1
|
Sauter ER, Butera G, Agurs-Collins T. Long-Term Randomized Controlled Trials of Diet Intervention Reports and Their Impact on Cancer: A Systematic Review. Cancers (Basel) 2024; 16:3296. [PMID: 39409915 PMCID: PMC11475623 DOI: 10.3390/cancers16193296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Most randomized controlled trials (RCTs) assessing the impact of diet on cancer have been short term (<1 year), mostly evaluating breast cancer survivors. Given the many-year interval that is generally required for an intervention to have an impact on cancer risk or prognosis, as well as the fact that lifestyle strategies such as diet modification frequently fail due to lack of adherence over the long term, we focused this systematic review only on longer-term (≥1 year) intervention reports. Diet intervention reports focused on reducing cancer risk in overweight and obese individuals target caloric restriction (every day, some days, or most hours of each day). METHODS This study is a systematic review of RCTs lasting at least 1 year, testing dietary interventions with a primary or secondary endpoint of cancer or a biomarker linked to cancer. RESULTS Fifty-one reports met our review criteria. Twenty of fifty-one (39%) reports are RCTs where the primary endpoint was cancer or a cancer-related biomarker, while the other reports evaluated reports where cancer or a cancer-related biomarker was a secondary endpoint. Thirteen of twenty (65%) primary reports evaluated isocaloric, and the remaining eight evaluated low-calorie diets. All but one of the primary and two secondary isocaloric diet reports evaluated the benefit of a low-fat diet (LFD), with the other three evaluating a Mediterranean diet (MedD). More LCD vs. isocaloric diet primary reports (71% vs. 38%) demonstrated cancer or cancer-related biomarker benefit; the difference in chance of benefit with secondary reports was 85% for LCD vs. 73% for isocaloric diets. Three of three MedD reports demonstrated benefit. Sixty-nine percent (20/29) of the secondary reports came from two large reports: the WHI diet modification trial (15 secondary reports) and the polyp prevention trial (5 secondary reports). Nineteen of twenty-two (86%) primary reports enrolled only women, and three enrolled both men and women. No study that met our criteria enrolled only men, comprising 1447 men in total vs. 62,054 women. Fifteen of twenty (75%) primary reports focus on healthy women or women with breast cancer. Adherence findings are discussed when provided. CONCLUSIONS More long-term RCTs evaluating cancer and cancer-related biomarker endpoints are needed, especially for cancers at sites other than the breast.
Collapse
Affiliation(s)
- Edward R. Sauter
- Division of Cancer Prevention, National Cancer Institute/National Institutes of Health (NIH), 9609 Medical Center Drive, Rockville, MD 20850, USA
| | - Gisela Butera
- Office of Research Services, NIH Library, Bethesda, MD 20892, USA;
| | - Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute/National Institutes of Health (NIH), 9609 Medical Center Drive, Rockville, MD 20850, USA;
| |
Collapse
|
2
|
Di Maso M, Augustin LSA, Jenkins DJA, Crispo A, Toffolutti F, Negri E, La Vecchia C, Ferraroni M, Polesel J. Adherence to a Cholesterol-Lowering Diet and the Risk of Pancreatic Cancer: A Case-Control Study. Nutrients 2024; 16:2508. [PMID: 39125388 PMCID: PMC11314520 DOI: 10.3390/nu16152508] [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: 06/13/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Pancreatic cancer risk has been associated with increased serum cholesterol level, which is in turn partially influenced by diet. This study aimed at evaluating the association between pancreatic cancer risk and the adherence to a plant-based cholesterol-lowering diet. METHODS Data were derived from an Italian case-control study including 258 pancreatic cancer patients and 551 controls. The cholesterol-lowering diet score was based on seven components: high intakes of (i) non-cellulosic polysaccharides (a proxy of viscous fibers), (ii) monounsaturated fatty acids, (iii) legumes, and (iv) seeds/corn oils (a proxy of phytosterols); and low intakes of (v) saturated fatty acids, (vi) dietary cholesterol, and (vii) food with a high glycemic index. The score was calculated adding one point for each fulfilled component, thus ranging from zero (no adherence) to seven (complete adherence). The odds ratios (ORs) and 95% confidence intervals (CIs) were estimated through the logistic regression model. RESULTS Scores 5-7 were associated with reduced cancer risk (OR = 0.30; 95% CI: 0.18-0.52) compared to scores 0-2. CONCLUSIONS Adherence to a plant-based cholesterol-lowering diet was associated with a reduced risk of pancreatic cancer.
Collapse
Affiliation(s)
- Matteo Di Maso
- Department of Clinical Sciences and Community Health, Department of Excellence 2023–2027, Branch of Medical Statistics, Biometry and Epidemiology “G.A. Maccacaro”, Università degli Studi di Milano, via Celoria 22, 20133 Milan, Italy; (M.D.M.); (M.F.)
| | - Livia S. A. Augustin
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori—IRCCS—“Fondazione G. Pascale”, via M. Semmola 1, 80131 Naples, Italy; (L.S.A.A.); (A.C.)
| | - David J. A. Jenkins
- Departments of Nutritional Science and Medicine, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Clinical Nutrition and Risk Factor Modification Centre, St. Michael’s Hospital, Toronto, ON M5C 2T2, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, St. Michael’s Hospital, Toronto, ON M5C 2T2, Canada
- Li Ka Shing Knowledge Institute, St. Michael’s Hospital, Toronto, ON M5C 2T2, Canada
| | - Anna Crispo
- Epidemiology and Biostatistics Unit, Istituto Nazionale Tumori—IRCCS—“Fondazione G. Pascale”, via M. Semmola 1, 80131 Naples, Italy; (L.S.A.A.); (A.C.)
| | - Federica Toffolutti
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via F. Gallini 2, 33081 Aviano, Italy; (F.T.); (J.P.)
| | - Eva Negri
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, Università di Bologna, via P. Palagi 9, 40138 Bologna, Italy;
| | - Carlo La Vecchia
- Department of Clinical Sciences and Community Health, Department of Excellence 2023–2027, Branch of Medical Statistics, Biometry and Epidemiology “G.A. Maccacaro”, Università degli Studi di Milano, via Celoria 22, 20133 Milan, Italy; (M.D.M.); (M.F.)
| | - Monica Ferraroni
- Department of Clinical Sciences and Community Health, Department of Excellence 2023–2027, Branch of Medical Statistics, Biometry and Epidemiology “G.A. Maccacaro”, Università degli Studi di Milano, via Celoria 22, 20133 Milan, Italy; (M.D.M.); (M.F.)
- Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, via F. Sforza 35, 20122 Milan, Italy
| | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via F. Gallini 2, 33081 Aviano, Italy; (F.T.); (J.P.)
| |
Collapse
|
3
|
Peng L, Du Q, Xiang L, Gu H, Luo H, Xu Z, He H, Xia B, Zhou Z, Wang Y, Chen Y. Adherence to the low-fat diet pattern reduces the risk of lung cancer in American adults aged 55 years and above: a prospective cohort study. J Nutr Health Aging 2024; 28:100240. [PMID: 38663125 DOI: 10.1016/j.jnha.2024.100240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/06/2024] [Accepted: 04/13/2024] [Indexed: 07/07/2024]
Abstract
OBJECTIVES There is little evidence on the association between low-fat dietary patterns and lung cancer risk among middle-aged and older adults. To fill this gap, we comprehensively investigated the association of adherence to a low-fat diet (LFD) and intake of different fat components including saturated, monounsaturated, and polyunsaturated fatty acids with incidence of lung cancer and its subtypes [non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC)] among adults aged 55 years and older. DESIGN A prospective cohort study with a mean follow-up time of 8.8 years. SETTING AND PARTICIPANTS This study used data from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. The study population included 98,459 PLCO participants age 55 and over at baseline who completed food frequency questionnaires providing detailed dietary information and had no history of cancer. METHODS Dietary intake was assessed using a validated food frequency questionnaire at baseline. A LFD score was calculated based on fat, protein, and carbohydrate intake as a percentage of total calories. Cox proportional hazards regression models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for the association between LFD score and intake of fat components (in quartiles) and incident lung cancer and its subtypes over follow-up. Restricted cubic spline analyses were conducted to examine possible nonlinear relationships. Subgroup analyses were performed to evaluate potential effect modifiers, and several sensitivity analyses were conducted to assess the stability of the findings. RESULTS During a follow-up of 869,807.9 person-years, 1,642 cases of lung cancer were observed, consisting of 1,408 (85.75%) cases of NSCLC and 234 (14.25%) cases of SCLC. The highest versus the lowest quartiles of the LFD score were found to be associated with a reduced risk of lung cancer (HR, 0.76; 95% CI, 0.66-0.89), NSCLC (HR, 0.79; 95% CI, 0.67-0.93), and SCLC (HR, 0.59; 95% CI, 0.38-0.92). The restricted cubic spline plots demonstrated a linear dose-response relationship between the LFD score and the risk of lung cancer as well as its subtypes. This risk reduction association for overall lung cancer was more pronounced in smokers (HR, 0.71; 95% CI, 0.60-0.84; P for interaction = 0.003). For fat components, high consumption of saturated fatty acids was associated with an increased lung cancer risk (HR, 1.35; 95% CI, 1.10-1.66), especially for SCLC (HR, 2.05; 95% CI, 1.20-3.53). No significant association was found between consumption of monounsaturated or polyunsaturated fatty acids and incident lung cancer and its subtypes. CONCLUSIONS Our findings suggest that adherence to LFD may reduce the lung cancer risk, particularly in smokers; while high saturated fatty acids consumption may increase lung cancer risk, especially for SCLC, among middle-aged and older adults in the US population.
