop specific lifestyle recommendations for colorectal cancer survivors.
Collapse
Affiliation(s)
- Doris S M Chan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Margarita Cariolou
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Georgios Markozannes
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| | - Katia Balducci
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Rita Vieira
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Sonia Kiss
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Nerea Becerra-Tomás
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Dagfinn Aune
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Nutrition, Oslo New University College, Oslo, Norway
- Department of Research, The Cancer Registry of Norway, Oslo, Norway
| | - Darren C Greenwood
- Leeds Institute for Data Analytics, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | - Esther M González-Gil
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Ellen Copson
- Cancer Sciences Academic Unit, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Andrew G Renehan
- The Christie NHS Foundation Trust, Manchester Cancer Research Centre, NIHR Manchester Biomedical Research Centre, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Martijn Bours
- Department of Epidemiology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Wendy Demark-Wahnefried
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Melissa M Hudson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Anne M May
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Roderick Skinner
- Department of Paediatric and Adolescent Haematology/Oncology, Great North Children's Hospital and Translational and Clinical Research Institute, and Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Karen Steindorf
- Division of Physical Activity, Prevention and Cancer, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anne Tjønneland
- Danish Cancer Society Research Center, Diet, Cancer and Health, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - Galina Velikova
- School of Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, UK
| | | | - Rajiv Chowdhury
- Department of Global Health, Robert Stempel College of Public Health and Social Work, Florida International University, Miami, Florida, USA
| | - Lynette Hill
- World Cancer Research Fund International, London, UK
| | - Sarah J Lewis
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Jaap Seidell
- Department of Health Sciences, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Matty P Weijenberg
- Department of Epidemiology, GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - John Krebs
- Department of Biology, University of Oxford, Oxford, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Konstantinos K Tsilidis
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
| |
Collapse
|
3
|
Oyelere AM, Kok DE, Bos D, Gunter MJ, Ferrari P, Keski-Rahkonen P, de Wilt JHW, van Halteren HK, Kouwenhoven EA, van Duijnhoven FJB, Kampman E. Coffee consumption is associated with a reduced risk of colorectal cancer recurrence and all-cause mortality. Int J Cancer 2024; 154:2054-2063. [PMID: 38346920 DOI: 10.1002/ijc.34879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/13/2024] [Accepted: 01/16/2024] [Indexed: 04/14/2024]
Abstract
Coffee consumption has been associated with a reduced risk of developing colorectal cancer (CRC). However, it is not clear whether coffee consumption is related to CRC progression. Hence, we assessed the association of coffee consumption with CRC recurrence and all-cause mortality using data from a prospective cohort study of 1719 stage I-III CRC patients in the Netherlands. Coffee consumption and other lifestyle characteristics were self-reported using questionnaires at the time of diagnosis. We retrieved recurrence and all-cause mortality data from the Netherlands Cancer Registry and the Personal Records Database, respectively. Cox proportional hazard regression models with and without restricted cubic splines were used to calculate hazard ratios (HR) and 95% confidence intervals (CI) adjusted for age, sex, education, smoking status, cancer stage and tumor location. We observed 257 recurrences during a 6.2-year median follow-up and 309 deaths during a 6.6-year median follow-up. Consuming more than 4 cups/d of coffee compared to an intake of <2 cups/d was associated with a 32% lower risk of CRC recurrence (95% CI: 0.49, 0.94,). The association between coffee consumption and all-cause mortality was U-shaped; coffee intake seemed optimal at 3-5 cups/d with the lowest risk at 4 cups/d (HR: 0.68, 95% CI: 0.53, 0.88). Our results suggest that coffee consumption may be associated with a lower risk of CRC recurrence and all-cause mortality. The association between coffee consumption and all-cause mortality appeared nonlinear. More studies are needed to understand the mechanism by which coffee consumption might improve CRC prognosis.
Collapse
Affiliation(s)
- Abisola M Oyelere
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Dieuwertje E Kok
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| | - Daniel Bos
- Department of Epidemiology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
- Department of Epidemiology and Biostatistics School of Public Health, Imperial College London, London, UK
| | - Pietro Ferrari
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Pekka Keski-Rahkonen
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), Lyon, France
| | - Johannes H W de Wilt
- Department of Surgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Henk K van Halteren
- Department of Internal Medicine, Admiraal de Ruyter Ziekenhuis, Goes, The Netherlands
| | | | | | - Ellen Kampman
- Division of Human Nutrition and Health, Wageningen University & Research, Wageningen, The Netherlands
| |
Collapse
|
4
|
Sharman R, Harris Z, Ernst B, Mussallem D, Larsen A, Gowin K. Lifestyle Factors and Cancer: A Narrative Review. Mayo Clin Proc Innov Qual Outcomes 2024; 8:166-183. [PMID: 38468817 PMCID: PMC10925935 DOI: 10.1016/j.mayocpiqo.2024.01.004] [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] [Indexed: 03/13/2024] Open
Abstract
Lifestyle factors and their impact on cancer prevention, prognosis, and survivorship are increasingly recognized in the medical literature. Lifestyle factors are primarily defined here as diet and physical activity. We conducted a narrative review of the primary published data, including randomized controlled trials and prospective studies, on the impact of primary lifestyle factors on oncogenesis and clinical outcomes in the preventative and survivorship setting. First, we discuss the oncogenic mechanisms behind primary lifestyle factors (diet, physical activity and, within these 2, obesity). Then, we discuss the impact of adherence to lifestyle guidelines and dietary patterns on cancer incidence based on primary data. Owing to the plethora of published literature, to summarize the data in a more efficient manner, we describe the role of physical activity on cancer incidence using summative systematic reviews. We end by synthesizing the primary data on lifestyle factors in the survivorship setting and conclude with potential future directions. In brief, the various large-scale studies investigating the role diet and physical activity have reported a beneficial effect on cancer prevention and survivorship. Although the impact of single lifestyle factors on cancer incidence risk reduction is generally supported, holistic approaches to address the potential synergistic impact of multiple lifestyle factors together in concert is limited. Future research to identify the potentially synergistic effects of lifestyle modifications on oncogenesis and clinical outcomes is needed, particularly in cancer subtypes beyond colorectal and breast cancers.
Collapse
Affiliation(s)
- Reya Sharman
- Division of Hematology Oncology, Department of Medicine, University of Arizona, Tucson, AZ
| | - Zoey Harris
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ
| | - Brenda Ernst
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ
| | - Dawn Mussallem
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Ashley Larsen
- Division of Hematology Oncology, Department of Medicine, University of Arizona, Tucson, AZ
| | - Krisstina Gowin
- Division of Hematology Oncology, Department of Medicine, University of Arizona, Tucson, AZ
| |
Collapse
|
5
|
Kyaw TS, Upadhyay V, Tolstykh I, Van Loon K, Laffan A, Stanfield D, Gempis D, Kenfield SA, Chan JM, Piawah S, Atreya CE, Ng K, Venook A, Kidder W, Turnbaugh PJ, Van Blarigan EL. Variety of Fruit and Vegetables and Alcohol Intake are Associated with Gut Microbial Species and Gene Abundance in Colorectal Cancer Survivors. Am J Clin Nutr 2023; 118:518-529. [PMID: 37474105 PMCID: PMC10550847 DOI: 10.1016/j.ajcnut.2023.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND Adherence to the American Cancer Society (ACS) guidelines of avoiding obesity, maintaining physical activity, and consuming a diet rich in fruits, vegetables, and whole grains is associated with longer survival in colorectal cancer (CRC) survivors. Dietary components of the ACS guidelines may act in part by changing the microbiome, which is implicated in CRC outcomes. OBJECTIVES We conducted a pilot cross-sectional study to explore associations between ACS guidelines and the gut microbiome. METHODS Stool samples and questionnaires were collected from 28 CRC survivors at the University of California, San Francisco from 2019 to 2020. ACS scores were calculated based on validated questionnaires. Gut microbial community structure from 16S amplicons and gene/pathway abundances from metagenomics were tested for associations with the ACS score and its components using ANOVA and general linear models. RESULTS The overall ACS score was not significantly associated with variations in the fecal microbiota. However, fruit and vegetable intake and alcohol intake accounted for 19% (P = 0.005) and 13% (P = 0.01) of variation in the microbiota, respectively. Fruit/vegetable consumption was associated with increased microbial diversity, increased Firmicutes, decreased Bacteroidota, and changes to multiple genes and metabolic pathways, including enriched pathways for amino acid and short-chain fatty acid biosynthesis and plant-associated sugar degradation. In contrast, alcohol consumption was positively associated with overall microbial diversity, negatively associated with Bacteroidota abundance, and associated with changes to multiple genes and metabolic pathways. The other components of the ACS score were not statistically significantly associated with the fecal microbiota in our sample. CONCLUSIONS These results guide future studies examining the impact of changes in the intake of fruits, vegetables, and alcoholic drinks on the gut microbiome of CRC survivors.
Collapse
Affiliation(s)
- Than S Kyaw
- Department of Microbiology and Immunology, University of California, San Francisco, CA, United States
| | - Vaibhav Upadhyay
- Department of Microbiology and Immunology, University of California, San Francisco, CA, United States; Department of Medicine, University of California, San Francisco, CA, United States
| | - Irina Tolstykh
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States
| | - Katherine Van Loon
- Department of Medicine, University of California, San Francisco, CA, United States; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Angela Laffan
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Dalila Stanfield
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Daryll Gempis
- Department of Microbiology and Immunology, University of California, San Francisco, CA, United States
| | - Stacey A Kenfield
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States; Department of Urology, University of California, San Francisco, United States
| | - June M Chan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States; Department of Urology, University of California, San Francisco, United States
| | - Sorbarikor Piawah
- Department of Medicine, University of California, San Francisco, CA, United States; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Chloe E Atreya
- Department of Medicine, University of California, San Francisco, CA, United States; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Kimmie Ng
- Dana Farber Cancer Institute, Boston, MA, United States
| | - Alan Venook
- Department of Medicine, University of California, San Francisco, CA, United States; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Wesley Kidder
- Department of Medicine, University of California, San Francisco, CA, United States; University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, United States
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, CA, United States; Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, United States.
| | - Erin L Van Blarigan
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States; Department of Urology, University of California, San Francisco, United States.
| |
Collapse
|
6
|
Gregg JR, Kim J, Logothetis C, Hanash S, Zhang X, Manyam G, Muir K, Giles GG, Stanford JL, Berndt SI, Kogevinas M, Brenner H, Eeles RA, Wei P, Daniel CR. Coffee Intake, Caffeine Metabolism Genotype, and Survival Among Men with Prostate Cancer. Eur Urol Oncol 2023; 6:282-288. [PMID: 35995710 PMCID: PMC9939555 DOI: 10.1016/j.euo.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/26/2022] [Accepted: 07/20/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Coffee intake may lower prostate cancer risk and progression, but postdiagnosis outcomes by caffeine metabolism genotype are not well characterized. OBJECTIVE To evaluate associations between coffee intake, caffeine metabolism genotype, and survival in a large, multicenter study of men with prostate cancer. DESIGN, SETTING, AND PARTICIPANTS Data from The PRACTICAL Consortium database for 5727 men with prostate cancer from seven US, Australian, and European studies were included. The cases included had data available for the CYP1A2 -163C>A rs762551 single-nucleotide variant associated with caffeine metabolism, coffee intake, and >6 mo of follow-up. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Multivariable-adjusted Cox proportional hazards models across pooled patient-level data were used to compare the effect of coffee intake (categorized as low [reference], high, or none/very low) in relation to overall survival (OS) and prostate cancer-specific survival (PCSS), with stratified analyses conducted by clinical disease risk and genotype. RESULTS AND LIMITATIONS High coffee intake appeared to be associated with longer PCSS (hazard ratio [HR] 0.85, 95% confidence interval [CI] 0.68-1.08; p = 0.18) and OS (HR 0.90, 95% CI 0.77-1.07; p = 0.24), although results were not statistically significant. In the group with clinically localized disease, high coffee intake was associated with longer PCSS (HR 0.66, 95% CI 0.44-0.98; p = 0.040), with comparable results for the group with advanced disease (HR 0.92, 95% CI 0.69-1.23; p = 0.6). High coffee intake was associated with longer PCSS among men with the CYP1A2 AA (HR 0.67, 95% CI 0.49-0.93; p = 0.017) but not the AC/CC genotype (p = 0.8); an interaction was detected (p = 0.042). No associations with OS were observed in subgroup analyses (p > 0.05). Limitations include the nominal statistical significance and residual confounding. CONCLUSIONS Coffee intake was associated with longer PCSS among men with a CYP1A2 -163AA (*1F/*1F) genotype, a finding that will require further replication. PATIENT SUMMARY It is likely that coffee intake is associated with longer prostate cancer-specific survival in certain groups, but more research is needed to fully understand which men may benefit and why.
Collapse
Affiliation(s)
- Justin R Gregg
- Department of Urology, Division of Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jeri Kim
- Merck & Co., Kenilworth, NJ, USA
| | - Christopher Logothetis
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sam Hanash
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiaotao Zhang
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ganiraju Manyam
- Department of Biostatistics, Division of Basic Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth Muir
- Division of Population Health, Health Services Research and Primary Care, University of Manchester, Manchester, UK; Warwick Medical School, University of Warwick, Coventry, UK
| | - Graham G Giles
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia; Precision Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Manolis Kogevinas
- ISGlobal, Barcelona, Spain; Hospital del Mar Medical Research Institute, Barcelona, Spain; Universitat Pompeu Fabra, Barcelona, Spain; CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany
| | - Rosalind A Eeles
- The Institute of Cancer Research, London, UK; Royal Marsden NHS Foundation Trust, London, UK
| | - Peng Wei
- Department of Biostatistics, Division of Basic Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carrie R Daniel
- Department of Epidemiology, Division of Cancer Prevention and Population Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
7
|
Salas S, Cottet V, Dossus L, Fassier P, Ginhac J, Latino-Martel P, Romieu I, Schneider S, Srour B, Touillaud M, Touvier M, Ancellin R. Nutritional Factors during and after Cancer: Impacts on Survival and Quality of Life. Nutrients 2022; 14:2958. [PMID: 35889914 PMCID: PMC9323157 DOI: 10.3390/nu14142958] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
The French National Cancer Institute conducted a collective expertise study with researchers and clinical experts from the French Network for Nutrition And Cancer Research (NACRe Network). The objective was to update the state of knowledge on the impacts of nutritional factors on clinical endpoints during or after cancer. Data from 150 meta-analyses, pooled analyses or intervention trials and 93 cohort studies were examined; they concerned 8 nutritional factors, 6 clinical events and 20 cancer locations. This report shows that some nutritional factors have impacts on mortality and on the risks of recurrence or second primary cancer in cancer patients. Therefore, high-risk nutritional conditions can be encountered for certain cancer sites: from the diagnosis and throughout the health care pathways, weight loss (lung and esophageal cancers), malnutrition (lung, esophageal, colorectal, pancreatic, gastric and liver cancers), weight gain (colorectal, breast and kidney cancers) and alcohol consumption (upper aerodigestive cancers) should be monitored; and after cancer treatments, excess weight should be detected (colorectal, breast and kidney cancers). These situations require nutritional assessments, and even support or management by health care professionals, in the context of tertiary prevention. This report also highlights some limitations regarding the existing literature and some needs for future research.
Collapse
Affiliation(s)
- Sébastien Salas
- AP-HM, Timone Hospital, Aix Marseille University, 13000 Marseille, France;
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
| | - Vanessa Cottet
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- INSERM UMR1231/CIC 1432, University Hospital, LabEx LipSTIC ANR-11-LABX-0021, University of Burgundy-Franche-Comté, 21000 Dijon, France
| | - Laure Dossus
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- International Agency for Research on Cancer, 69000 Lyon, France
| | - Philippine Fassier
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Gustave Roussy Institute, 94800 Villejuif, France
| | - Julie Ginhac
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Nutritional Epidemiology Research Team (EREN), Sorbonne Paris Nord University, INSERM, INRAE, CNAM, Epidemiology and Statistics Research Centre (CRESS), University of Paris, 93022 Bobigny, France
| | - Paule Latino-Martel
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Nutritional Epidemiology Research Team (EREN), Sorbonne Paris Nord University, INSERM, INRAE, CNAM, Epidemiology and Statistics Research Centre (CRESS), University of Paris, 93022 Bobigny, France
| | - Isabelle Romieu
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- International Agency for Research on Cancer, 69000 Lyon, France
| | - Stéphane Schneider
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- University Hospital, University of Côte d’Azur, 06000 Nice, France
| | - Bernard Srour
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Nutritional Epidemiology Research Team (EREN), Sorbonne Paris Nord University, INSERM, INRAE, CNAM, Epidemiology and Statistics Research Centre (CRESS), University of Paris, 93022 Bobigny, France
| | - Marina Touillaud
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Léon-Bérard Cancer Centre, UA8 Inserm, 69000 Lyon, France
| | - Mathilde Touvier
- The French Network for Nutrition and Cancer Research (NACRe Network), 78350 Jouy-en-Josas, France; (L.D.); (P.F.); (J.G.); (P.L.-M.); (I.R.); (S.S.); (B.S.); (M.T.); (M.T.)
- Nutritional Epidemiology Research Team (EREN), Sorbonne Paris Nord University, INSERM, INRAE, CNAM, Epidemiology and Statistics Research Centre (CRESS), University of Paris, 93022 Bobigny, France
| | - Raphaëlle Ancellin
- The French National Cancer Institute (INCa), 92012 Boulogne-Billancourt, France;
| |
Collapse
|
8
|
Bartolomeu AR, Romualdo GR, Lisón CG, Besharat ZM, Corrales JAM, Chaves MÁG, Barbisan LF. Caffeine and Chlorogenic Acid Combination Attenuate Early-Stage Chemically Induced Colon Carcinogenesis in Mice: Involvement of oncomiR miR-21a-5p. Int J Mol Sci 2022; 23:ijms23116292. [PMID: 35682971 PMCID: PMC9181067 DOI: 10.3390/ijms23116292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/27/2022] [Accepted: 05/28/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of most common cancers worldwide, with high rates of mortality. Epidemiological findings demonstrate that coffee consumption reduces the risk of developing CRC by ~13%. In general, in vivo and in vitro findings demonstrate the antiproliferative, antioxidant and proapoptotic effects of brewed coffee or major bioavailable coffee compounds. Thus, it was assessed whether caffeine (CAF) and/or chlorogenic acid (CGA) attenuates the early-stage of chemically induced mouse colon carcinogenesis. Male Swiss mice were submitted to a 1,2-dimethylhydrazine/deoxycholic acid (DMH/DCA)-induced colon carcinogenesis model. These animals received CAF (50 mg/kg), CGA (25 mg/kg) or CAF+CGA (50 + 25 mg/kg) intragastrically for five times/week for ten weeks. CAF+CGA had the most pronounced effects on decreasing epithelial cell proliferation (Ki-67) and increasing apoptosis (cleaved caspase-3) in colonic crypts. This treatment also decreased the levels of proinflammatory cytokines IL-6, IL-17 and TNF-α, and downregulated the oncomiR miR-21a-5p in the colon. Accordingly, the analysis of miR-21a-5p targets demonstrated the genes involved in the negative regulation of proliferation and inflammation, and the positive regulation of apoptosis. Ultimately, CAF+CGA attenuated preneoplastic aberrant crypt foci (ACF) development. Our findings suggest that a combination of coffee compounds reduces early-stage colon carcinogenesis by the modulation of miR-21a-5p expression, highlighting the importance of coffee intake to prevent CRC.
Collapse
Affiliation(s)
- Ariane Rocha Bartolomeu
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (A.R.B.); (G.R.R.)
| | - Guilherme Ribeiro Romualdo
- Department of Pathology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (A.R.B.); (G.R.R.)
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
| | - Carmen Griñán Lisón
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- GENYO (Centre for Genomics and Oncological Research), Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain
- UGC de Oncología Médica, Complejo Hospitalario de Jaen, 23007 Jaen, Spain
| | - Zein Mersini Besharat
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy;
| | - Juan Antonio Marchal Corrales
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- Department of Human Anatomy and Embryology, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Maria Ángel García Chaves
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, University of Granada, 18012 Granada, Spain; (C.G.L.); (J.A.M.C.); (M.Á.G.C.)
- Department of Biochemistry and Molecular Biology III and Immunology, University of Granada, 18016 Granada, Spain
| | - Luís Fernando Barbisan
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil
- Correspondence:
| |
Collapse
|
9
|
Nutritional Treatment of Patients with Colorectal Cancer. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19116881. [PMID: 35682464 PMCID: PMC9180662 DOI: 10.3390/ijerph19116881] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/02/2022] [Accepted: 06/02/2022] [Indexed: 12/12/2022]
Abstract
Colorectal cancer is one of the most common cancers in Europe and the world. Cancer treatments have side effects and cause significant deterioration of the patient’s nutritional status. Patient malnutrition may worsen the health condition and prevent the deliberate effects of the therapy. The aim of this review was to describe the available data about clinical nutrition in colorectal cancer patients. A large proportion of colorectal cancer patients suffer from malnutrition, which negatively affects the survival prognosis, quality of life, and oncological therapy. Therefore, monitoring nutritional status during the treatment is essential and can be used to arrange proper nutritional therapy to enhance patient responses, prevent side effects, and shorten recovery time. The principles of nutrition during anticancer therapy should mainly consider light and low-fat foods, the exclusion of lactose and gluten-containing foods in certain cases, or the introduction of special dietary products such as oral nutrition supplements and it should be tailored to patients’ individual needs.
Collapse
|
10
|
Moy RH, Nguyen A, Loo JM, Yamaguchi N, Kajba CM, Santhanam B, Ostendorf BN, Wu YG, Tavazoie S, Tavazoie SF. Functional genetic screen identifies ITPR3/calcium/RELB axis as a driver of colorectal cancer metastatic liver colonization. Dev Cell 2022; 57:1146-1159.e7. [PMID: 35487218 PMCID: PMC9446818 DOI: 10.1016/j.devcel.2022.04.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/02/2022] [Accepted: 04/06/2022] [Indexed: 11/29/2022]
Abstract
Metastatic colonization is the primary cause of death from colorectal cancer (CRC). We employed genome-scale in vivo short hairpin RNA (shRNA) screening and validation to identify 26 promoters of CRC liver colonization. Among these genes, we identified a cluster that contains multiple targetable genes, including ITPR3, which promoted liver-metastatic colonization and elicited similar downstream gene expression programs. ITPR3 is a caffeine-sensitive inositol 1,4,5-triphosphate (IP3) receptor that releases calcium from the endoplasmic reticulum and enhanced metastatic colonization by inducing expression of RELB, a transcription factor that is associated with non-canonical NF-κB signaling. Genetic, cell biological, pharmacologic, and clinical association studies revealed that ITPR3 and RELB drive CRC colony formation by promoting cell survival upon substratum detachment or hypoxic exposure. RELB was sufficient to drive colonization downstream of ITPR3. Our findings implicate the ITPR3/calcium/RELB axis in CRC metastatic colony formation and uncover multiple clinico-pathologically associated targetable proteins as drivers of CRC metastatic colonization.