Collapse
Affiliation(s)
- Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qingqing Du
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haoyun Luo
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiquan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Boning Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhihang Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ying Chen
- Health Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Chen B, Patel S, Bao L, Nadeem D, Krittanawong C. Pro-Inflammatory Food, Gut Microbiota, and Cardiovascular and Pancreatic Diseases. Biomolecules 2024; 14:210. [PMID: 38397447 PMCID: PMC10886602 DOI: 10.3390/biom14020210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/25/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Recent studies have shown that a pro-inflammatory diet and dysbiosis, especially a high level of trimethylamine-N-oxide (TMAO), are associated with various adverse health conditions. Cardiovascular diseases and pancreatic diseases are two major morbidities in the modern world. Through this narrative review, we aimed to summarize the association between a pro-inflammatory diet, gut microbiota, and cardiovascular and pancreatic diseases, along with their underlying mechanisms. Our review revealed that TMAO is associated with the development of cardiovascular diseases by promoting platelet aggregation, atherosclerotic plaque formation, and vascular inflammation. TMAO is also associated with the development of acute pancreatitis. The pro-inflammatory diet is associated with an increased risk of pancreatic cancer and cardiovascular diseases through mechanisms that include increasing TMAO levels, activating the lipopolysaccharides cascade, and the direct pro-inflammatory effect of certain nutrients. Meanwhile, an anti-inflammatory diet decreases the risk of cardiovascular diseases and pancreatic cancer.
Collapse
Affiliation(s)
- Bing Chen
- Department of Gastroenterology, Hepatology, and Nutrition, Geisinger Medical Center, Danville, PA 17822, USA
| | - Shriraj Patel
- Department of Medicine, Geisinger Medical Center, Danville, PA 17822, USA
| | - Lingyu Bao
- Section on Molecular Morphogenesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20817, USA
| | - Danial Nadeem
- Department of Gastroenterology, Hepatology, and Nutrition, Geisinger Medical Center, Danville, PA 17822, USA
| | - Chayakrit Krittanawong
- Cardiology Division, NYU School of Medicine and NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
5
|
Mumtaz S, Akhtar N, Ahmed A, Qazi AS. Dietary Pattern and Cancer. Cancer Treat Res 2024; 191:191-216. [PMID: 39133409 DOI: 10.1007/978-3-031-55622-7_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Diet play an important role in the development of cancer. A lot of research has been done on the role of individual nutrients or phytochemicals and cancer risk. Both harmful and beneficial associations of this nutrient have been observed with cancer. However, there is an interaction of individual dietary constituents to influence disease risk. On the other hand, examining the diet as a whole as is done in dietary patterns research may produce more accurate estimates and data that can be more easily translated into dietary recommendations. Dietary patterns and cancer research are becoming increasingly common in the epidemiology literature, and novel dietary patterns are being generated at a rapid pace. However, major issues remain over whether one general "healthy" dietary pattern can be suggested for cancer prevention or whether several diets should be advocated for different forms of cancer protection. It is challenging to study typical human diet in animal model that is appropriate for cancer prevention. Some dietary patterns, such as the ketogenic diet or macronutrient composition alteration, have been investigated more extensively in animal models than in humans in terms of cancer prevention, and bigger human observational studies are now needed to advise dietary guidelines. The question of whether to adapt nutritional guidelines to population subgroups based on susceptibility factors (for example, family history, sex, age, other lifestyle factors or comorbidities, metabolomics signatures, or microbiota-based profiles) is still open and will be crucial in moving the field forward.
Collapse
Affiliation(s)
- Sara Mumtaz
- National University of Medical Sciences (NUMS), Rawalpindi, Pakistan.
| | - Nosheen Akhtar
- National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| | | | - Asma Saleem Qazi
- National University of Medical Sciences (NUMS), Rawalpindi, Pakistan
| |
Collapse
|
6
|
Michalak N, Małecka-Wojciesko E. Modifiable Pancreatic Ductal Adenocarcinoma (PDAC) Risk Factors. J Clin Med 2023; 12:4318. [PMID: 37445352 DOI: 10.3390/jcm12134318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
This study aims to summarize the modifiable risk factors for pancreatic ductal adenocarcinoma (PDAC) that have been known for a long time, as well as information from the most recent reports. As a cancer with a late diagnosis and poor prognosis, accurate analysis of PDAC risk factors is warranted. The incidence of this cancer continues to rise, and the five-year survival rate is the lowest with respect to other tumors. The influence of cigarette smoking, alcohol consumption, and chronic pancreatitis in increasing the risk of pancreatic ductal adenocarcinoma is continually being confirmed. There are also newly emerging reports relating to the impact of lifestyle, including physical activity, the gut and oral microbiome, and hepatotropic viruses. A precise understanding of PDAC risk factors can help to identify groups of high-risk patients, and this may contribute to population awareness and education as well as earlier diagnoses with possible better treatment outcomes.
Collapse
Affiliation(s)
- Natalia Michalak
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland
| | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Medical University of Lodz, 90-419 Lodz, Poland
| |
Collapse
|
7
|
Steel H, Park SY, Lim T, Stram DO, Boushey CJ, Hébert JR, Le Marchand L, Wu AH, Setiawan VW. Diet Quality and Pancreatic Cancer Incidence in the Multiethnic Cohort. Cancer Epidemiol Biomarkers Prev 2023; 32:123-131. [PMID: 36306381 PMCID: PMC10072126 DOI: 10.1158/1055-9965.epi-22-0564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/25/2022] [Accepted: 10/21/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Data on diet quality and pancreatic cancer are limited. We examined the relationship between diet quality, assessed by the Healthy Eating Index-2015 (HEI-2015), the Alternative Healthy Eating Index-2010 (AHEI-2010), the alternate Mediterranean Diet (aMED) score, the Dietary Approaches to Stop Hypertension (DASH) score and the energy-adjusted Dietary Inflammatory Index (E-DII), and pancreatic cancer incidence in the Multiethnic Cohort Study. METHODS Diet quality scores were calculated from a validated food frequency questionnaire administered at baseline. Cox models were used to calculate HR and 95% confidence intervals (CI) adjusted for age, sex, race/ethnicity, education, diabetes, family history of pancreatic cancer, physical activity, smoking variables, total energy intake, body mass index (BMI), and alcohol consumption. Stratified analyses by sex, race/ethnicity, smoking status, and BMI were conducted. RESULTS Over an average follow-up of 19.3 years, 1,779 incident pancreatic cancer cases were identified among 177,313 participants (average age of 60.2 years at baseline, 1993-1996). Overall, we did not observe associations between the dietary pattern scores and pancreatic cancer (aMED: 0.98; 95% CI, 0.83-1.16; HEI-2015: 1.03; 95% CI, 0.88-1.21; AHEI-2010: 1.03; 95% CI, 0.88-1.20; DASH: 0.92; 95% CI, 0.79-1.08; E-DII: 1.05; 95% CI, 0.89-1.23). An inverse association was observed with DASH for ever smokers (HR, 0.75; 0.61-0.93), but not for nonsmokers (HR, 1.05; 0.83-1.32). CONCLUSIONS The DASH diet showed an inverse association with pancreatic cancer among ever smokers, but does not show a protective association overall. IMPACT Modifiable measures are needed to reduce pancreatic cancer burden in these high-risk populations; our study adds to the discussion of the benefit of dietary changes.
Collapse
Affiliation(s)
- Heather Steel
- Department of Population and Public Health Sciences, Keck School of Medicine of University of Southern California, Los Angeles, California
| | - Song-Yi Park
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Tiffany Lim
- Department of Population and Public Health Sciences, Keck School of Medicine of University of Southern California, Los Angeles, California
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles
| | - Daniel O Stram
- Department of Population and Public Health Sciences, Keck School of Medicine of University of Southern California, Los Angeles, California
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles
| | - Carol J Boushey
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - James R Hébert
- Department of Epidemiology and Biostatistics and Cancer Prevention and Control Program, Arnold School of Public Health, University of South Carolina, Columbia, South Carolina
- Department of Nutrition, Connecting Health Innovations LLC, Columbia, South Carolina
| | - Loïc Le Marchand
- Cancer Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Anna H Wu
- Department of Population and Public Health Sciences, Keck School of Medicine of University of Southern California, Los Angeles, California
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles
| | - Veronica Wendy Setiawan
- Department of Population and Public Health Sciences, Keck School of Medicine of University of Southern California, Los Angeles, California
- Norris Comprehensive Cancer Center, Keck School of Medicine of University of Southern California, Los Angeles
- Center for Genetic Epidemiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
8
|
Ibrahim MO, Abuhijleh H, Tayyem R. What Dietary Patterns and Nutrients are Associated with Pancreatic Cancer? Literature Review. Cancer Manag Res 2023; 15:17-30. [PMID: 36643074 PMCID: PMC9832506 DOI: 10.2147/cmar.s390228] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/23/2022] [Indexed: 01/07/2023] Open
Abstract
This narrative review summarizes the main findings of observational studies (case-control and cohort) as well as systematic reviews and meta-analyses on the role of nutrients and dietary patterns on pancreatic cancer (PC) risk and elucidates possible mechanisms for the association between nutrients or specific food components and the risk of PC. A literature search of MEDLINE (PubMed), Google Scholar, ScienceDirect, and Scopus was performed. An extensive search of related articles published in the English language from 1985 to 2022 was carried out. Our search included macro- and micronutrient intake as well as dietary patterns associated with PC. In conclusion, the consumption of a diet high in nutrients such as sugar, fats, and red and processed meats can increase the risk of PC. Conversely, a high dietary intake of fresh fruit and vegetables and their associated nutrients like fiber, antioxidants, and polyphenols may prevent PC. Dietary patterns loaded with red and processed meats were also linked to an increased risk of PC, whereas dietary patterns rich in plant-based foods like vegetables, fruits, whole grains, and legumes were associated with a reduced risk of PC. Dietary fiber, fat-soluble vitamins, water-soluble vitamins, and minerals might also play a protective role against PC.
Collapse
Affiliation(s)
- Mohammed O Ibrahim
- Department of Nutrition and Food Technology, Faculty of Agriculture, Mu’tah University, Karak, Jordan
| | - Haya Abuhijleh
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Reema Tayyem
- Department of Human Nutrition, College of Health Sciences, QU Health, Qatar University, Doha, Qatar,Correspondence: Reema Tayyem, Department of Human Nutrition, College of Health Sciences, Qatar University, Doha, 2713, Qatar, Email
| |
Collapse
|
9
|
Ruze R, Chen Y, Xu R, Song J, Yin X, Wang C, Xu Q. Obesity, diabetes mellitus, and pancreatic carcinogenesis: Correlations, prevention, and diagnostic implications. Biochim Biophys Acta Rev Cancer 2023; 1878:188844. [PMID: 36464199 DOI: 10.1016/j.bbcan.2022.188844] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/26/2022] [Indexed: 12/03/2022]
Abstract
The prevalence of obesity, diabetes mellitus (DM), and pancreatic cancer (PC) has been consistently increasing in the last two decades worldwide. Sharing various influential risk factors in genetics and environmental inducers in pathogenesis, the close correlations of these three diseases have been demonstrated in plenty of clinical studies using multiple parameters among different populations. On the contrary, most measures aimed to manage and treat obesity and DM effectively reduce the risk and prevent PC occurrence, yet certain drugs can inversely promote pancreatic carcinogenesis instead. Most importantly, an elevation of blood glucose with or without a reduction in body weight, along with other potential tools, may provide valuable clues for detecting PC at an early stage in patients with obesity and DM, favoring a timely intervention and prolonging survival. Herein, the epidemiological and etiological correlations among these three diseases and the supporting clinical evidence of their connections are first summarized to favor a better and more thorough understanding of obesity- and DM-related pancreatic carcinogenesis. After comparing the distinct impacts of different weight-lowering and anti-diabetic treatments on the risk of PC, the possible diagnostic implications of hyperglycemia and weight loss in PC screening are also addressed in detail.
Collapse
Affiliation(s)
- Rexiati Ruze
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Yuan Chen
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Ruiyuan Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Jianlu Song
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Xinpeng Yin
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China; Chinese Academy of Medical Sciences and Peking Union Medical College, No. 9 Dongdan Santiao, Beijing, China
| | - Chengcheng Wang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China.
| | - Qiang Xu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Beijing, China.
| |
Collapse
|
10
|
Peng L, Xiang L, Xu Z, Gu H, Zhu Z, Tang Y, Jiang Y, He H, Wang Y, Zhao X. Association between low-fat diet and liver cancer risk in 98,455 participants: Results from a prospective study. Front Nutr 2022; 9:1013643. [PMID: 36466389 PMCID: PMC9716652 DOI: 10.3389/fnut.2022.1013643] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/26/2022] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Low-fat diet reduces the risk of chronic metabolic diseases such as obesity and diabetes, which exhibit overlapping mechanisms with liver cancer. However, the association between low-fat diet and liver cancer risk remains unclear. AIM To investigate whether adherence to low-fat diet is associated with a reduced risk of liver cancer in a prospective study. MATERIALS AND METHODS Data of participants in this study were collected from the Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. A low-fat diet score was calculated to reflect adherence to low-fat dietary pattern, with higher scores indicating greater adherence. Cox regression was used to calculate hazard ratios (HRs) and 95% confidence intervals (CIs) for liver cancer incidence with adjustment for potential covariates. Restricted cubic spline model was used to characterize liver cancer risk across the full range of the low-fat diet score. Prespecified subgroup analyses were used to identify potential impact modifiers. Sensitivity analyses were performed to test the robustness of this association. RESULTS A total of 98,455 participants were included in the present analysis. The mean (standard deviation) age, low-fat diet score, and follow-up time were 65.52 (5.73) years, 14.99 (6.27) points, and 8.86 (1.90) years, respectively. During 872639.5 person-years of follow-up, 91 liver cancers occurred, with an overall incidence rate of 0.01 cases per 100 person-years. In the fully adjusted Cox model, the highest versus the lowest quartile of low-fat diet score was found to be associated with a reduced risk of liver cancer (HR Q4 vs. Q1: 0.458; 95% CI: 0.218, 0.964; P = 0.035 for trend), which remained associated through a series of sensitivity analyses. The restricted cubic spline model showed a linear dose-response association between low-fat diet score and liver cancer incidence (p = 0.482 for non-linear). Subgroup analyses did not show significant interaction between low-fat diet score and potential impact modifiers in the incidence of liver cancer. CONCLUSION In this study, low-fat diet score is associated with reduced liver cancer risk in the US population, indicating that adherence to low-fat diet may be helpful for liver cancer prevention. Future studies should validate our findings in other populations.
Collapse
Affiliation(s)
- Linglong Peng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ling Xiang
- Department of Clinical Nutrition, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiquan Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhiyong Zhu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunhao Tang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yahui Jiang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongmei He
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yaxu Wang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaodong Zhao
- The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Olakowski M, Bułdak Ł. Modifiable and Non-Modifiable Risk Factors for the Development of Non-Hereditary Pancreatic Cancer. Medicina (B Aires) 2022; 58:medicina58080978. [PMID: 35893093 PMCID: PMC9394367 DOI: 10.3390/medicina58080978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic cancer is becoming an increasing healthcare concern. Though it is a 14th most common cancer worldwide, its incidence is steadily rising. Results of currently available therapies are still not satisfactory. Therefore, great attention should be put on the identification and reduction of risk factors for pancreatic cancer. A thorough up-to-date review of available data on the impact of well-established and novel risk factors of pancreatic cancer development have been performed. Several risk factors associated with lifestyle have significant impact on the risk of pancreatic cancer (i.e., smoking, obesity, alcohol consumption). Physicians should also be aware of the novel findings suggesting increasing role of microbiome, including viral and bacterial infections, in the development of pancreatic cancer. A growing body of evidence suggest also an increased risk during certain occupational exposures. In general, lifestyle seems to be a major contributor in the development of pancreatic cancer. Special attention should be given to individuals with a vicious cluster consisting of metabolic syndrome, tobacco smoking and alcohol consumption. Physicians should urge patients to comply to healthy diet, cessation of smoking and moderation of alcohol consumption, which may halve pancreatic cancer incidence. Further studies are warranted to explore the potential use of therapeutic approach on novel risk factors (e.g., microbiome).
Collapse
Affiliation(s)
- Marek Olakowski
- Department of Gastrointestinal Surgery, Medical University of Silesia, Medyków 14, 40-752 Katowice, Poland;
| | - Łukasz Bułdak
- Department of Internal Medicine and Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland
- Correspondence:
| |
Collapse
|
12
|
Saliakoura M, Sebastiano MR, Nikdima I, Pozzato C, Konstantinidou G. Restriction of extracellular lipids renders pancreatic cancer dependent on autophagy. J Exp Clin Cancer Res 2022; 41:16. [PMID: 34998392 PMCID: PMC8742413 DOI: 10.1186/s13046-021-02231-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/21/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND KRAS is the predominant oncogene mutated in pancreatic ductal adenocarcinoma (PDAC), the fourth cause of cancer-related deaths worldwide. Mutant KRAS-driven tumors are metabolically programmed to support their growth and survival, which can be used to identify metabolic vulnerabilities. In the present study, we aimed to understand the role of extracellularly derived fatty acids in KRAS-driven pancreatic cancer. METHODS To assess the dependence of PDAC cells on extracellular fatty acids we employed delipidated serum or RNAi-mediated suppression of ACSL3 (to inhibit the activation and cellular retention of extracellular fatty acids) followed by cell proliferation assays, qPCR, apoptosis assays, immunoblots and fluorescence microscopy experiments. To assess autophagy in vivo, we employed the KrasG12D/+;p53flox/flox;Pdx1-CreERT2 (KPC) mice crossed with Acsl3 knockout mice, and to assess the efficacy of the combination therapy of ACSL3 and autophagy inhibition we used xenografted human cancer cell-derived tumors in immunocompromised mice. RESULTS Here we show that depletion of extracellularly derived lipids either by serum lipid restriction or suppression of ACSL3, triggers autophagy, a process that protects PDAC cells from the reduction of bioenergetic intermediates. Combined extracellular lipid deprivation and autophagy inhibition exhibits anti-proliferative and pro-apoptotic effects against PDAC cell lines in vitro and promotes suppression of xenografted human pancreatic cancer cell-derived tumors in mice. Therefore, we propose lipid deprivation and autophagy blockade as a potential co-targeting strategy for PDAC treatment. CONCLUSIONS Our work unravels a central role of extracellular lipid supply in ensuring fatty acid provision in cancer cells, unmasking a previously unappreciated metabolic vulnerability of PDAC cells.