Collapse
Affiliation(s)
- Ryan H Moy
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Alexander Nguyen
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Jia Min Loo
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Norihiro Yamaguchi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Christina M Kajba
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Balaji Santhanam
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Benjamin N Ostendorf
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Y Gloria Wu
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Saeed Tavazoie
- Department of Biochemistry and Molecular Biophysics and Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
11
|
Impact of Diet and Exercise on Colorectal Cancer. Hematol Oncol Clin North Am 2022; 36:471-489. [DOI: 10.1016/j.hoc.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
12
|
Cheng E, Ou FS, Ma C, Spiegelman D, Zhang S, Zhou X, Bainter TM, Saltz LB, Niedzwiecki D, Mayer RJ, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Giovannucci EL, Van Blarigan EL, Brown JC, Ng K, Gross CP, Meyerhardt JA, Fuchs CS. Diet- and Lifestyle-Based Prediction Models to Estimate Cancer Recurrence and Death in Patients With Stage III Colon Cancer (CALGB 89803/Alliance). J Clin Oncol 2022; 40:740-751. [PMID: 34995084 PMCID: PMC8887946 DOI: 10.1200/jco.21.01784] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/08/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
PURPOSE Current tools in predicting survival outcomes for patients with colon cancer predominantly rely on clinical and pathologic characteristics, but increasing evidence suggests that diet and lifestyle habits are associated with patient outcomes and should be considered to enhance model accuracy. METHODS Using an adjuvant chemotherapy trial for stage III colon cancer (CALGB 89803), we developed prediction models of disease-free survival (DFS) and overall survival by additionally incorporating self-reported nine diet and lifestyle factors. Both models were assessed by multivariable Cox proportional hazards regression and externally validated using another trial for stage III colon cancer (CALGB/SWOG 80702), and visual nomograms of prediction models were constructed accordingly. We also proposed three hypothetical scenarios for patients with (1) good-risk, (2) average-risk, and (3) poor-risk clinical and pathologic features, and estimated their predictive survival by considering clinical and pathologic features with or without adding self-reported diet and lifestyle factors. RESULTS Among 1,024 patients (median age 60.0 years, 43.8% female), we observed 394 DFS events and 311 deaths after median follow-up of 7.3 years. Adding self-reported diet and lifestyle factors to clinical and pathologic characteristics meaningfully improved performance of prediction models (c-index from 0.64 [95% CI, 0.62 to 0.67] to 0.69 [95% CI, 0.67 to 0.72] for DFS, and from 0.67 [95% CI, 0.64 to 0.70] to 0.71 [95% CI, 0.69 to 0.75] for overall survival). External validation also indicated good performance of discrimination and calibration. Adding most self-reported favorable diet and lifestyle exposures to multivariate modeling improved 5-year DFS of all patients and by 6.3% for good-risk, 21.4% for average-risk, and 42.6% for poor-risk clinical and pathologic features. CONCLUSION Diet and lifestyle factors further inform current recurrence and survival prediction models for patients with stage III colon cancer.
Collapse
Affiliation(s)
- En Cheng
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | - Chao Ma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Donna Spiegelman
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
- Center on Methods for Implementation and Prevention Science, Yale School of Public Health, New Haven, CT
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Xin Zhou
- Department of Biostatistics, Yale School of Public Health, New Haven, CT
- Center on Methods for Implementation and Prevention Science, Yale School of Public Health, New Haven, CT
| | - Tiffany M. Bainter
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, MN
| | | | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, Durham, NC
| | - Robert J. Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Renaud Whittom
- Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada
| | | | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | | | | | - Edward L. Giovannucci
- Department of Epidemiology, and Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Erin L. Van Blarigan
- Department of Epidemiology and Biostatistics, and Urology, University of California, San Francisco, CA
| | - Justin C. Brown
- Cancer Metabolism Program, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Cary P. Gross
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
- Section of General Internal Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research Center, Yale Cancer Center, New Haven, CT
| | | | - Charles S. Fuchs
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT
- Hematology and Oncology Product Development, Genentech & Roche, South San Francisco, CA
| |
Collapse
|
13
|
Xie R, Chen X, Zhou J, Huang L, Jiang W. LncRNA CCHE1 participate in the postoperative distant recurrence of urothelial bladder cancer possibly by regulating ROCK1 expression. Crit Rev Eukaryot Gene Expr 2022; 32:23-31. [DOI: 10.1615/critreveukaryotgeneexpr.2022039524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
14
|
Ugai T, Haruki K, Väyrynen JP, Borowsky J, Fujiyoshi K, Lau MC, Akimoto N, Zhong R, Kishikawa J, Arima K, Shi SS, Zhao M, Fuchs CS, Zhang X, Giannakis M, Song M, Nan H, Meyerhardt JA, Wang M, Nowak JA, Ogino S. Coffee Intake of Colorectal Cancer Patients and Prognosis According to Histopathologic Lymphocytic Reaction and T-Cell Infiltrates. Mayo Clin Proc 2022; 97:124-133. [PMID: 34996545 PMCID: PMC8820462 DOI: 10.1016/j.mayocp.2021.09.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 08/19/2021] [Accepted: 09/10/2021] [Indexed: 01/03/2023]
Abstract
Given previous biologic evidence of immunomodulatory effects of coffee, we hypothesized that the association between coffee intake of colorectal cancer patients and survival differs by immune responses. Using a molecular pathologic epidemiology database of 4465 incident colorectal cancer cases, including 1262 cases with molecular data, in the Nurses' Health Study and the Health Professionals Follow-up Study, we examined the association between coffee intake of colorectal cancer patients and survival in strata of levels of histopathologic lymphocytic reaction and T-cell infiltrates in tumor tissue. We did not observe a significant association of coffee intake with colorectal cancer-specific mortality (multivariable-adjusted hazard ratio [HR] for 1-cup increase of coffee intake per day, 0.93; 95% CI, 0.84 to 1.03). Although statistical significance was not reached at the stringent level (α=.005), the association of coffee intake with colorectal cancer-specific mortality differed by Crohn disease-like lymphoid reaction (Pinteraction=.007). Coffee intake was associated with lower colorectal cancer-specific mortality in patients with high Crohn disease-like reaction (multivariable HR for 1-cup increase of coffee intake per day, 0.55; 95% CI, 0.37 to 0.81; Ptrend=.002) but not in patients with intermediate Crohn disease-like reaction (the corresponding HR, 1.02; 95% CI, 0.72 to 1.44) or negative/low Crohn disease-like reaction (the corresponding HR, 0.95; 95% CI, 0.83 to 1.07). The associations of coffee intake with colorectal cancer-specific mortality did not significantly differ by levels of other lymphocytic reaction or any T-cell subset (Pinteraction>.18). There is suggestive evidence for differential prognostic effects of coffee intake by Crohn disease-like lymphoid reaction in colorectal cancer.
Collapse
Affiliation(s)
- Tomotaka Ugai
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.
| | - Koichiro Haruki
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Juha P Väyrynen
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Jennifer Borowsky
- Conjoint Gastroenterology Department, QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Kenji Fujiyoshi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Mai Chan Lau
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Naohiko Akimoto
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rong Zhong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Junko Kishikawa
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kota Arima
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shan-Shan Shi
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Melissa Zhao
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, CT; Department of Medicine, Yale School of Medicine, New Haven, CT; Smilow Cancer Hospital, New Haven, CT
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Marios Giannakis
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston; Division of Gastroenterology, Massachusetts General Hospital, Boston
| | - Hongmei Nan
- Department of Global Health, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis; Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Molin Wang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA; Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA; Broad Institute of MIT and Harvard, Cambridge, MA.
| |
Collapse
|
15
|
Ghebrial M, Aktary ML, Wang Q, Spinelli JJ, Shack L, Robson PJ, Kopciuk KA. Predictors of CRC Stage at Diagnosis among Male and Female Adults Participating in a Prospective Cohort Study: Findings from Alberta's Tomorrow Project. Curr Oncol 2021; 28:4938-4952. [PMID: 34898587 PMCID: PMC8628758 DOI: 10.3390/curroncol28060414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 11/22/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of morbidity and mortality in Canada. CRC screening and other factors associated with early-stage disease can improve CRC treatment efficacy and survival. This study examined factors associated with CRC stage at diagnosis among male and female adults using data from a large prospective cohort study in Alberta, Canada. Baseline data were obtained from healthy adults aged 35–69 years participating in Alberta’s Tomorrow Project. Factors associated with CRC stage at diagnosis were evaluated using Partial Proportional Odds models. Analyses were stratified to examine sex-specific associations. A total of 267 participants (128 males and 139 females) developed CRC over the study period. Among participants, 43.0% of males and 43.2% of females were diagnosed with late-stage CRC. Social support, having children, and caffeine intake were predictors of CRC stage at diagnosis among males, while family history of CRC, pregnancy, hysterectomy, menopausal hormone therapy, lifetime number of Pap tests, and household physical activity were predictive of CRC stage at diagnosis among females. These findings highlight the importance of sex differences in susceptibility to advanced CRC diagnosis and can help inform targets for cancer prevention programs to effectively reduce advanced CRC and thus improve survival.
Collapse
Affiliation(s)
- Monica Ghebrial
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Michelle L. Aktary
- Faculty of Kinesiology, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Qinggang Wang
- Cancer Epidemiology and Prevention Research, Cancer Care Alberta, Alberta Health Services, Calgary, AB T2S 3C3, Canada;
| | - John J. Spinelli
- School of Population and Public Health, University of British Columbia, Vancouver, BC V6T 1Z3, Canada;
- Population Oncology, BC Cancer, Vancouver, BC V5Z 1L3, Canada
| | - Lorraine Shack
- Cancer Surveillance and Reporting, Alberta Health Services, Calgary, AB T2S 3C3, Canada;
| | - Paula J. Robson
- Department of Agricultural, Food and Nutritional Science and School of Public Health, University of Alberta, Edmonton, AB T6G 2P5, Canada;
- Cancer Care Alberta and Cancer Strategic Clinical Network, Alberta Health Services, Edmonton, AB T5J 3H1, Canada
| | - Karen A. Kopciuk
- Cancer Epidemiology and Prevention Research, Cancer Care Alberta, Alberta Health Services, Calgary, AB T2S 3C3, Canada;
- Departments of Oncology, Community Health Sciences and Mathematics and Statistics, University of Calgary, Calgary, AB T2N 4N2, Canada
- Correspondence:
| |
Collapse
|
16
|
Ancellin R, Cottet V, Dossus L, Fassier P, Gaillot de Saintignon J, Ginhac J, Romieu I, Salas S, Schneider S, Srour B, Touillaud M, Touvier M, Latino-Martel P. [Impact of nutritional factors during and after cancer]. Bull Cancer 2021; 108:455-464. [PMID: 33836862 DOI: 10.1016/j.bulcan.2021.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/07/2021] [Indexed: 02/07/2023]
Abstract
Nutritional factors (diet, weight, alcohol, physical activity) are identified as factors having an impact on the onset of several cancer sites. Less abundant scientific data also underline their impact on the tumor progression. A review of the scientific literature was carried out by a group of experts established by the French National Cancer Institute (INCa) to better document the influence of nutritional factors during and after cancer on outcomes such as overall mortality, cancer specific mortality, recurrence, second primary cancers and quality of life. This analysis of the literature completes messages of reduction of alcohol consumption, prevention of undernutrition or excess weight and adherence to dietary recommendations, avoiding the use of dietary supplements, fasting or restrictive diets and strengthens messages promoting the practice of physical activity and the fight against sedentary lifestyle.
Collapse
Affiliation(s)
- Raphaëlle Ancellin
- Institut national du cancer, 52, avenue André-Morizet, 92100 Boulogne-Billancourt, France.
| | - Vanessa Cottet
- CHU de Dijon, université de Bourgogne-Franche-Comté, Inserm UMR1231 & CIC1432, 7, boulevard Jeanne-d'Arc, 21079 Dijon, France
| | - Laure Dossus
- Centre international de recherche sur le cancer (CIRC), 150, cours Albert-Thomas, 69008 Lyon, France
| | | | | | - Julie Ginhac
- Cellule de coordination, domaine de Vilvert, réseau national alimentation cancer recherche (Réseau NACRe), 78350 Jouy-en-Josas, France
| | - Isabelle Romieu
- Center for Research on Population Health, National Institute of Public Health, Mexico, Mexique; Emory University, Hubert Department of Global Health, Atlanta, Georgia, États-Unis
| | - Sébastien Salas
- CHU de Timone, 264, rue Saint-Pierre, 13005 Marseille, France
| | - Stéphane Schneider
- Université Côte d'Azur, CHU de Nice, Hôpital de l'Archet, 151, route de Saint-Antoine, 06200 Nice, France
| | - Bernard Srour
- Cellule de coordination, domaine de Vilvert, réseau national alimentation cancer recherche (Réseau NACRe), 78350 Jouy-en-Josas, France; Université Sorbonne Paris Nord, université de Paris (CRESS), Inrae U1125, Cnam, équipe de recherche en épidémiologie nutritionnelle (EREN), centre de recherche en épidémiologie et statistiques, Inserm U1153, 74, rue Marcel-Cachin, 93017 Bobigny, France
| | - Marina Touillaud
- Centre Léon-Bérard, Inserm UA8 « Radiations : défense, santé et environnement », 28, rue Laennec, 69008 Lyon, France
| | - Mathilde Touvier
- Université Sorbonne Paris Nord, université de Paris (CRESS), Inrae U1125, Cnam, équipe de recherche en épidémiologie nutritionnelle (EREN), centre de recherche en épidémiologie et statistiques, Inserm U1153, 74, rue Marcel-Cachin, 93017 Bobigny, France
| | - Paule Latino-Martel
- Cellule de coordination, domaine de Vilvert, réseau national alimentation cancer recherche (Réseau NACRe), 78350 Jouy-en-Josas, France
| |
Collapse
|
17
|
Abstract
The substantial burden of colorectal cancer and its increasing trend in young adults highlight the importance of dietary and lifestyle modifications for improved cancer prevention and survivorship. In this chapter, we review the cutting-edge evidence for the interplay between diet/lifestyle and the gut microbiota in the incidence and prognosis of colorectal cancer. We focus on factors for which there are data supporting their importance for the gut microbiota and colorectal cancer, including excess body weight, fiber, red and processed meat, and coffee. We discuss the potential precision nutrition approaches for modifying and exploiting the gut microbiota for improved cancer prevention and survivorship.