Collapse
Affiliation(s)
- Maria Saliakoura
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | | - Ioanna Nikdima
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | - Chiara Pozzato
- Institute of Pharmacology, University of Bern, 3010, Bern, Switzerland
| | | |
Collapse
|
13
|
Guo Z, Hong Y, Cheng Y. Dietary inflammatory index and pancreatic cancer risk: a systematic review and dose-response meta-analysis. Public Health Nutr 2021; 24:6427-6435. [PMID: 33843543 PMCID: PMC11148588 DOI: 10.1017/s1368980021001579] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECTIVE The meta-analysis was conducted to test the link between pancreatic cancer (PC) risk and dietary inflammatory index (DII®) score. DESIGN Systematic review and meta-analysis. SETTING We searched PubMed, Embase, Web of Science and the Cochrane Library up to 22 November 2020 to identify the relevant studies. Studies that reported the risk estimates and the corresponding 95 % CI for the DII category and PC risk were included. The effect sizes were pooled using the random-effects model. Dose-response analysis was conducted where possible. PARTICIPANTS Two prospective cohort studies of 634 705 participants (3152 incident cases), and four case-control studies of 2737 cases and 4861 controls. RESULTS Overall, the pooled risk ratio (RR) indicated that individuals in the highest category compared with the lowest category had an increased PC risk (RR = 1·45; 95 % CI 1·11, 1·90; P = 0·006). Meanwhile, significant heterogeneity was also revealed. The dose-response meta-analysis indicated that a 1-unit increase in the DII score was associated with the PC risk (RR = 1·08; 95 % CI 1·002, 1·166; P = 0·045; I2 = 94·1 %, P < 0·001). Nonlinear result showed an increased risk of moving from fewer to more inflammatory borders with increasing DII score (Pnonlinearity = 0·003; I2 = 76·5 %, P < 0·001). Subgroup analyses found that significant positive association between PC risk and DII score appeared to be in case-control studies (RR = 1·70; 95 % CI 1·16, 2·50; P = 0·007) and studies with ≤ 31 DII components (RR = 1·76; 95 % CI 1·14, 2·72; P = 0·011). CONCLUSION These findings suggested dietary habits with high inflammatory features (high DII score) might increase PC risk.
Collapse
Affiliation(s)
- Zhangyou Guo
- Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming 650118, People's Republic of China
| | - Yuan Hong
- Medical Laboratory, The First Affiliated Hospital of Kunming Medical University/Institute of Experimental Diagnostics of Yunnan Province/Key Laboratory of Laboratory Medicine of Yunnan Province, Kunming 650032, People's Republic of China
| | - Yao Cheng
- Department of Hepatobiliary Surgery of the Second Affiliated Hospital of Chongqing Medical University, Chongqing400010, People's Republic of China
| |
Collapse
|
14
|
Li D, Zheng J, Hatia R, Hassan M, Daniel CR. Dietary Intake of Fatty Acids and Risk of Pancreatic Cancer: A Case-Control Study. J Nutr 2021; 152:439-447. [PMID: 34665254 PMCID: PMC8826846 DOI: 10.1093/jn/nxab372] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Epidemiological findings on dietary fat intake and risk of pancreatic cancer (PanC) are inconsistent. OBJECTIVES This study aimed to determine the association between types of dietary fat intake and PanC. METHODS We conducted a hospital-based case-control study in 957 pathologically confirmed PanC cases and 938 cancer-free controls. Cases and controls were frequency matched by age, sex, and race. Dietary information was collected using a self-administered validated FFQ. Unconditional logistic regression models were used to estimate the ORs and 95% CIs of PanC risk by quintiles of fat intake with the lowest quintile as referent and with adjustment for other risk factors and dietary factors. RESULTS We observed no difference in (median) intake of total fat standardized for energy among cases versus controls. The multivariable-adjusted OR (95% CI) of the highest versus the lowest quintile of intake (ORQ5 compared with Q1) was 2.51 (1.68-3.72) for fat from animal sources and 0.41 (0.29-0.58) for fat from plant sources. Intakes of total MUFA, total PUFA, and linoleic (n-6) and long chain n-3 fatty acids were inversely associated with PanC (ORQ5 compared with Q1 and 95% CI: 0.55 [0.36-0.82], 0.59 [0.42-0.82], 0.64 [0.43-0.84], and 0.60 [0.42-0.84], respectively). Arachidonic acid (n-6) and several SFAs were positively associated with PanC. CONCLUSION Although some observed associations with pancreatic cancer risk could be explained by reverse causation, the potential protective associations with intakes of largely plant-derived PUFAs and MUFAs and fish-derived long chain n-3 PUFAs warrant further prospective investigation.
Collapse
Affiliation(s)
| | - Jiali Zheng
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rikita Hatia
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Manal Hassan
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carrie R Daniel
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
Hu JX, Zhao CF, Chen WB, Liu QC, Li QW, Lin YY, Gao F. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J Gastroenterol 2021; 27:4298-4321. [PMID: 34366606 PMCID: PMC8316912 DOI: 10.3748/wjg.v27.i27.4298] [Citation(s) in RCA: 223] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/18/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023] Open
Abstract
Despite rapid advances in modern medical technology and significant improvements in survival rates of many cancers, pancreatic cancer is still a highly lethal gastrointestinal cancer with a low 5-year survival rate and difficulty in early detection. At present, the incidence and mortality of pancreatic cancer are increasing year by year worldwide, no matter in the United States, Europe, Japan, or China. Globally, the incidence of pancreatic cancer is projected to increase to 18.6 per 100000 in 2050, with the average annual growth of 1.1%, meaning that pancreatic cancer will pose a significant public health burden. Due to the special anatomical location of the pancreas, the development of pancreatic cancer is usually diagnosed at a late stage with obvious clinical symptoms. Therefore, a comprehensive understanding of the risk factors for pancreatic cancer is of great clinical significance for effective prevention of pancreatic cancer. In this paper, the epidemiological characteristics, developmental trends, and risk factors of pancreatic cancer are reviewed and analyzed in detail.
Collapse
Affiliation(s)
- Jian-Xiong Hu
- Intensive Care Unit (ICU), Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China
| | - Cheng-Fei Zhao
- School of Pharmacy and Medical Technology, Putian University, Putian 351100, Fujian Province, China
- Key Laboratory of Pharmaceutical Analysis and Laboratory Medicine in University of Fujian Province, Putian University, Putian 351100, Fujian Province, China
| | - Wen-Biao Chen
- Department of Basic Medicine, Quanzhou Medical College, Quanzhou 362011, Fujian Province, China
| | - Qi-Cai Liu
- Department of Reproductive Medicine Centre, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| | - Qu-Wen Li
- Department of Priority Laboratory for Zoonoses Research, Fujian Center for Disease Control and Prevention, Fuzhou 350001, Fujian Province, China
| | - Yan-Ya Lin
- Intensive Care Unit (ICU), Affiliated Hospital of Putian University, Putian 351100, Fujian Province, China
| | - Feng Gao
- Department of Pathology, First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China
| |
Collapse
|
16
|
Cai J, Chen H, Lu M, Zhang Y, Lu B, You L, Zhang T, Dai M, Zhao Y. Advances in the epidemiology of pancreatic cancer: Trends, risk factors, screening, and prognosis. Cancer Lett 2021; 520:1-11. [PMID: 34216688 DOI: 10.1016/j.canlet.2021.06.027] [Citation(s) in RCA: 148] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/09/2021] [Accepted: 06/25/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a malignancy with poor prognosis and high mortality. The recent increase in pancreatic cancer incidence and mortality has resulted in an increased number of studies on its epidemiology. This comprehensive and systematic literature review summarizes the advances in the epidemiology of pancreatic cancer, including its epidemiological trends, risk factors, risk prediction models, screening modalities, and prognosis. The risk factors for pancreatic cancers can be categorized as those related to individual characteristics, lifestyle and environment, and disease status. Several prediction models for pancreatic cancer have been developed in populations with new-onset diabetes or a family history of pancreatic cancer; however, these models require further validation. Despite recent progress in pancreatic cancer screening, the quantity and quality of related studies are also unsatisfactory, especially with respect to the identification of high-risk populations and development of effective screening modality. Apart from the populations with familial genetic risk and those at a high risk of sporadic pancreatic cancer, risk factors such as new-onset diabetes may be a new direction for timely intervention. We hope this work will provide new ideas for further prevention and treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Jie Cai
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Hongda Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Ming Lu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Yuhan Zhang
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Bin Lu
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China
| | - Lei You
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Min Dai
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
17
|
Crudele L, Piccinin E, Moschetta A. Visceral Adiposity and Cancer: Role in Pathogenesis and Prognosis. Nutrients 2021; 13:2101. [PMID: 34205356 PMCID: PMC8234141 DOI: 10.3390/nu13062101] [Citation(s) in RCA: 33] [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: 05/25/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
The prevalence of being overweight and obese has been expanded dramatically in recent years worldwide. Obesity usually occurs when the energetic introit overtakes energy expenditure from metabolic and physical activity, leading to fat accumulation mainly in the visceral depots. Excessive fat accumulation represents a risk factor for many chronic diseases, including cancer. Adiposity, chronic low-grade inflammation, and hyperinsulinemia are essential factors of obesity that also play a crucial role in tumor onset. In recent years, several strategies have been pointed toward boundary fat accumulation, thus limiting the burden of cancer attributable to obesity. While remodeling fat via adipocytes browning seems a tempting prospect, lifestyle interventions still represent the main pathway to prevent cancer and enhance the efficacy of treatments. Specifically, the Mediterranean Diet stands out as one of the best dietary approaches to curtail visceral adiposity and, therefore, cancer risk. In this Review, the close relationship between obesity and cancer has been investigated, highlighting the biological mechanisms at the basis of this link. Finally, strategies to remodel fat, including browning and lifestyle interventions, have been taken into consideration as a major perspective to limit excess body weight and tumor onset.
Collapse
Affiliation(s)
- Lucilla Crudele
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.C.); (E.P.)
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Elena Piccinin
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.C.); (E.P.)
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (L.C.); (E.P.)
- INBB, National Institute for Biostructures and Biosystems, 00136 Rome, Italy
- National Cancer Center, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| |
Collapse
|
18
|
Systematic Review of the Integrative Medicine Recommendations for Patients with Pancreatic Cancer. SURGERIES 2021. [DOI: 10.3390/surgeries2020022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Introduction: Integrative medicine (IM) is a relatively new field where non-traditional therapies with peer-reviewed evidence are incorporated or integrated with more traditional approaches. Methods: A systematic review of the literature from the last 10 years was done by searching clinical trials and randomized-controlled trials on Pubmed that discuss nutrition, supplementation, and lifestyle changes associated with “Pancreatic Cancer.” Results: Only 50 articles ultimately met the inclusion criteria for this review. A total of 15 articles discussed the role of obesity and 10 discussed the influence of stress in increasing the risk of pancreatic cancer. Six discussed the potential beneficial role of Vitamins, 5 of cannabinoids, 4 an anti-inflammatory diet, 3 of nut consumption, 2 of green tea consumption, 2 of curcumin supplementation, 1 role of melatonin, and 1 of probiotics. One article each was found on the theoretical benefits of adhering to either a Mediterranean or ketogenic diet. Discussion: As more surgeons become interested in IM, it is hoped that more diseases where the curative treatment is mainly surgical can benefit from the all-encompassing principles of IM in an effort to improve quality of life and survival in patients with pancreatic cancer.
Collapse
|
19
|
Khalaf N, El-Serag HB, Abrams HR, Thrift AP. Burden of Pancreatic Cancer: From Epidemiology to Practice. Clin Gastroenterol Hepatol 2021; 19:876-884. [PMID: 32147593 PMCID: PMC8559554 DOI: 10.1016/j.cgh.2020.02.054] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 02/05/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is the seventh leading cause of cancer-related deaths worldwide with 432,242 related deaths in 2018. Unlike other cancers, the incidence of pancreatic cancer continues to increase, with little improvement in survival rates. We review the epidemiologic features of pancreatic cancer, covering surveillance and early detection in high-risk persons. We summarize data on worldwide incidence and mortality and analyze the 1975-2016 data from 9 registries of the National Cancer Institute's Surveillance, Epidemiology, and End Results study, on the overall burden of pancreatic cancer as well as age-, sex-, and race-specific incidence, survival rates and trends. It is important to increase our knowledge of the worldwide and regional epidemiologic features of and risk factors for pancreatic cancer, to identify new approaches for prevention, surveillance, and treatment.
Collapse
Affiliation(s)
- Natalia Khalaf
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas.
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine and Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas
| | - Hannah R Abrams
- Department of Internal Medicine, Baylor College of Medicine, Houston, Texas
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
20
|
Nojomi M, Tehrani Banihashemi A, Niksima H, Hashemian M, Mottaghi A, Malekzaddeh R. The relationship between dietary patterns, dietary quality index, and dietary inflammatory index with the risk of all types of cancer: Golestan cohort study. Med J Islam Repub Iran 2021; 35:48. [PMID: 34268236 PMCID: PMC8271224 DOI: 10.47176/mjiri.35.48] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Indexed: 01/29/2023] Open
Abstract
Background: Dietary patterns and diet quality index (DQI) are widely discussed in relation with different health conditions and have recently been taken into consideration for all cancer types. Since chronic inflammation has been recognized as an important biologic risk factor for cancer occurrence, especially in epithelial tissues, proinflammatory or anti-inflammatory characteristics of diet has become the center of attention. In the present study, we aimed to identify whether a specific dietary pattern, Mediterranean dietary score (MDs), and dietary inflammatory index (DII) were associated with overall cancer risk in Iranian population.
Methods: This study was performed in the context of the Golestan cohort study. Participants with extreme daily energy intake or those who did not answer more than 30 question of the Food Frequency questionnaire (FFQ) were excluded. Dietary patterns, MDs, and DII were measured from FFQ. Age, sex, total energy, place of residence, smoking, wealth score, ethnicity, opiate use, BMI, education, marital status, and physical activity score were considered as confounding variables. Using Cox proportional hazards regression models, hazard ratios (HRs) and 95% confidence interval of cancer were estimated.
Results: HRs (95% CIs) of all cancers by quartiles of Western dietary pattern, DII, and MDs showed that the forth quartile of the Western dietary pattern is attributed to 23% higher cancer risk (HRs: 1.23, CI: 1.09-1.40, P< 0.001, adjusted for age and sex) compared to the first quartile. It also remained significant after further adjustments (HRs = 1.20, CI: 1.06-1.36, P< 0.001). There was a higher cancer risk in the fourth quartile of DII in comparison with the first quartile (HRs = 1.16, CI: 1.01-1.32, P trend < 0.001, adjusted for age and sex). The lower adherence to the Mediterranean dietary pattern also largely contributes to 27% higher cancer risk (HRs: 1.27, CI: 1.12-1.44), P trend < 0.001, adjusted for age and sex), which also remained remarkable after further adjustments ((HRs =1.19, CI: 1.05-1.35, P trend < 0.001).
Conclusion: Cancer is highly correlated to dietary intake and dietary patterns, such as the Western dietary pattern, while the Mediterranean diet score was inversely associated with cancer risk. Further investigations are required to get a broader insight into cancer determinants in population.
Collapse
Affiliation(s)
- Marzieh Nojomi
- Preventive Medicine & Public Health Research Center, Psychosocial Health Research Institute, Department of Community and Family Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Sociology & Anthropology, Nipissing University, North Bay, Ontario, Canada
| | - Arash Tehrani Banihashemi
- Preventive Medicine & Public Health Research Center, Department of Community and Family Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hassan Niksima
- Preventive Medicine & Public Health Research Center, Department of Community and Family Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Hashemian
- Metabolic Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Azadeh Mottaghi
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology & Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Malekzaddeh
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
21
|
Jin Q, Hart PA, Shi N, Joseph JJ, Donneyong M, Conwell DL, Clinton SK, Cruz-Monserrate Z, Brasky TM, Tinker LF, Liu S, Shadyab AH, Thomson CA, Qi L, Rohan T, Tabung FK. Dietary Patterns of Insulinemia, Inflammation and Glycemia, and Pancreatic Cancer Risk: Findings from the Women's Health Initiative. Cancer Epidemiol Biomarkers Prev 2021; 30:1229-1240. [PMID: 33827986 DOI: 10.1158/1055-9965.epi-20-1478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/11/2020] [Accepted: 03/29/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Pancreatic cancer risk is increasing in countries with high consumption of Western dietary patterns and rising obesity rates. We examined the hypothesis that specific dietary patterns reflecting hyperinsulinemia (empirical dietary index for hyperinsulinemia; EDIH), systemic inflammation (empirical dietary inflammatory pattern; EDIP), and postprandial glycemia [glycemic index (GI); glycemic load (GL)] are associated with pancreatic cancer risk, including the potential modifying role of type 2 diabetes (T2D) and body mass index (BMI). METHODS We calculated dietary scores from baseline (1993-1998) food frequency questionnaires among 129,241 women, 50-79 years-old in the Women's Health Initiative. We used multivariable-adjusted Cox regression to estimate HRs and 95% confidence intervals (95% CI) for pancreatic cancer risk. RESULTS During a median 19.9 years of follow-up, 850 pancreatic cancer cases were diagnosed. We observed no association between dietary scores and pancreatic cancer risk overall. However, risk was elevated among participants with longstanding T2D (present >3 years before pancreatic cancer diagnosis) for EDIH. For each 1 SD increment in dietary score, the HRs (95% CIs) were: EDIH, 1.33 (1.06-1.66); EDIP, 1.26 (0.98-1.63); GI, 1.26 (0.96-1.67); and GL, 1.23 (0.96-1.57); although interactions were not significant (all P interaction >0.05). Separately, we observed inverse associations between GI [0.86 (0.76-0.96), P interaction = 0.0068] and GL [0.83 (0.73-0.93), P interaction = 0.0075], with pancreatic cancer risk among normal-weight women. CONCLUSIONS We observed no overall association between the dietary patterns evaluated and pancreatic cancer risk, although women with T2D appeared to have greater cancer risk. IMPACT The elevated risk for hyperinsulinemic diets among women with longstanding T2D and the inverse association among normal-weight women warrant further examination.