Collapse
Affiliation(s)
- Kai Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
18
|
Hoang T, Kim H, Kim J. Dietary Intake in Association with All-Cause Mortality and Colorectal Cancer Mortality among Colorectal Cancer Survivors: A Systematic Review and Meta-Analysis of Prospective Studies. Cancers (Basel) 2020; 12:cancers12113391. [PMID: 33207660 PMCID: PMC7697273 DOI: 10.3390/cancers12113391] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/11/2020] [Accepted: 11/13/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Given that an extensive range of dietary factors has not been investigated among colorectal cancer (CRC) survivors to date, we carried out a systematic review and meta-analysis to determine the effects of both prediagnostic and postdiagnostic dietary intake on all-cause mortality and CRC-specific mortality among CRC survivors. In total, 45 studies were included in the final analysis of 35 food items, 8 macronutrients, 27 micronutrients, 2 dietary patterns, and 13 dietary indexes in association with all-cause mortality and CRC-specific mortality. We found that an unhealthy dietary pattern increased the risks of both all-cause mortality and CRC-specific mortality. The role of prediagnostic and postdiagnostic intake such as macronutrients and fatty acids could be different in the risk of all-cause mortality. Overall, comprehensive evidence for the effect of substantial numbers of prediagnostic and postdiagnostic dietary items on mortality outcomes is reported in this study. Abstract We carried out a systematic review and meta-analysis to determine the effects of both prediagnostic and postdiagnostic dietary intake on all-cause mortality and CRC-specific mortality among CRC survivors. An extensive search of PubMed and Embase was conducted to identify eligible studies. We applied a random-effects model to estimate the pooled relative risks (RRs)/hazard ratios (HRs) and their 95% confidence intervals (CIs). As a result, a total of 45 studies were included in the final analysis. Pooled effect sizes from at least three study populations showed that whole grains and calcium were inversely associated with all-cause mortality, with RRs/HRs (95% CIs) of 0.83 (0.69–0.99) and 0.84 (0.73–0.97), respectively. In contrast, a positive association between an unhealthy dietary pattern and both all-cause mortality (RR/HR = 1.47, 95% CI = 1.05–2.05) and CRC-specific mortality (RR/HR = 1.52, 95% CI = 1.13–2.06) was observed among CRC survivors. In the subgroup analysis by CRC diagnosis, prediagnostic and postdiagnostic dietary intake such as carbohydrates, proteins, lipids, and fiber were observed to have different effects on all-cause mortality. Overall, an unhealthy dietary pattern increased the risks of both all-cause mortality and CRC-specific mortality. The role of prediagnostic and postdiagnostic intake of dietary elements such as macronutrients and fatty acids could be different in the risk of all-cause mortality.
Collapse
|
19
|
Mackintosh C, Yuan C, Ou FS, Zhang S, Niedzwiecki D, Chang IW, O'Neil BH, Mullen BC, Lenz HJ, Blanke CD, Venook AP, Mayer RJ, Fuchs CS, Innocenti F, Nixon AB, Goldberg RM, O'Reilly EM, Meyerhardt JA, Ng K. Association of Coffee Intake With Survival in Patients With Advanced or Metastatic Colorectal Cancer. JAMA Oncol 2020; 6:1713-1721. [PMID: 32940631 PMCID: PMC7499248 DOI: 10.1001/jamaoncol.2020.3938] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Importance Several compounds found in coffee possess antioxidant, anti-inflammatory, and insulin-sensitizing effects, which may contribute to anticancer activity. Epidemiological studies have identified associations between increased coffee consumption and decreased recurrence and mortality of colorectal cancer. The association between coffee consumption and survival in patients with advanced or metastatic colorectal cancer is unknown. Objective To evaluate the association of coffee consumption with disease progression and death in patients with advanced or metastatic colorectal cancer. Design, Setting, and Participants This prospective observational cohort study included 1171 patients with previously untreated locally advanced or metastatic colorectal cancer who were enrolled in Cancer and Leukemia Group B (Alliance)/SWOG 80405, a completed phase 3 clinical trial comparing the addition of cetuximab and/or bevacizumab to standard chemotherapy. Patients reported dietary intake using a semiquantitative food frequency questionnaire at the time of enrollment. Data were collected from October 27, 2005, to January 18, 2018, and analyzed from May 1 to August 31, 2018. Exposures Consumption of total, decaffeinated, and caffeinated coffee measured in cups per day. Main Outcomes and Measures Overall survival (OS) and progression-free survival (PFS). Results Among the 1171 patients included in the analysis (694 men [59%]; median age, 59 [interquartile range, 51-67] years). The median follow-up time among living patients was 5.4 years (10th percentile, 1.3 years; IQR, 3.2-6.3 years). A total of 1092 patients (93%) had died or had disease progression. Increased consumption of coffee was associated with decreased risk of cancer progression (hazard ratio [HR] for 1-cup/d increment, 0.95; 95% CI, 0.91-1.00; P = .04 for trend) and death (HR for 1-cup/d increment, 0.93; 95% CI, 0.89-0.98; P = .004 for trend). Participants who consumed 2 to 3 cups of coffee per day had a multivariable HR for OS of 0.82 (95% CI, 0.67-1.00) and for PFS of 0.82 (95% CI, 0.68-0.99), compared with those who did not drink coffee. Participants who consumed at least 4 cups of coffee per day had a multivariable HR for OS of 0.64 (95% CI, 0.46-0.87) and for PFS of 0.78 (95% CI, 0.59-1.05). Significant associations were noted for both caffeinated and decaffeinated coffee. Conclusions and Relevance Coffee consumption may be associated with reduced risk of disease progression and death in patients with advanced or metastatic colorectal cancer. Further research is warranted to elucidate underlying biological mechanisms.
Collapse
Affiliation(s)
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Fang-Shu Ou
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Sui Zhang
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - I-Wen Chang
- Southeast Clinical Oncology Research Consortium, Winston-Salem, North Carolina
| | - Bert H O'Neil
- Department of Medicine, Indiana University School of Medicine, Indianapolis
| | - Brian C Mullen
- Alliance Statistics and Data Management Center, Mayo Clinic, Rochester, Minnesota
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles
| | - Charles D Blanke
- SWOG Group Chair's Office/Knight Cancer Institute, Oregon Health and Science University, Portland
| | - Alan P Venook
- Department of Medicine, University of California, San Francisco, School of Medicine
| | - Robert J Mayer
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Charles S Fuchs
- Yale Cancer Center and Smilow Cancer Hospital, New Haven, Connecticut
| | - Federico Innocenti
- Eshelman School of Pharmacy and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill
| | - Andrew B Nixon
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| | | | - Eileen M O'Reilly
- Weill Cornell Medical College, Cornell University and Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
Abstract
AIM Conduct a systematic review of available evidence on food and beverage intake during cancer treatment. OBJECTIVE Determine what food or beverages consumed during cancer treatment might prevent recurrence, subsequent malignancies, treatment-related toxicity, or death. BACKGROUND Food and beverage intake, as well as weight status, can integrate with cancer treatment to mitigate treatment-related toxicities, support treatment success, and prevent recurrence. Yet, evidence-based recommendations are lacking. METHODS We searched PubMed, Embase, and Cochran for research studies conducted within the last 10 years on food and beverage consumption during cancer treatment, with no restrictions on age or cancer type. Two reviewers independently extracted information on intervention type, diet, and outcomes; these data were confirmed by a third reviewer. RESULTS Nineteen studies were selected from 1551 potential studies. Nine were randomized controlled trials, analyzing high protein diets, short-term fasting, low-fat diets, FODMAP diet, or comparing consumption of 1 specific food or nutrient, including Concord grape juice, onions, and fiber. The remaining 10 studies were observational or retrospective and tracked treatment symptoms, general dietary intake, or weight status as well as consumption of specific foods including nuts, coffee, sugar-sweetened beverages. CONCLUSIONS Available evidence suggests food can be effective at ameliorating cancer treatment-related toxicities and improving prognosis, but more research is needed.
Collapse
|
21
|
Shao C, Zuo Q, Lin J, Yu RJ, Fu Y, Xiao M, Sun LL, Lin L. Effect of Chinese Herbal Medicine on the Survival of Colorectal Cancer Patients With Liver-Limited Metastases: A Retrospective Cohort Study, 2008 to 2017. Integr Cancer Ther 2020; 18:1534735419883687. [PMID: 31658839 PMCID: PMC6820185 DOI: 10.1177/1534735419883687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Background: Colorectal cancer (CRC) remains one of the leading contributors to cancer-related mortality and morbidity worldwide. Traditional Chinese medicines have been widely employed to treat various types of cancer in China. This investigation aims to determine the association between Chinese herbal medicine (CHM) therapy and survival outcomes in CRC patients with liver-limited metastases. Methods: A retrospective cohort study was performed among patients with colorectal liver metastases at the First Affiliated Hospital of Guangzhou University of Chinese Medicine in Guangzhou, China. Data from a series of consecutive patients were collected via an electronic medical record system or telephone follow-up. We defined high exposure as a period of CHM therapy lasting more than 6 months. The primary outcome was overall survival. Results: The study included the data of 191 patients from January 2008 to December 2017; 126 patients (65.97%) met the inclusion criteria of high exposure to CHM. Multivariate analyses revealed that high exposure to CHM was associated with better overall survival (hazard ratio = 0.444, 95% confidence interval = [0.213, 0.926], P = .030). The association was further confirmed by a subgroup exploratory analysis. Conclusion: Long-term CHM therapy is correlated with improved survival outcomes in CRC patients with liver-limited metastases.