Collapse
Affiliation(s)
- Qi Jin
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio
| | - Phil A Hart
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Ni Shi
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Joshua J Joseph
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | | | - Darwin L Conwell
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Steven K Clinton
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio.,The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, Ohio.,Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Zobeida Cruz-Monserrate
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.,The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, Ohio
| | - Theodore M Brasky
- The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, Ohio.,Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, Ohio
| | - Lesley F Tinker
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Simin Liu
- Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Aladdin H Shadyab
- Department of Family Medicine and Public Health, School of Medicine, University of California San Diego, La Jolla, California
| | - Cynthia A Thomson
- Department of Health Promotion Sciences, Mel & Enid Zuckerman College of Public Health, The University of Arizona, Tucson, Arizona
| | - Lihong Qi
- School of Medicine, University of California Davis, Davis, California
| | - Thomas Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Fred K Tabung
- Interdisciplinary Ph.D. Program in Nutrition, The Ohio State University, Columbus, Ohio. .,The Ohio State University Comprehensive Cancer Center - James Cancer Hospital and Solove Research Institute, Columbus, Ohio.,Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio.,Division of Epidemiology, College of Public Health, The Ohio State University, Columbus, Ohio
| |
Collapse
|
22
|
Zhong GC, Li QJ, Yang PF, Wang YB, Hao FB, Wang K, Hu JJ, Wu JJ. Low-carbohydrate diets and the risk of pancreatic cancer: a large prospective cohort study. Carcinogenesis 2021; 42:724-732. [PMID: 33480980 DOI: 10.1093/carcin/bgab006] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/24/2020] [Accepted: 01/18/2021] [Indexed: 12/14/2022] Open
Abstract
Low-carbohydrate diets have become a popular approach for weight loss in recent years. However, whether low-carbohydrate diets are associated with the risk of pancreatic cancer remains to be elucidated. Hence, we examined the association of low-carbohydrate diets with the risk of pancreatic cancer in a US population. A population-based cohort of 95 962 individuals was identified. A low-carbohydrate-diet score was calculated to quantify adherence to this dietary pattern, with higher scores indicating greater adherence. Cox regression was used to calculate risk estimate for the association of the low-carbohydrate-diet score with the risk of pancreatic cancer. Subgroup analysis was used to identify the potential effect modifiers. After an average follow-up of 8.87 years (875856.9 person-years), we documented a total of 351 pancreatic cancer cases. In the fully adjusted model, the highest versus the lowest quartiles of the overall low-carbohydrate-diet score were found to be associated with a reduced risk of pancreatic cancer (hazard ratioquartile 4 versus 1: 0.61; 95% confidence interval: 0.45, 0.82; Ptrend < 0.001). Subgroup analysis found that the inverse association of low-carbohydrate diets with the risk of pancreatic cancer was more pronounced in individuals aged ≥65 years than in those aged <65 years (Pinteraction = 0.015). Similar results were obtained for animal and vegetable low-carbohydrate-diet scores. In conclusion, low-carbohydrate diets, regardless of the type of protein and fat, are associated with a lower risk of pancreatic cancer in the US population, suggesting that adherence to low-carbohydrate diets may be beneficial for pancreatic cancer prevention. Future studies should validate our findings in other populations.
Collapse
Affiliation(s)
- Guo-Chao Zhong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qu-Jin Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peng-Fei Yang
- Department of Nephrology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Bing Wang
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fa-Bao Hao
- Department of Neurosurgery, Qingdao Women and Children's Hospital, Qingdao University, Qingdao, Shandong, China
| | - Kang Wang
- Department of Endocrine and Breast Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie-Jun Hu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jing-Jing Wu
- Department of Nutrition and Food Hygiene, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
23
|
Zanini S, Renzi S, Limongi AR, Bellavite P, Giovinazzo F, Bermano G. A review of lifestyle and environment risk factors for pancreatic cancer. Eur J Cancer 2021; 145:53-70. [PMID: 33423007 DOI: 10.1016/j.ejca.2020.11.040] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 11/04/2020] [Accepted: 11/24/2020] [Indexed: 12/12/2022]
Abstract
Pancreatic cancer (PaCa) is one of the deadliest cancers known and its incidence is increasing in the developed countries. Because of the lack of biomarkers that allow early detection and the tendency of the disease to be asymptomatic, the diagnosis comes often too late for effective surgical or chemotherapy intervention. Lifestyle factors, that may cause common genetic modifications occurring in the disease, interfere with pancreatic physiology or function, and play a role in PaCa development, have been of concern recently, since a strategy to prevent this severe cancer is needed. This review identifies the latest evidences related to increased risk of developing PaCa due to dietary habits such as high alcohol, fructose and red or processed meat intake, and pathological conditions such as diabetes, obesity and infections in addition to stress and smoking behaviour. It aims to highlight the importance of intervening on modifiable risk factors: the action on these factors could prevent a considerable number of new cases of PaCa.
Collapse
Affiliation(s)
- Sara Zanini
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Serena Renzi
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK
| | - Antonina R Limongi
- Department of Science, University of Basilicata, Potenza, Italy; BioInnova Srl, Potenza, Italy
| | - Paolo Bellavite
- Department of Medicine, Section of General Pathology, University of Verona, Italy
| | | | - Giovanna Bermano
- Centre for Obesity Research and Education [CORE], School of Pharmacy & Life Sciences, Robert Gordon University, Aberdeen, UK.
| |
Collapse
|
24
|
Integrated bioinformatics analysis identified COL11A1 as an immune infiltrates correlated prognosticator in pancreatic adenocarcinoma. Int Immunopharmacol 2020; 90:106982. [PMID: 33129696 DOI: 10.1016/j.intimp.2020.106982] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/21/2020] [Accepted: 09/04/2020] [Indexed: 12/26/2022]
Abstract
Pancreatic adenocarcinoma (PAAD) is the most common pancreatic cancer, with high mortality rate and limited treatment options. Tumor infiltrating cells and genes in microenvironment are emerging as pivotal players in PAAD progression and prognosis. In this study, we obtained genes expression data set GSE119794 of PAAD, which contains data from 10 tumor and 10 normal samples. A total of 262 differentially expressed genes (DEGs), including 169 up-regulated and 93 down-regulated genes, were obtained based on expression fold change and significance. Combining the pathway analysis of DEGs and GSEA analysis of all genes, four KEGG pathways were enriched. The 4 pathways include pancreatic secretion, protein digestion and absorption, fat digestion and absorption, and PPAR signaling pathways. Functional enrichment of Gene Ontology significantly enriched extracellular matrix, an important component in microenvironment. In the Protein-protein interaction (PPI) network, we screened out 3 hub genes of COL11A1, KRT19 and CXCL5 by CytoHubba. At last, the expression level, prognostic significance and correlation with tumor infiltrates were validated in TCGA database, with GEPIA and TIMER. The validation identified Collagen Type XI Alpha 1 Chain (COL11A1), an extracellular matrix structural constituent, as a hazardous prognosticator with significant correlation with macrophage, neutrophil and dendritic cells. In sum, we identified COL11A1 as an immune infiltrates correlated prognosticator in pancreatic adenocarcinoma.
Collapse
|
25
|
The Role of Dysfunctional Adipose Tissue in Pancreatic Cancer: A Molecular Perspective. Cancers (Basel) 2020; 12:cancers12071849. [PMID: 32659999 PMCID: PMC7408631 DOI: 10.3390/cancers12071849] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Pancreatic cancer (PC) is a lethal malignancy with rising incidence and limited therapeutic options. Obesity is a well-established risk factor for PC development. Moreover, it negatively affects outcome in PC patients. Excessive fat accumulation in obese, over- and normal-weight individuals induces metabolic and inflammatory changes of adipose tissue microenvironment leading to a dysfunctional adipose “organ”. This may drive the association between abnormal fat accumulation and pancreatic cancer. In this review, we describe several molecular mechanisms that underpin this association at both local and systemic levels. We focus on the role of adipose tissue-derived circulating factors including adipokines, hormones and pro-inflammatory cytokines, as well as on the impact of the local adipose tissue in promoting PC. A discussion on potential therapeutic interventions, interfering with pro-tumorigenic effects of dysfunctional adipose tissue in PC, is included. Considering the raise of global obesity, research efforts to uncover the molecular basis of the relationship between pancreatic cancer and adipose tissue dysfunction may provide novel insights for the prevention of this deadly disease. In addition, these efforts may uncover novel targets for personalized interventional strategies aimed at improving the currently unsatisfactory PC therapeutic options.
Collapse
|
26
|
Stopa KB, Kusiak AA, Szopa MD, Ferdek PE, Jakubowska MA. Pancreatic Cancer and Its Microenvironment-Recent Advances and Current Controversies. Int J Mol Sci 2020; 21:E3218. [PMID: 32370075 PMCID: PMC7246785 DOI: 10.3390/ijms21093218] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) causes annually well over 400,000 deaths world-wide and remains one of the major unresolved health problems. This exocrine pancreatic cancer originates from the mutated epithelial cells: acinar and ductal cells. However, the epithelia-derived cancer component forms only a relatively small fraction of the tumor mass. The majority of the tumor consists of acellular fibrous stroma and diverse populations of the non-neoplastic cancer-associated cells. Importantly, the tumor microenvironment is maintained by dynamic cell-cell and cell-matrix interactions. In this article, we aim to review the most common drivers of PDAC. Then we summarize the current knowledge on PDAC microenvironment, particularly in relation to pancreatic cancer therapy. The focus is placed on the acellular stroma as well as cell populations that inhabit the matrix. We also describe the altered metabolism of PDAC and characterize cellular signaling in this cancer.
Collapse
Affiliation(s)
- Kinga B. Stopa
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7A, 30-387 Krakow, Poland;
| | - Agnieszka A. Kusiak
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland; (A.A.K.); (M.D.S.)
| | - Mateusz D. Szopa
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland; (A.A.K.); (M.D.S.)
| | - Pawel E. Ferdek
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, ul. Gronostajowa 7, 30-387 Krakow, Poland; (A.A.K.); (M.D.S.)
| | - Monika A. Jakubowska
- Malopolska Centre of Biotechnology, Jagiellonian University, ul. Gronostajowa 7A, 30-387 Krakow, Poland;
| |
Collapse
|
27
|
Rawla P, Thandra KC, Sunkara T. Pancreatic cancer and obesity: epidemiology, mechanism, and preventive strategies. Clin J Gastroenterol 2019; 12:285-291. [PMID: 30788774 DOI: 10.1007/s12328-019-00953-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 02/14/2019] [Indexed: 12/27/2022]
Abstract
The prevalence of obesity has seen a global increase in the past decades, escalating to one of the major epidemiological challenges today. Global economic growth has caused changes in dietary and physical activity patterns fueling obesity across age, gender, and income groups. The implications are many, as obesity has been associated with numerous serious health conditions, ultimately affecting morbidity and mortality. There is a growing recognition of the risk a high body mass index confers on the development and outcome of several malignancies, including pancreatic cancer. Pancreatic cancer is a highly lethal disease with exceptionally poor outcome, with incidences rising worldwide. Due to vague symptoms and no screening recommendations, a vast majority of patients are diagnosed at late stages, with already advanced disease and no opportunity for surgical intervention. Obesity mediates risk for pancreatic cancer through insufficiently understood mechanisms, possibly including inflammation and hormonal misbalance. As excess abdominal adiposity is among the few modifiable risk factors for pancreatic cancer onset, enduring weight loss could manifest an effective preventive measure. Lifestyle modifications on a population level aimed to reduce obesity could also scale down the grim pancreatic cancer rates. In cases when these measures alone are insufficiently effective, bariatric surgery can be an advantageous alternative. Extremely obese patients exhibit many health benefits following bariatric surgery along with weight loss, consequently reducing the chances of pancreatic cancer, especially if additionally adopting healthy lifestyle habits.
Collapse
Affiliation(s)
- Prashanth Rawla
- Department of Internal Medicine/Hospitalist, SOVAH Health, 320 Hospital Dr, Martinsville, VA, 24115, USA.
| | - Krishna Chaitanya Thandra
- Department of Pulmonary and Critical Care Medicine, Sentara Virginia Beach General Hospital, Virginia Beach, VA, USA
| | - Tagore Sunkara
- Division of Gastroenterology and Hepatology, Mercy Medical Center, Des Moines, IA, 50314, USA
| |
Collapse
|
28
|
Parohan M, Sadeghi A, Khatibi SR, Nasiri M, Milajerdi A, Khodadost M, Sadeghi O. Dietary total antioxidant capacity and risk of cancer: a systematic review and meta-analysis on observational studies. Crit Rev Oncol Hematol 2019; 138:70-86. [DOI: 10.1016/j.critrevonc.2019.04.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 11/30/2018] [Accepted: 04/01/2019] [Indexed: 01/04/2023] Open
|
29
|
Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int J Mol Sci 2019; 20:ijms20030676. [PMID: 30764482 PMCID: PMC6387440 DOI: 10.3390/ijms20030676] [Citation(s) in RCA: 216] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 01/23/2019] [Accepted: 01/31/2019] [Indexed: 12/24/2022] Open
Abstract
Systemic and local chronic inflammation might enhance the risk of pancreatic ductal adenocarcinoma (PDAC), and PDAC-associated inflammatory infiltrate in the tumor microenvironment concurs in enhancing tumor growth and metastasis. Inflammation is closely correlated with immunity, the same immune cell populations contributing to both inflammation and immune response. In the PDAC microenvironment, the inflammatory cell infiltrate is unbalanced towards an immunosuppressive phenotype, with a prevalence of myeloid derived suppressor cells (MDSC), M2 polarized macrophages, and Treg, over M1 macrophages, dendritic cells, and effector CD4⁺ and CD8⁺ T lymphocytes. The dynamic and continuously evolving cross-talk between inflammatory and cancer cells might be direct and contact-dependent, but it is mainly mediated by soluble and exosomes-carried cytokines. Among these, tumor necrosis factor alpha (TNFα) plays a relevant role in enhancing cancer risk, cancer growth, and cancer-associated cachexia. In this review, we describe the inflammatory cell types, the cytokines, and the mechanisms underlying PDAC risk, growth, and progression, with particular attention on TNFα, also in the light of the potential risks or benefits associated with anti-TNFα treatments.
Collapse
|
30
|
Chlebowski RT, Anderson GL, Manson JE, Prentice RL, Aragaki AK, Snetselaar L, Beresford SAA, Kuller LH, Johnson K, Lane D, Luo J, Rohan TE, Jiao L, Barac A, Womack C, Coday M, Datta M, Thomson CA. Low-Fat Dietary Pattern and Cancer Mortality in the Women's Health Initiative (WHI) Randomized Controlled Trial. JNCI Cancer Spectr 2019; 2:pky065. [PMID: 31360880 PMCID: PMC6649760 DOI: 10.1093/jncics/pky065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/19/2018] [Accepted: 10/03/2018] [Indexed: 01/08/2023] Open
Abstract
Background In the Women’s Health Initiative Dietary Modification trial, a low-fat dietary pattern reduced deaths after breast cancer. Mortality from other cancer sites has not been reported. Methods A low-fat dietary pattern influence on deaths from and after site-specific cancers was examined during 8.5 years (median) of dietary intervention and cumulatively during 17.7 years (median) of follow-up. A total 48 835 postmenopausal women, ages 50–79 years, were randomly assigned from 1993 to 1998 at 40 US clinical centers to dietary intervention (40%, n = 19 541 or a usual diet comparison group (60%, n = 29 294). Dietary intervention influence on mortality from protocol-specified cancers (breast, colon and rectum, endometrium and ovary), individually and as a composite, represented the primary analyses. Results During the dietary intervention period, a reduction in deaths after breast cancer (HR = 0.65 95% CI = 0.45 to 0.94, P = .02) was the only statistically significant cancer mortality finding. During intervention, the HRs for deaths after the protocol-specified cancer composite were 0.90 (95% CI = 0.73 to 1.10) and 0.95 (95% CI = 0.85 to 1.06) for deaths after all cancers. During 17.7 years of follow-up with 3867 deaths after all cancers, reduction in deaths after breast cancer continued in the dietary intervention group (HR = 0.85, 95% CI = 0.74 to 0.99, P = .03). However, no dietary intervention influence on deaths from or after any other cancer or cancer composite was seen. Conclusions A low-fat dietary pattern reduced deaths after breast cancer. No reduction in mortality from or after any other cancer or cancer composite was seen.
Collapse
Affiliation(s)
| | - Garnet L Anderson
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - JoAnn E Manson
- Harvard Medical School, Brigham and Women's Hospital, Boston, MA
| | - Ross L Prentice
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Aaron K Aragaki
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | | | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA
| | - Karen Johnson
- Department of Preventive Medicine, University of Tennessee, Memphis, TN
| | - Dorothy Lane
- Preventive Medicine, Stony Brook University, Stony Brook, NY
| | - Juhua Luo
- School of Public Health, University of Indiana, Bloomington, IN
| | - Thomas E Rohan
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Li Jiao
- Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Ana Barac
- MedStar Health Research Institute and Georgetown/Howard Universities Center for Clinical and Translational Sciences, Washington, DC
| | | | - Mace Coday
- Department of Preventive Medicine, University of Tennessee, Memphis, TN
| | - Mridul Datta
- School of Public Health, University of Purdue, West Lafayette, IN
| | - Cynthia A Thomson
- Mel and Enid Zuckerman College of Public Health, University of Arizona Cancer Center, Tucson, AZ
| |
Collapse
|
31
|
Kovač U, Skubic C, Bohinc L, Rozman D, Režen T. Oxysterols and Gastrointestinal Cancers Around the Clock. Front Endocrinol (Lausanne) 2019; 10:483. [PMID: 31379749 PMCID: PMC6653998 DOI: 10.3389/fendo.2019.00483] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 07/03/2019] [Indexed: 12/24/2022] Open
Abstract
This review focuses on the role of oxidized sterols in three major gastrointestinal cancers (hepatocellular carcinoma, pancreatic, and colon cancer) and how the circadian clock affects the carcinogenesis by regulating the lipid metabolism and beyond. While each field of research (cancer, oxysterols, and circadian clock) is well-studied within their specialty, little is known about the intertwining mechanisms and how these influence the disease etiology in each cancer type. Oxysterols are involved in pathology of these cancers, but final conclusions about their protective or damaging effects are elusive, since the effect depends on the type of oxysterol, concentration, and the cell type. Oxysterol concentrations, the expression of key regulators liver X receptors (LXR), farnesoid X receptor (FXR), and oxysterol-binding proteins (OSBP) family are modulated in tumors and plasma of cancer patients, exposing these proteins and selected oxysterols as new potential biomarkers and drug targets. Evidence about how cholesterol/oxysterol pathways are intertwined with circadian clock is building. Identified key contact points are different forms of retinoic acid receptor related orphan receptors (ROR) and LXRs. RORs and LXRs are both regulated by sterols/oxysterols and the circadian clock and in return also regulate the same pathways, representing a complex interplay between sterol metabolism and the clock. With this in mind, in addition to classical therapies to modulate cholesterol in gastrointestinal cancers, such as the statin therapy, the time is ripe also for therapies where time and duration of the drug application is taken as an important factor for successful therapies. The final goal is the personalized approach with chronotherapy for disease management and treatment in order to increase the positive drug effects.