Collapse
Affiliation(s)
- Cui Shao
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qian Zuo
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jietao Lin
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, China
| | - Rong Jian Yu
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China
| | - Yuanfeng Fu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Xiao
- Guangzhou University of Chinese Medicine, Guangzhou, China.,Shenzhen Chinese Medicine Hospital, Shenzhen, China
| | - Ling Ling Sun
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, China
| | - Lizhu Lin
- Guangzhou University of Traditional Chinese Medicine First Affiliated Hospital, Guangzhou, China
| |
Collapse
|
22
|
Souza LDSD, Carrero Horta IP, de Souza Rosa L, Barbosa Lima LG, Santos da Rosa J, Montenegro J, da Silva Santos L, Nana de Castro RB, Freitas-Silva O, Teodoro AJ. Effect of the roasting levels of Coffea arabica L. extracts on their potential antioxidant capacity and antiproliferative activity in human prostate cancer cells. RSC Adv 2020; 10:30115-30126. [PMID: 35518253 PMCID: PMC9056273 DOI: 10.1039/d0ra01179g] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/24/2020] [Indexed: 12/11/2022] Open
Abstract
Coffee, besides being one of the most consumed stimulating beverages in the world, has important bioactive activities, which have been attracting increasing attention from researchers. However, the standard process of roasting causes changes in its chemical composition. In the present study, extracts obtained from green and roasted beans (light, medium and dark) of Coffea arabica Linnaeus were submitted to high-power ultrasonic extraction and atomization by spray drying. Colorimetric analysis was used to classify the roasting levels of the dried extract samples. The effects of the roasting process on the bioactivity of the dried extracts were verified through the following assays: caffeine, chlorogenic acid and caffeic acid, by HPLC-PDA; total phenolics by Folin–Ciocalteu; antioxidant activity by DPPH, FRAP, ABTS and ORAC; antiproliferative activity, using the MTT assay; and cell cycle and apoptosis by flow cytometry in metastatic prostate cancer cell lines from bone (PC-3) and brain (DU-145). The results showed that the lowest levels of caffeine, chlorogenic and caffeic acids were observed in dark roasted coffee. In comparison to medium and dark extracts in PC-3 cells, the green and light coffee extracts had higher antioxidant activities and promoted cytotoxicity followed by cell cycle arrest in phase S and apoptosis induction. Thus, the roasting level of the coffee extracts may be related to the potential chemoprotective effects of Coffea arabica L. in prostate cancer cells. Coffee, besides being one of the most consumed stimulating beverages in the world, has important bioactive activities, which have been attracting increasing attention from researchers.![]()
Collapse
Affiliation(s)
| | | | - Lana de Souza Rosa
- Laboratory of Functional Foods
- Universidade Federal do Estado do Rio de Janeiro
- Rio de Janeiro
- Brazil
| | | | - Jeane Santos da Rosa
- Empresa Brasileira de Pesquisa Agropecuária
- Embrapa Agroindústria de Alimentos
- Rio de Janeiro
- Brazil
| | - Julia Montenegro
- Laboratory of Functional Foods
- Universidade Federal do Estado do Rio de Janeiro
- Rio de Janeiro
- Brazil
| | - Lauriza da Silva Santos
- Laboratory of Functional Foods
- Universidade Federal do Estado do Rio de Janeiro
- Rio de Janeiro
- Brazil
| | | | - Otniel Freitas-Silva
- Laboratory of Functional Foods
- Universidade Federal do Estado do Rio de Janeiro
- Rio de Janeiro
- Brazil
- Empresa Brasileira de Pesquisa Agropecuária
| | - Anderson Junger Teodoro
- Laboratory of Functional Foods
- Universidade Federal do Estado do Rio de Janeiro
- Rio de Janeiro
- Brazil
| |
Collapse
|
23
|
Song M, Chan AT, Sun J. Influence of the Gut Microbiome, Diet, and Environment on Risk of Colorectal Cancer. Gastroenterology 2020; 158:322-340. [PMID: 31586566 PMCID: PMC6957737 DOI: 10.1053/j.gastro.2019.06.048] [Citation(s) in RCA: 430] [Impact Index Per Article: 107.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 06/11/2019] [Accepted: 06/16/2019] [Indexed: 02/07/2023]
Abstract
Researchers have discovered associations between elements of the intestinal microbiome (including specific microbes, signaling pathways, and microbiota-related metabolites) and risk of colorectal cancer (CRC). However, it is unclear whether changes in the intestinal microbiome contribute to the development of sporadic CRC or result from it. Changes in the intestinal microbiome can mediate or modify the effects of environmental factors on risk of CRC. Factors that affect risk of CRC also affect the intestinal microbiome, including overweight and obesity; physical activity; and dietary intake of fiber, whole grains, and red and processed meat. These factors alter microbiome structure and function, along with the metabolic and immune pathways that mediate CRC development. We review epidemiologic and laboratory evidence for the influence of the microbiome, diet, and environmental factors on CRC incidence and outcomes. Based on these data, features of the intestinal microbiome might be used for CRC screening and modified for chemoprevention and treatment. Integrated prospective studies are urgently needed to investigate these strategies.
Collapse
Affiliation(s)
- Mingyang Song
- Departments of Epidemiology and Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital, Boston, Massachusetts; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, Microbiology/Immunology, UIC Cancer Center, University of Illinois at Chicago, Illinois.
| |
Collapse
|
24
|
Shojaei-Zarghani S, Yari Khosroushahi A, Rafraf M, Asghari-Jafarabadi M, Azami-Aghdash S. Dietary natural methylxanthines and colorectal cancer: a systematic review and meta-analysis. Food Funct 2020; 11:10290-10305. [DOI: 10.1039/d0fo02518f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Some evidence suggests that caffeine, theophylline, and theobromine, as natural methylxanthines (MTXs), possess anti-cancer effects.
Collapse
Affiliation(s)
- Sara Shojaei-Zarghani
- Student Research Committee
- Faculty of Nutrition and Food Science
- Tabriz University of Medical Sciences
- Tabriz
- Iran
| | - Ahmad Yari Khosroushahi
- Drug Applied Research Center
- Tabriz University of Medical Sciences
- Tabriz
- Iran
- Department of Medical Nanotechnology
| | - Maryam Rafraf
- Nutrition Research Center
- Department of Community Nutrition
- Faculty of Nutrition and Food Science
- Tabriz University of Medical Sciences
- Tabriz
| | | | - Saber Azami-Aghdash
- Faculty of Management and Medical Informatics
- Tabriz University of Medical Sciences
- Tabriz
- Iran
| |
Collapse
|
25
|
Wu J, Yu Y, Huang L, Li Z, Guo P, Xu YW. Dairy Product Consumption and Bladder Cancer Risk: A Meta-Analysis. Nutr Cancer 2019; 72:377-385. [PMID: 31295044 DOI: 10.1080/01635581.2019.1637909] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
To explore the potential relationship between dairy product consumption and bladder cancer risk, we retrieved eligible studies published up to March 15, 2018, via online database search and manual review of the selected articles. Summary relative risk (RR) estimates were calculated using random-effects models based on high to low intake values. Inter-study heterogeneity was explored using stratified analyses of study design, geographic region, or whether studies adjusted for the confounders age, sex, body mass index, smoking, and education level. We extracted data from 16 studies on milk (5,193 subjects) and 10 studies on dairy products (20,434 subjects). The total study population included 220,952 individuals. Dairy product intake and bladder cancer risk were significantly associated, and milk intake and bladder cancer risk more so. Stratified analysis revealed that the trend was more pronounced in case-control studies, and in studies with impact factor <3 and in Asia. The relationship was confirmed after adjusting for sex and Newcastle-Ottawa Scale score of 7 and 8. Our study shows an inverse association between milk consumption and bladder cancer risk.
Collapse
Affiliation(s)
- Jianhua Wu
- Department of Nephrology, Hui Ya Hospital of the First Affiliated Hospital, Sun Yat-Sen University, HuiZhou, China
| | - Yi Yu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Liqi Huang
- Department of Nephrology, Hui Ya Hospital of the First Affiliated Hospital, Sun Yat-Sen University, HuiZhou, China
| | - Zirui Li
- Department of Nephrology, Hui Ya Hospital of the First Affiliated Hospital, Sun Yat-Sen University, HuiZhou, China
| | - Penghui Guo
- Department of Nephrology, Hui Ya Hospital of the First Affiliated Hospital, Sun Yat-Sen University, HuiZhou, China
| | - Yuan Wen Xu
- Department of Nephrology, Hui Ya Hospital of the First Affiliated Hospital, Sun Yat-Sen University, HuiZhou, China
| |
Collapse
|
26
|
Singh J, Hussain Y, Luqman S, Meena A. Targeting Ca 2+ signalling through phytomolecules to combat cancer. Pharmacol Res 2019; 146:104282. [PMID: 31129179 DOI: 10.1016/j.phrs.2019.104282] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 05/10/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Cancer is amongst the life-threatening public health issue worldwide, hence responsible for millions of death every year. It is affecting human health regardless of their gender, age, eating habits, and ecological location. Many drugs and therapies are available for its cure still the need for effective targeted drugs and therapies are of paramount importance. In the recent past, Ca2+ signalling (including channels/transporters/pumps) are being studied as a plausible target for combating the cancer menace. Many evidence has shown that the intracellular Ca2+ homeostasis is altered in cancer cells and the remodelling is linked with tumor instigation, angiogenesis, progression, and metastasis. Focusing on these altered Ca2+ signalling tool kit for cancer treatment is a cross-cutting and emerging area of research. In addition, there are numerous phytomolecules which can be exploited as a potential Ca2+ (channels/transporters/ pumps) modulators in the context of targeting Ca2+ signalling in the cancer cell. In the present review, a list of plant-based potential Ca2+ (channel/transporters/pumps) modulators has been reported which could have application in the framework of repurposing the potential drugs to target Ca2+ signalling pathways in cancer cells. This review also aims to gain attention in and support for prospective research in this field.
Collapse
Affiliation(s)
- Jyoti Singh
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Jawaharlal Nehru University, New Delhi, 110067, India
| | - Yusuf Hussain
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Suaib Luqman
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India
| | - Abha Meena
- Molecular Bioprospection Department of Biotechnology Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, 226015, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, Uttar Pradesh, India.
| |
Collapse
|
27
|
Martini S, Conte A, Tagliazucchi D. Antiproliferative Activity and Cell Metabolism of Hydroxycinnamic Acids in Human Colon Adenocarcinoma Cell Lines. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3919-3931. [PMID: 30892877 DOI: 10.1021/acs.jafc.9b00522] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
In this study, we investigated the antiproliferative activity and the stability and metabolic fate of the main dietary hydroxycinnamates, using two colonic adenocarcinoma cell models (Caco-2 and SW480). Dihydrocaffeic and dihydroferulic acids were the most effective against cell proliferation in both cell lines with IC50 values of 71.7 ± 1.1 and 83.1 ± 1.1 μmol/L, respectively ( P < 0.05) in Caco-2. At 200 μmol/L, caffeic and ferulic acids inhibited SW480 proliferation by 40.8 ± 1.6 and 59.9 ± 1.3%, respectively. Hydroxycinnamic acids with a catechol-type structure were degraded in Caco-2 cell medium, resulting in the production of H2O2. Intracellular Caco-2 UDP-glucuronosyltransferases and catechol- O-methyltransferases were able to form glucuronide and methyl conjugates. However, only the sulfate conjugates were detected after incubation with SW480. In addition, simple hydroxycinnamates were released from quinic and aspartic conjugates. The remarkable effect of dihydrocaffeic and dihydroferulic acids against cell proliferation is of paramount importance, since these compounds are the main metabolites detectable at the colonic level.
Collapse
Affiliation(s)
- Serena Martini
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| | - Angela Conte
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| | - Davide Tagliazucchi
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Amendola 2 , 42100 Reggio Emilia , Italy
| |
Collapse
|
28
|
Moreno-Ceballos M, Arroyave JC, Cortes-Mancera FM, Röthlisberger S. Chemopreventive effect of coffee against colorectal cancer and hepatocellular carcinoma. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2019. [DOI: 10.1080/10942912.2019.1593193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Manuel Moreno-Ceballos
- Biomedical Innovation and Research Group, Faculty of Applied and Exact Sciences, Instituto Tecnologico Metropolitano, Medellin, Colombia
| | - Johanna C. Arroyave
- Biomedical Innovation and Research Group, Faculty of Applied and Exact Sciences, Instituto Tecnologico Metropolitano, Medellin, Colombia
| | - Fabian M. Cortes-Mancera
- Biomedical Innovation and Research Group, Faculty of Applied and Exact Sciences, Instituto Tecnologico Metropolitano, Medellin, Colombia
| | - Sarah Röthlisberger
- Biomedical Innovation and Research Group, Faculty of Applied and Exact Sciences, Instituto Tecnologico Metropolitano, Medellin, Colombia
| |
Collapse
|
29
|
Gregg JR, Lopez DS, Reichard C, Zheng J, Wu W, Ye Y, Chapin B, Kim J, Daniel CR, Davis J. Coffee, Caffeine Metabolism Genotype and Disease Progression in Patients with Localized Prostate Cancer Managed with Active Surveillance. J Urol 2019; 201:308-314. [PMID: 30179617 PMCID: PMC9798525 DOI: 10.1016/j.juro.2018.08.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Active surveillance is increasingly used as a management strategy for localized prostate cancer. Coffee intake has been associated with a lower prostate cancer incidence. We assessed whether coffee was associated with disease progression in men on active surveillance. MATERIALS AND METHODS A total of 411 patients with newly diagnosed Gleason score 6 or 7 prostate cancer were enrolled on a prospective active surveillance protocol for at least 6 months and completed a baseline dietary assessment. The active surveillance protocol included a biennial monitoring regimen with disease progression defined as an increase in the Gleason score. Cox proportional hazards models were used to evaluate associations of coffee intake with progression-free survival. We also evaluated patient genotype in the caffeine metabolism related single nucleotide polymorphism rs762551. RESULTS Median followup was 36 months (range 6 to 126) and the Gleason score progressed in 76 of the 411 patients (18.5%). Compared to 0 cups per day, in the multivariable model adjusting for prostate specific antigen, patient age and tumor length, less than 1 cup (HR 0.85, 95% CI 0.40-1.71), 1 to 1.9 cups (HR 0.64, 95% CI 0.29-1.43), 2 to 3.9 cups (HR 0.71, 95% CI 0.35-1.47) and 4 cups or more (HR 1.67, 95% CI 0.81-3.45) were not significantly associated with progression-free survival (p for nonlinearity = 0.01). Patients with low/moderate coffee intake and the AA fast caffeine metabolizer genotype were less likely to experience grade progression than nonconsumers (HR 0.36, 95% CI 0.15-0.88, p = 0.03). CONCLUSIONS Low to moderate coffee intake appears safe in men on active surveillance of localized prostate cancer. Further work is needed to determine whether high consumption is associated with shorter progression-free survival in sensitive groups.
Collapse
Affiliation(s)
| | | | | | - Jiali Zheng
- University of Texas MD Anderson Cancer Center
| | - Wenhui Wu
- University of Texas MD Anderson Cancer Center
| | - Yuanqing Ye
- University of Texas MD Anderson Cancer Center
| | | | - Jeri Kim
- University of Texas MD Anderson Cancer Center
| | | | - John Davis
- University of Texas MD Anderson Cancer Center
| |
Collapse
|
30
|
Thanikachalam K, Khan G. Colorectal Cancer and Nutrition. Nutrients 2019; 11:nu11010164. [PMID: 30646512 PMCID: PMC6357054 DOI: 10.3390/nu11010164] [Citation(s) in RCA: 418] [Impact Index Per Article: 83.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 01/10/2019] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
Colorectal Cancer is the third most common cancer diagnosed in the US. While the incidence and the mortality rate of colorectal cancer has decreased due to effective cancer screening measures, there has been an increase in number of young patients diagnosed in colon cancer due to unclear reasons at this point of time. While environmental and genetic factors play a major role in the pathogenesis of colon cancer, extensive research has suggested that nutrition may play both a causal and protective role in the development of colon cancer. In this review article, we aim to provide a review of factors that play a major role in development of colorectal cancer.
Collapse
Affiliation(s)
- Kannan Thanikachalam
- Department of Hematology/Oncology, Henry Ford Health System, Detroit, MI 48202, USA.
| | - Gazala Khan
- Department of Hematology/Oncology, Henry Ford Health System, Detroit, MI 48202, USA.
| |
Collapse
|
31
|
The Impact of Coffee and Its Selected Bioactive Compounds on the Development and Progression of Colorectal Cancer In Vivo and In Vitro. Molecules 2018; 23:molecules23123309. [PMID: 30551667 PMCID: PMC6321559 DOI: 10.3390/molecules23123309] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022] Open
Abstract
Coffee is one of the most popular beverages worldwide. Coffee contains bioactive compounds that affect the human body such as caffeine, caffeic acid, chlorogenic acids, trigonelline, diterpenes, and melanoidins. Some of them have demonstrated potential anticarcinogenic effects in animal models and in human cell cultures, and may play a protective role against colorectal cancer. Colorectal cancer (CRC) is the third leading cause of cancer-related mortality in the USA and other countries. Dietary patterns, as well as the consumption of beverages, may reduce the risk of CRC incidence. In this review, we focus on published epidemiological studies concerning the association of coffee consumption and the risk of development of colorectal cancer, and provide a description of selected biologically active compounds in coffee that have been investigated as potential cancer-combating compounds: Caffeine, caffeic acid (CA), chlorogenic acids (CGAs), and kahweol in relation to colorectal cancer progression in in vitro settings. We review the impact of these substances on proliferation, viability, invasiveness, and metastasis, as well as on susceptibility to chemo- and radiotherapy of colorectal cancer cell lines cultured in vitro.
Collapse
|
32
|
Preventing Lethal Prostate Cancer with Diet, Supplements, and Rx: Heart Healthy Continues to Be Prostate Healthy and "First Do No Harm" Part I. Curr Urol Rep 2018; 19:104. [PMID: 30368693 DOI: 10.1007/s11934-018-0846-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW To discuss the overall and latest observations of the effect of diet, lifestyle, supplements, and some prescription heart healthy medications for prostate cancer prevention. RECENT FINDINGS The concept of maximizing heart health to prevent aggressive prostate cancer continues to be solidified with the addition of more prospective observational and randomized controlled trial data. Heart healthy is prostate healthy, but heart unhealthy is prostate unhealthy. The primary goal of reducing the risk of all-cause and cardiovascular disease (CVD) morbidity and mortality also allows for maximizing prostate cancer prevention. The obesity epidemic in children and adults along with recent diverse research has only strengthened the nexus between heart and prostate health. Greater dietary adherence toward a variety of healthy foods is associated with a graded improved probability of CVD and potentially aggressive cancer risk reduction. Preventing prostate cancer via dietary supplements should encourage a "first do no harm", or less is more approach until future evidence can reverse the concerning trend that more supplementation has resulted in either no impact or an increased risk of prostate cancer. Supplements to reduce side effects of some cancer treatments appear to have more encouraging data. Medications that improve heart health including statins, aspirin, and metformin (S.A.M.), and specific beta-blocker medications are primarily generic or low-cost and should continue to garner research interest. A watershed moment in medical education has arrived where the past perception of a diverse number of trees seemingly separated by vast distances, in reality, now appear to exist within the same forest.
Collapse
|
33
|
Soares PV, Kannen V, Jordão Junior AA, Garcia SB. Coffee, but Neither Decaffeinated Coffee nor Caffeine, Elicits Chemoprotection Against a Direct Carcinogen in the Colon of Wistar Rats. Nutr Cancer 2018; 71:615-623. [PMID: 30362831 DOI: 10.1080/01635581.2018.1506489] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Colorectal cancer (CRC) is the third most frequent malignancy worldwide. Coffee is the second most consumed drink in the globe and suggested to decrease the CRC risk. Here, we explored whether coffee, decaffeinated coffee, or caffeine impact on the development of colorectal carcinogenesis induced by the direct carcinogen N-methyl-N-nitro-N-nitrosoguanidine (MNNG) in rats. To this end, sixty-four young male Wistar rats were divided into eight groups of eight animals each. We analyzed the frequency of dysplastic crypts and expression of metallothionein as a biomarker of the cancer risk, as well the expression of phosphorylated H2A histone family/member X (γH2AX) for DNA damage and cyclooxygenase-2 (COX-2) for inflammatory response. We also studied the oxidative stress profile in hepatic and colonic frozen samples (malondialdehyde [MDA], glutathione [GSH], and α-tocopherol). We found that coffee but neither decaffeinated coffee nor caffeine decreased the development of dysplastic crypts in MNNG-exposed rats. All treatments reduced DNA damage intensity in colonocytes. Only decaffeinated coffee increased the numbers of metallothionein positive crypts in comparison with coffee-treated rats. Coffee and caffeine inhibited COX-2 expression in the colon. Both decaffeinated coffee and caffeine decreased hepatic α-tocopherol levels. We suggest that coffee may have other compounds that elicit greater chemoprotective effects than caffeine reducing the CRC risk.
Collapse
Affiliation(s)
| | - Vinicius Kannen
- b Department of Toxicology, Bromatology, and Clinical Analysis , University of Sao Paulo , Ribeirao Preto , Brazil
| | | | | |
Collapse
|
34
|
Song M, Chan AT. The Potential Role of Exercise and Nutrition in Harnessing the Immune System to Improve Colorectal Cancer Survival. Gastroenterology 2018; 155:596-600. [PMID: 30076837 PMCID: PMC6571501 DOI: 10.1053/j.gastro.2018.07.038] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Mingyang Song
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, and Harvard Medical School, Boston, MA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
35
|
Guercio BJ, Zhang S, Niedzwiecki D, Li Y, Babic A, Morales-Oyarvide V, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Zoltick ES, Stampfer M, Ng K, Wu K, Willett WC, Giovannucci EL, Meyerhardt JA, Fuchs CS. Associations of artificially sweetened beverage intake with disease recurrence and mortality in stage III colon cancer: Results from CALGB 89803 (Alliance). PLoS One 2018; 13:e0199244. [PMID: 30024889 PMCID: PMC6053135 DOI: 10.1371/journal.pone.0199244] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 06/01/2018] [Indexed: 12/16/2022] Open
Abstract
Purpose Observational studies have demonstrated increased colon cancer recurrence and mortality in states of excess energy balance, as denoted by factors including sedentary lifestyle, diabetes, increased dietary glycemic load, and increased intake of sugar-sweetened beverages. Nonetheless, the relation between artificially sweetened beverages, a popular alternative for sugar-sweetened beverages, and colon cancer recurrence and survival is unknown. Methods We analyzed data from 1,018 patients with stage III colon cancer who prospectively reported dietary intake during and after chemotherapy while enrolled in a National Cancer Institute-sponsored trial of adjuvant chemotherapy. Using Cox proportional hazards regressions, we assessed associations of artificially sweetened beverage intake with cancer recurrence and mortality. Results Patients consuming one or more 12-ounce servings of artificially sweetened beverages per day experienced an adjusted hazard ratio for cancer recurrence or mortality of 0.54 (95% confidence interval, 0.36 to 0.80) when compared to those who largely abstained (Ptrend = .004). Similarly, increasing artificially sweetened beverage intake was also associated with a significant improvement in both recurrence-free survival (Ptrend = .005) and overall survival (Ptrend = .02). Substitution models demonstrated that replacing a 12-ounce serving of a sugar-sweetened beverage with an isovolumetric serving of an artificially sweetened beverage per day was associated with a 23% lower risk of cancer recurrence and mortality (relative risk, 0.77; 95% confidence interval, 0.63 to 0.95; P = .02). Conclusion Higher artificially sweetened beverage consumption may be associated with significantly reduced cancer recurrence and death in patients with stage III colon cancer. This association may be mediated by substitution for sugar-sweetened alternatives. Further studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Brendan J. Guercio
- Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Donna Niedzwiecki
- Alliance Statistics and Data Center, Duke University, Durham, North Carolina, United States of America
| | - Yanping Li
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Ana Babic
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | | | - Leonard B. Saltz
- Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Robert J. Mayer
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Rex B. Mowat
- Toledo Community Hospital Oncology Program, Toledo, Ohio, United States of America
| | | | - Alexander Hantel
- Edward-Elmhurst Healthcare, Naperville, Illinois, United States of America
| | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, United States of America
| | - Daniel Atienza
- Virginia Oncology Associates, Norfolk, Virginia, United States of America
| | - Michael Messino
- Southeast Clinical Oncology Research (SCOR) Consortium, Mission Hospitals, Incorporated, Asheville, North Carolina, United States of America
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer Center, Chicago, Illinois, United States of America
| | - Alan Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, California, United States of America
| | - Shuji Ogino
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Division of Molecular Pathological Epidemiology (MPE), Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
- Department of Pathology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Emilie S. Zoltick
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, United States of America
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Meir Stampfer
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimmie Ng
- Dana-Farber/Partners CancerCare, Boston, Massachusetts, United States of America
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Walter C. Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Edward L. Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Charles S. Fuchs
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, United States of America
- * E-mail:
| |
Collapse
|
36
|
Brown JC, Zhang S, Niedzwiecki D, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson A, Atienza D, Messino M, Kindler H, Venook A, Ogino S, Li Y, Zhang X, Ng K, Willett WC, Giovannucci EL, Fuchs CS, Meyerhardt JA. Grain Intake and Clinical Outcome in Stage III Colon Cancer: Results From CALGB 89803 (Alliance). JNCI Cancer Spectr 2018; 2:pky017. [PMID: 29877501 PMCID: PMC5977856 DOI: 10.1093/jncics/pky017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 12/15/2022] Open
Abstract
Background Energy balance–related risk factors for colon cancer recurrence and mortality—type II diabetes, hyperinsulinemia, inflammation, and visceral obesity—are positively correlated with consumption of refined grains and negatively correlated with consumption of whole grains. We examined the relationship between the consumption of refined and whole grains with cancer recurrence and mortality in a cohort of patients with colon cancer. Methods We conducted a prospective observational study of 1024 patients with stage III colon cancer who participated in a randomized trial of postoperative chemotherapy. Patients reported consumption of refined and whole grains using a food frequency questionnaire during and six months after chemotherapy. The primary outcome was disease-free survival (DFS). Multivariable-adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) were estimated using Cox regression models. All P values are two-sided. Results During a median follow-up of 7.3 years, 394 patients experienced a DFS event. The hazard ratio for DFS was 1.56 (95% CI = 1.09 to 2.24) for patients consuming three or more servings per day of refined grains compared with patients consuming less than one serving per day (Ptrend = .005). The hazard ratio for DFS was 0.89 (95% CI = 0.66 to 1.20) for patients consuming three or more servings per day of whole grains compared with patients consuming less than one serving per day (Ptrend = .54). The hazard ratio for DFS of substituting one serving per day of refined grain with one serving per day of whole grain was 0.87 (95% CI = 0.79 to 0.96, P = .007). Conclusions The choice of grain consumed may be associated with cancer recurrence and mortality. Future studies are necessary to confirm our findings and to inform the design of randomized trials.
Collapse
Affiliation(s)
| | - Sui Zhang
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | | | | | - Rex B Mowat
- Toledo Community Hospital Oncology Program, Toledo, OH
| | | | | | - Al Benson
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL
| | | | - Michael Messino
- Southeast Clinical Oncology Research (SCOR) Consortium, Mission Hospitals, Inc., Asheville, NC
| | - Hedy Kindler
- University of Chicago Comprehensive Cancer, Chicago, IL
| | - Alan Venook
- University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA
| | - Shuji Ogino
- Dana-Farber/Partners CancerCare, Boston, MA.,Harvard T.H. Chan School of Public Health, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | - Yanping Li
- Harvard T.H. Chan School of Public Health, Boston, MA
| | | | - Kimmie Ng
- Dana-Farber/Partners CancerCare, Boston, MA
| | | | - Edward L Giovannucci
- Harvard T.H. Chan School of Public Health, Boston, MA.,Brigham and Women's Hospital, Boston, MA
| | | | | |
Collapse
|
37
|
Zhan X, Wang J, Pan S, Lu C. Tea consumption and the risk of ovarian cancer: A meta-analysis of epidemiological studies. Oncotarget 2018; 8:37796-37806. [PMID: 28445129 PMCID: PMC5514950 DOI: 10.18632/oncotarget.16890] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 03/27/2017] [Indexed: 01/06/2023] Open
Abstract
A large number of epidemiological studies have provided conflicting results about the relationship between tea consumption and ovarian cancer. This study aimed to clarify the association between tea consumption and ovarian cancer. A literature search of the MEDICINE, Scopus, PubMed, and Web of Science databases was performed in April 2016. A total of 18 (11 case-control and 7 cohort) studies, representing data for 701,857 female subjects including 8,683 ovarian cancer cases, were included in the meta-analysis. A random-effects meta-analysis was used to compute the pooled relative risks (RR), meta regression, and publication bias, and heterogeneity analyses were performed for the included trials. We found that tea consumption had a significant protective effect against ovarian cancer (relative risk [RR] = 0.86; 95% confidence interval [CI]: 0.76, 0.96). The relationship was confirmed particularly after adjusting for family history of cancer (RR = 0.85; 95% CI: 0.72, 0.97), menopause status (RR = 0.85; 95% CI: 0.72, 0.98), education (RR = 0.82; 95% CI: 0.68, 0.96), BMI (RR = 0.85; 95% CI: 0.70, 1.00), smoking (RR = 0.83; 95% CI: 0.72, 0.93) and Jadad score of 3 (RR = 0.76; 95% CI: 0.56, 0.95) and 5 (RR = 0.74; 95% CI: 0.59, 0.89). The Begg's and Egger's tests (all P > 0.01) showed no evidence of publication bias. In conclusion, our meta-analysis showed an inverse association between tea consumption and ovarian cancer risk. High quality cohort-clinical trials should be conducted on different tea types and their relationship with ovarian cancer.
Collapse
Affiliation(s)
- Xin Zhan
- Obstetrics and Gynecology Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, PR China
| | - Jie Wang
- Obstetrics and Gynecology Department, The First Affiliated Hospital of Zhejiang Chinese Medical University Zhejiang Provincial Hospital of TCM, Hangzhou, Zhejiang 310016, PR China
| | - Shufen Pan
- Department of Obstetrics and Gynecology, Central Hospital of Wenzhou, Luchengqu, Wenzhou, Zhejiang 325000, PR China
| | - Caijuan Lu
- Obstetrics and Gynecology Department, Hangzhou First People's Hospital, Hangzhou, Zhejiang 310006, PR China
| |
Collapse
|
38
|
Hu Y, Ding M, Yuan C, Wu K, Smith-Warner SA, Hu FB, Chan AT, Meyerhardt JA, Ogino S, Fuchs CS, Giovannucci EL, Song M. Association Between Coffee Intake After Diagnosis of Colorectal Cancer and Reduced Mortality. Gastroenterology 2018; 154:916-926.e9. [PMID: 29158191 PMCID: PMC5847429 DOI: 10.1053/j.gastro.2017.11.010] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/04/2017] [Accepted: 11/13/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Few studies have examined the association between coffee intake and survival after diagnosis of colorectal cancer (CRC). We performed a prospective study to investigate the association between coffee intake after a diagnosis of CRC and mortality. METHODS We collected data from the Nurses' Health Study (1984-2012) and Health Professionals Follow-up Study (1986-2012), following 1599 patients diagnosed with stage 1, 2, or 3 CRC. CRC was reported on questionnaires and ascertained by review of medical records and pathology reports; intake of food and beverages was determined from responses to semi-quantitative food frequency questionnaires. Participants were asked how often during the previous year that they consumed coffee, with 1 cup as the standard portion size. The first questionnaire response collected at least 6 months but not more than 4 years after diagnosis was used for assessment of post-diagnostic intake (median time from diagnosis to the dietary assessment, 2.2 years). The last semi-quantitative food frequency questionnaire prior to diagnosis was used to assess pre-diagnostic dietary intake. RESULTS During a median of 7.8 years of follow-up, we documented 803 deaths, of which 188 were because of CRC. In the multivariable adjusted models, compared with nondrinkers, patients who consumed at least 4 cups of coffee per day had a 52% lower risk of CRC-specific death (hazard ratio [HR] 0.48; 95% CI, 0.28-0.83; P for trend=.003) and 30% reduced risk of all-cause death (HR, 0.70; 95% CI, 0.54-0.91; P for trend <.001). High intake of caffeinated and decaffeinated coffee (2 or more cups/day) was associated with lower risk of CRC-specific mortality and all-cause mortality. When coffee intake before vs after CRC diagnosis were examined, compared with patients consistently consuming low amounts (less than 2 cups/day), those who maintained a high intake (2 or more cups/day) had a significantly lower risk of CRC-specific death (multivariable HR, 0.63; 95% CI, 0.44-0.89) and death from any cause (multivariable HR, 0.71; 95% CI, 0.60-0.85). CONCLUSIONS In an analysis data from the Nurses' Health Study and Health Professionals Follow-up Study, we associated intake of caffeinated and decaffeinated coffee after diagnosis of CRC with lower risk of CRC-specific death and overall death. Studies are needed to determine the mechanisms by which coffee might reduce CRC progression.
Collapse
Affiliation(s)
- Yang Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Ming Ding
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Chen Yuan
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Stephanie A Smith-Warner
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrew T Chan
- Channing Division of Network Medicine, Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, Massachusetts
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Charles S Fuchs
- Yale Cancer Center, New Haven, Connecticut; Department of Medicine, Yale School of Medicine, New Haven, Connecticut; Smilow Cancer Hospital, New Haven, Connecticut
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Channing Division of Network Medicine, Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts; Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
39
|
Affiliation(s)
| | - Marc J Gunter
- International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
40
|
Fadelu T, Zhang S, Niedzwiecki D, Ye X, Saltz LB, Mayer RJ, Mowat RB, Whittom R, Hantel A, Benson AB, Atienza DM, Messino M, Kindler HL, Venook A, Ogino S, Ng K, Wu K, Willett W, Giovannucci E, Meyerhardt J, Bao Y, Fuchs CS. Nut Consumption and Survival in Patients With Stage III Colon Cancer: Results From CALGB 89803 (Alliance). J Clin Oncol 2018; 36:1112-1120. [PMID: 29489429 PMCID: PMC5891130 DOI: 10.1200/jco.2017.75.5413] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Purpose Observational studies have reported increased colon cancer recurrence and mortality in patients with states of hyperinsulinemia, including type 2 diabetes, obesity, sedentary lifestyle, and high glycemic load diet. Nut intake has been associated with a lower risk of type 2 diabetes, metabolic syndrome, and insulin resistance. However, the effect of nut intake on colon cancer recurrence and survival is not known. Patients and Methods We conducted a prospective, observational study of 826 eligible patients with stage III colon cancer who reported dietary intake on food frequency questionnaires while enrolled onto a randomized adjuvant chemotherapy trial. Using Cox proportional hazards regression, we assessed associations of nut intake with cancer recurrence and mortality. Results After a median follow-up of 6.5 years, compared with patients who abstained from nuts, individuals who consumed two or more servings of nuts per week experienced an adjusted hazard ratio (HR) for disease-free survival of 0.58 (95% CI, 0.37 to 0.92; Ptrend = .03) and an HR for overall survival of 0.43 (95% CI, 0.25 to 0.74; Ptrend = .01). In subgroup analysis, the apparent benefit was confined to tree nut intake (HR for disease-free survival, 0.54; 95% CI, 0.34 to 0.85; Ptrend = .04; and HR for overall survival, 0.47; 95% CI, 0.27 to 0.82; Ptrend = .04). The association of total nut intake with improved outcomes was maintained across other known or suspected risk factors for cancer recurrence and mortality. Conclusion Diets with a higher consumption of nuts may be associated with a significantly reduced incidence of cancer recurrence and death in patients with stage III colon cancer.
Collapse
Affiliation(s)
- Temidayo Fadelu
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Sui Zhang
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Donna Niedzwiecki
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Xing Ye
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Leonard B Saltz
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Robert J Mayer
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Rex B Mowat
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Renaud Whittom
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Alexander Hantel
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Al B Benson
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Daniel M Atienza
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Michael Messino
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Hedy L Kindler
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Alan Venook
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Shuji Ogino
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Kimmie Ng
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Kana Wu
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Walter Willett
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Edward Giovannucci
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Jeffrey Meyerhardt
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Ying Bao
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| | - Charles S Fuchs
- Temidayo Fadelu, Sui Zhang, Robert J. Mayer, Shuji Ogino, Kimmie Ng, Jeffrey Meyerhardt, and Charles S. Fuchs, Dana-Farber/Partners CancerCare; Shuji Ogino, Kana Wu, Walter Willett, and Edward Giovannucci, Harvard TH Chan School of Public Health; Shuji Ogino, Edward Giovannucci, and Ying Bao, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA; Donna Niedzwiecki and Xing Ye, Alliance Statistics and Data Center, Duke University, Durham; Michael Messino, Southeast Clinical Oncology Research Consortium, Mission Hospitals, Asheville, NC; Leonard B. Saltz, Memorial Sloan Kettering Cancer Center, New York, NY; Rex B. Mowat, Toledo Community Hospital Oncology Program, Toledo, OH; Renaud Whittom, Hôpital du Sacré-Coeur de Montréal, Montreal, Quebec, Canada; Alexander Hantel, Loyola University Stritch School of Medicine, Naperville; Al B. Benson, Robert H Lurie Comprehensive Cancer Center, Northwestern University; Hedy L. Kindler, University of Chicago Comprehensive Cancer, Chicago, IL; Daniel M. Atienza, Virginia Oncology Associates, Norfolk, VA; Alan Venook, University of California at San Francisco Comprehensive Cancer Center, San Francisco, CA; and Charles S. Fuchs, Yale Cancer Center, Yale School of Medicine, New Haven, CT
| |
Collapse
|
41
|
Abstract
Diet, physical activity, and body weight have been shown to play an important role in cancer survivorship. The impact of each of these lifestyle factors differs slightly among cancer types, and adherence to recommended diet and physical activity guidelines has been associated with positive outcomes, including decrease in the risk of cancer recurrence and improvement of quality of life. Although there are compelling data that appropriate diet, physical activity, and body weight have beneficial effects in cancer survivorship, additional trials are needed to understand the relationship.
Collapse
Affiliation(s)
- Karishma Mehra
- Department of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Alyssa Berkowitz
- Department of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA
| | - Tara Sanft
- Department of Medical Oncology, Yale School of Medicine, 333 Cedar Street, New Haven, CT 06520, USA.
| |
Collapse
|
42
|
van Zutphen M, Kampman E, Giovannucci EL, van Duijnhoven FJB. Lifestyle after Colorectal Cancer Diagnosis in Relation to Survival and Recurrence: A Review of the Literature. CURRENT COLORECTAL CANCER REPORTS 2017; 13:370-401. [PMID: 29104517 PMCID: PMC5658451 DOI: 10.1007/s11888-017-0386-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW This review summarizes the evidence regarding diet, physical activity, smoking, and body composition after colorectal cancer (CRC) diagnosis in relation to all-cause and CRC-specific mortality and disease recurrence and gives suggestions for future research directions. RECENT FINDINGS Overall, this review suggests that some, albeit not all, of the well-known modifiable risk factors for cancer incidence might also be associated with CRC survival. CRC prognosis appears to be worse with increased physical inactivity, smoking, or being underweight after CRC diagnosis. Emerging evidence suggests that diets associated with a positive energy balance, e.g., high consumption of sugar-sweetened beverages, may negatively impact survival in CRC survivors. In contrast, there is currently little evidence to support the recommendation to limit red and processed meat or alcohol intake after CRC diagnosis. Whether being overweight and obese after CRC diagnosis improves or worsens CRC prognosis remains controversial and may depend on the measure used to assess body fatness. SUMMARY Further research on post-diagnosis lifestyle patterns is needed to understand the multifactorial influence on CRC prognosis. Disease recurrence and the development of comorbidities should be included as key outcomes in future studies and lifestyle should preferably be repeatedly measured.
Collapse
Affiliation(s)
- Moniek van Zutphen
- Division of Human Nutrition, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Ellen Kampman
- Division of Human Nutrition, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| | - Edward L. Giovannucci
- Department of Nutrition, Department of Epidemiology, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Bldg. 2, Room 371, Boston, MA 02115 USA
| | - Fränzel J. B. van Duijnhoven
- Division of Human Nutrition, Wageningen University and Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands
| |
Collapse
|
43
|
Tsujimoto T, Kajio H, Sugiyama T. Association Between Caffeine Intake and All-Cause and Cause-Specific Mortality: A Population-Based Prospective Cohort Study. Mayo Clin Proc 2017; 92:1190-1202. [PMID: 28697850 DOI: 10.1016/j.mayocp.2017.03.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/17/2017] [Accepted: 03/07/2017] [Indexed: 11/17/2022]
Abstract
OBJECTIVE To assess whether caffeine intake is associated with all-cause and cause-specific mortality. PATIENTS AND METHODS We conducted a prospective cohort study using data from the National Health and Nutrition Examination Survey 1999-2010. Cox proportional hazards models were used to compare the multivariate-adjusted hazard ratios (HRs) of participants with a caffeine intake of 10 to 99, 100 to 199, and 200 mg/d or more with those of participants with a caffeine intake of less than 10 mg/d. RESULTS In total, 17,594 participants were included, and the mean ± SD and median (interquartile range) follow-up was 6.5±2.8 years and 6.4 (3.6-9.5) years, respectively; 17,568 participants (99.8%) completed the follow-up, and 1310 died. Compared with those who had a caffeine intake of less than 10 mg/d, HRs and 95% CIs for all-cause mortality were significantly lower in participants with a caffeine intake of 10 to 99 mg/d (HR, 0.81; 95% CI, 0.66-1.00; P=.05), 100 to 199 mg/d (HR, 0.63; 95% CI, 0.51-0.78; P<.001), and 200 or more mg/d (HR, 0.69; 95% CI, 0.58-0.83; P<.001). A similar association was observed in participants who consumed less than 1 cup of coffee per week, and the HR was lowest in those with a caffeine intake of 100 to 199 mg/d (HR, 0.46; 95% CI, 0.22-0.93). There was no association between caffeine intake and cardiovascular mortality, whereas the HRs for noncardiovascular mortality were significantly lower in those with a caffeine intake of 10 to 99 mg/d (HR, 0.74; 95% CI, 0.57-0.95; P=.01), 100 to 199 mg/d (HR, 0.60; 95% CI, 0.46-0.77; P<.001), and 200 or more mg/d (HR, 0.65; 95% CI, 0.53-0.80; P<.001). CONCLUSION Moderate caffeine intake was associated with a decreased risk of all-cause mortality, regardless of the presence or absence of coffee consumption.
Collapse
Affiliation(s)
- Tetsuro Tsujimoto
- Department of Diabetes, Endocrinology, and Metabolism, Center Hospital, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Hiroshi Kajio
- Department of Diabetes, Endocrinology, and Metabolism, Center Hospital, National Center for Global Health and Medicine, Tokyo, Japan
| | - Takehiro Sugiyama
- Department of Clinical Study and Informatics, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan; Department of Public Health/Health Policy, the University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Liu H, Zhou Y, Tang L. Caffeine induces sustained apoptosis of human gastric cancer cells by activating the caspase‑9/caspase‑3 signalling pathway. Mol Med Rep 2017; 16:2445-2454. [PMID: 28677810 PMCID: PMC5547974 DOI: 10.3892/mmr.2017.6894] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 04/25/2017] [Indexed: 12/21/2022] Open
Abstract
Caffeine is one of the most widely consumed substances found in beverages, and has demonstrated anticancer effects in several types of cancer. The present study aimed to examine the anticancer effects of caffeine on gastric cancer (GC) cells (MGC‑803 and SGC‑7901) in vitro, and to determine whether the apoptosis‑related caspase‑9/-3 pathway is associated with these effects. The sustained antiproliferative effects of caffeine on gastric cancer were also investigated. GC cell viability and proliferation were evaluated using cell counting and colony forming assays, following treatment with various concentrations of caffeine. Flow cytometry was performed to assess cell cycle dynamics and apoptosis. Western blot analysis was conducted to detect the activity of the caspase‑9/-3 pathway. The results indicated that caffeine treatment significantly suppressed GC cell growth and viability and induced apoptosis by activating the caspase‑9/-3 pathway. Furthermore, the anticancer effects of caffeine appeared to be sustained, as the caspase‑9/-3 pathway remained active following caffeine withdrawal. In conclusion, caffeine may function as a sustained anticancer agent by activating the caspase‑9/-3 pathway, which indicates that it may be useful as a therapeutic candidate in gastric cancer.
Collapse
Affiliation(s)
- Hanyang Liu
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| | - Yan Zhou
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| | - Liming Tang
- Department of Gastrointestinal Surgery, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou, Jiangsu 213000, P.R. China
| |
Collapse
|
45
|
Amigo-Benavent M, Wang S, Mateos R, Sarriá B, Bravo L. Antiproliferative and cytotoxic effects of green coffee and yerba mate extracts, their main hydroxycinnamic acids, methylxanthine and metabolites in different human cell lines. Food Chem Toxicol 2017; 106:125-138. [PMID: 28506698 DOI: 10.1016/j.fct.2017.05.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 04/28/2017] [Accepted: 05/10/2017] [Indexed: 12/18/2022]
Abstract
This work aimed at studying the effects of green coffee bean (GCBE) and yerba mate (YME) extracts, their main phenolic components (5-caffeoylquinic acid, 5-CQA; 3,5-dicaffeoylquinic acid, 3,5-DCQA) and metabolites (ferulic acid, FA; caffeic acid, CA; dihydrocaffeic acid, DHCA; and dihydroferulic acid, DHFA) along with caffeine (CAF) on the viability and proliferation of different human cell lines. Extracts (10-1000 μg/mL) and standards (10-1000 μM) were assayed in colon (Caco-2), lung (A549), oesophageal (OE-33), urinary bladder (T24) human carcinoma cells, and a non-cancer cell line (CCD-18Co). YME significantly reduced viability of cancer cells at all assayed concentrations, the higher doses also reducing cell proliferation. GCBE effects on cell viability were more effective at 100 and 1000 μg/mL, showing modest effects on cell proliferation. The highest doses of 5-CQA and 3,5-DCQA reduced cell viability and proliferation in all cell lines, whereas FA, DHCA and DHFA had lower and variable effects. Caffeine had no effect. Dietary-attainable concentrations (0.1, 1 and 10 μg/mL) of YME were tested for cytotoxicity and reactive oxygen species generation, showing no cytotoxic effect. Low concentrations of all tested compounds were non-cytotoxic to CCD-18Co cells. CONCLUSION YME and to a lower degree GCBE, their phenolic components and metabolites may decrease cancer cell viability and proliferation.
Collapse
Affiliation(s)
- M Amigo-Benavent
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/ Jose Antonio Nováis 10, 28040 Madrid, Spain
| | - S Wang
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/ Jose Antonio Nováis 10, 28040 Madrid, Spain
| | - R Mateos
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/ Jose Antonio Nováis 10, 28040 Madrid, Spain
| | - B Sarriá
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/ Jose Antonio Nováis 10, 28040 Madrid, Spain.
| | - L Bravo
- Institute of Food Science, Technology and Nutrition (ICTAN), Spanish National Research Council (CSIC), C/ Jose Antonio Nováis 10, 28040 Madrid, Spain.
| |
Collapse
|
46
|
Schmit SL, Rennert HS, Rennert G, Gruber SB. Coffee Consumption and the Risk of Colorectal Cancer. Cancer Epidemiol Biomarkers Prev 2017; 25:634-9. [PMID: 27196095 DOI: 10.1158/1055-9965.epi-15-0924] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/18/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Coffee contains several bioactive compounds relevant to colon physiology. Although coffee intake is a proposed protective factor for colorectal cancer, current evidence remains inconclusive. METHODS We investigated the association between coffee consumption and risk of colorectal cancer in 5,145 cases and 4,097 controls from the Molecular Epidemiology of Colorectal Cancer (MECC) study, a population-based case-control study in northern Israel. We also examined this association by type of coffee, by cancer site (colon and rectum), and by ethnic subgroup (Ashkenazi Jews, Sephardi Jews, and Arabs). Coffee data were collected by interview using a validated, semi-quantitative food frequency questionnaire. RESULTS Coffee consumption was associated with 26% lower odds of developing colorectal cancer [OR (drinkers vs. non-drinkers), 0.74; 95% confidence interval (CI), 0.64-0.86; P < 0.001]. The inverse association was also observed for decaffeinated coffee consumption alone (OR, 0.82; 95% CI, 0.68-0.99; P = 0.04) and for boiled coffee (OR, 0.82; 95% CI, 0.71-0.94; P = 0.004). Increasing consumption of coffee was associated with lower odds of developing colorectal cancer. Compared with <1 serving/day, intake of 1 to <2 servings/day (OR, 0.78; 95% CI, 0.68-0.90; P < 0.001), 2 to 2.5 servings/day (OR, 0.59; 95% CI, 0.51-0.68; P < 0.001), and >2.5 servings/day (OR, 0.46; 95% CI, 0.39-0.54; P < 0.001) were associated with significantly lower odds of colorectal cancer (Ptrend < 0.001), and the dose-response trend was statistically significant for both colon and rectal cancers. CONCLUSIONS Coffee consumption may be inversely associated with risk of colorectal cancer in a dose-response manner. IMPACT Global coffee consumption patterns suggest potential health benefits of the beverage for reducing the risk of colorectal cancer. Cancer Epidemiol Biomarkers Prev; 25(4); 634-9. ©2016 AACR.
Collapse
Affiliation(s)
- Stephanie L Schmit
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California. Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Hedy S Rennert
- Department of Community Medicine and Epidemiology, Carmel Medical Center, Haifa, Israel
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Carmel Medical Center, Haifa, Israel. Clalit Health Services National Cancer Control Center, Haifa, Israel. Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California. Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California.
| |
Collapse
|
47
|
Abstract
Colorectal cancer is one of the so-called westernized diseases and the second leading cause of cancer death worldwide. On the basis of global epidemiological and scientific studies, evidence suggests that the risk of colorectal cancer is increased by processed and unprocessed meat consumption but suppressed by fibre, and that food composition affects colonic health and cancer risk via its effects on colonic microbial metabolism. The gut microbiota can ferment complex dietary residues that are resistant to digestion by enteric enzymes. This process provides energy for the microbiota but culminates in the release of short-chain fatty acids including butyrate, which are utilized for the metabolic needs of the colon and the body. Butyrate has a remarkable array of colonic health-promoting and antineoplastic properties: it is the preferred energy source for colonocytes, it maintains mucosal integrity and it suppresses inflammation and carcinogenesis through effects on immunity, gene expression and epigenetic modulation. Protein residues and fat-stimulated bile acids are also metabolized by the microbiota to inflammatory and/or carcinogenic metabolites, which increase the risk of neoplastic progression. This Review will discuss the mechanisms behind these microbial metabolite effects, which could be modified by diet to achieve the objective of preventing colorectal cancer in Western societies.
Collapse
|