Collapse
|
32
|
Matejcic M, Lesueur F, Biessy C, Renault AL, Mebirouk N, Yammine S, Keski-Rahkonen P, Li K, Hémon B, Weiderpass E, Rebours V, Boutron-Ruault MC, Carbonnel F, Kaaks R, Katzke V, Kuhn T, Boeing H, Trichopoulou A, Palli D, Agnoli C, Panico S, Tumino R, Sacerdote C, Quirós JR, Duell EJ, Porta M, Sánchez MJ, Chirlaque MD, Barricarte A, Amiano P, Ye W, Peeters PH, Khaw KT, Perez-Cornago A, Key TJ, Bueno-de-Mesquita HB, Riboli E, Vineis P, Romieu I, Gunter MJ, Chajès V. Circulating plasma phospholipid fatty acids and risk of pancreatic cancer in a large European cohort. Int J Cancer 2018; 143:2437-2448. [PMID: 30110135 DOI: 10.1002/ijc.31797] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 02/11/2024]
Abstract
There are both limited and conflicting data on the role of dietary fat and specific fatty acids in the development of pancreatic cancer. In this study, we investigated the association between plasma phospholipid fatty acids and pancreatic cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort. The fatty acid composition was measured by gas chromatography in plasma samples collected at recruitment from375 incident pancreatic cancer cases and375 matched controls. Associations of specific fatty acids with pancreatic cancer risk were evaluated using multivariable conditional logistic regression models with adjustment for established pancreatic cancer risk factors. Statistically significant inverse associations were found between pancreatic cancer incidence and levels of heptadecanoic acid (ORT3-T1 [odds ratio for highest versus lowest tertile] =0.63; 95%CI[confidence interval] = 0.41-0.98; ptrend = 0.036), n-3 polyunsaturated α-linolenic acid (ORT3-T1 = 0.60; 95%CI = 0.39-0.92; ptrend = 0.02) and docosapentaenoic acid (ORT3-T1 = 0.52; 95%CI = 0.32-0.85; ptrend = 0.008). Industrial trans-fatty acids were positively associated with pancreatic cancer risk among men (ORT3-T1 = 3.00; 95%CI = 1.13-7.99; ptrend = 0.029), while conjugated linoleic acids were inversely related to pancreatic cancer among women only (ORT3-T1 = 0.37; 95%CI = 0.17-0.81; ptrend = 0.008). Among current smokers, the long-chain n-6/n-3 polyunsaturated fatty acids ratio was positively associated with pancreatic cancer risk (ORT3-T1 = 3.40; 95%CI = 1.39-8.34; ptrend = 0.007). Results were robust to a range of sensitivity analyses. Our findings suggest that higher circulating levels of saturated fatty acids with an odd number of carbon atoms and n-3 polyunsaturated fatty acids may be related to lower risk of pancreatic cancer. The influence of some fatty acids on the development of pancreatic cancer may be sex-specific and modulated by smoking.
Collapse
Affiliation(s)
- M Matejcic
- International Agency for Research on Cancer, Lyon, France
| | - F Lesueur
- Genetic Epidemiology of Cancer team, Inserm, U900, Paris, France
- Institut Curie, Paris, France
- PSL University, Paris, France
- Mines ParisTech, Fontainebleau, France
| | - C Biessy
- International Agency for Research on Cancer, Lyon, France
| | - A L Renault
- Genetic Epidemiology of Cancer team, Inserm, U900, Paris, France
- Institut Curie, Paris, France
- PSL University, Paris, France
- Mines ParisTech, Fontainebleau, France
| | - N Mebirouk
- Genetic Epidemiology of Cancer team, Inserm, U900, Paris, France
- Institut Curie, Paris, France
- PSL University, Paris, France
- Mines ParisTech, Fontainebleau, France
| | - S Yammine
- International Agency for Research on Cancer, Lyon, France
| | | | - K Li
- International Agency for Research on Cancer, Lyon, France
| | - B Hémon
- International Agency for Research on Cancer, Lyon, France
| | - E Weiderpass
- Genetic Epidemiology Group, Folkhälsan Research Center, Helsinki, Finland
- Department of Community Medicine, Faculty of Health Sciences, University of Tromsø - The Arctic University of Norway, Tromsø, Norway
- Department of Research, Cancer Registry of Norway, Institute of Population-Based Cancer Research, Oslo, Norway
- Public Health Division of Gipuzkoa, BioDonostia Research institute, San Sebastian, Spain
| | - V Rebours
- Department of Gastroenterology and Pancreatology, Beaujon Hospital, University Paris 7, Clichy, France
| | - M C Boutron-Ruault
- INSERM, Centre for Research in Epidemiology and Population Health, U1018, Health across Generations Team, Institut Gustave Roussy, Villejuif, France
- Université Paris Sud, UMRS, Villejuif, France
| | - F Carbonnel
- INSERM, Centre for Research in Epidemiology and Population Health, U1018, Health across Generations Team, Institut Gustave Roussy, Villejuif, France
- Université Paris Sud, UMRS, Villejuif, France
- Department of Gastroenterology, Bicêtre University Hospital, Assistance Publique des Hôpitaux de Paris, Le Kremlin Bicêtre, France
| | - R Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - V Katzke
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - T Kuhn
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - H Boeing
- Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Nuthetal, Germany
| | - A Trichopoulou
- Hellenic Health Foundation, Athens, Greece
- WHO Collaborating Center for Nutrition and Health, Unit of Nutritional Epidemiology and Nutrition in Public Health, Department of Hygiene, Epidemiology and Medical Statistics, University of Athens Medical School, Athens, Greece
| | - D Palli
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute - ISPO, Florence, Italy
| | - C Agnoli
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Panico
- Clinical Medicine and Surgery Department, Università degli Studi di Napoli Federico II, Naples, Italy
| | - R Tumino
- Cancer Registry and Histopathology Department, ASP, "Civic - M.P. Arezzo" Hospital, Ragusa, Italy
| | - C Sacerdote
- Unit of Cancer Epidemiology, Citta' della Salute e della Scienza Hospital, University of Turin and Centre for Cancer Prevention (CPO), Turin, Italy
| | - J R Quirós
- EPIC Asturias, Public Health Directorate, Asturias, Spain
| | - E J Duell
- Unit of Nutrition and Cancer, Catalan Institute of Oncology (ICO-IDIBELL), Barcelona, Spain
| | - M Porta
- Hospital del Mar Research Institute - IMIM, CIBER Epidemiología y Salud Pública (CIBERESP) and Universitat Autònoma de Barcelona, Barcelona, Spain
| | - M J Sánchez
- Escuela Andaluza de Salud Pública. Instituto de Investigación Biosanitaria ibs.GRANADA. Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - M D Chirlaque
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Epidemiology, Regional Health Council, IMIB-Arrixaca, Murcia, Spain
- Department of Health and Social Sciences, Universidad de Murcia, Murcia, Spain
| | - A Barricarte
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Navarra Public Health Institute, Pamplona, Spain
| | - P Amiano
- Public Health Division of Gipuzkoa, BioDonostia Research institute, San Sebastian, Spain
- CIBER in Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - W Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- The Medical Biobank at Umeå University, Umeå, Sweden
| | - P H Peeters
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - K T Khaw
- University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - A Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - T J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - H B Bueno-de-Mesquita
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
- Department of Social & Preventive Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - E Riboli
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College, London, United Kingdom
| | - P Vineis
- MRC-PHE Center for Environment and Health, School of Public Health, Imperial College, London, United Kingdom
| | - I Romieu
- International Agency for Research on Cancer, Lyon, France
| | - M J Gunter
- International Agency for Research on Cancer, Lyon, France
| | - V Chajès
- International Agency for Research on Cancer, Lyon, France
| |
Collapse
|
33
|
Nadella S, Burks J, Al-Sabban A, Inyang G, Wang J, Tucker RD, Zamanis ME, Bukowski W, Shivapurkar N, Smith JP. Dietary fat stimulates pancreatic cancer growth and promotes fibrosis of the tumor microenvironment through the cholecystokinin receptor. Am J Physiol Gastrointest Liver Physiol 2018; 315:G699-G712. [PMID: 29927319 PMCID: PMC6293257 DOI: 10.1152/ajpgi.00123.2018] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The gastrointestinal peptide cholecystokinin (CCK) is released from the duodenum in response to dietary fat to aid in digestion, and plasma CCK levels are elevated with the consumption of high-fat diets. CCK is also a trophic peptide for the pancreas and has also been shown to stimulate growth of pancreatic cancer. In the current investigation, we studied the influence of a diet high in saturated fat on the growth of pancreatic cancer in syngeneic murine models before the mice became obese to exclude the confounding factors associated with obesity. The high-fat diet significantly increased growth and metastasis of pancreatic cancer compared with the control diet, and the stimulatory effect was blocked by the CCK-receptor antagonist proglumide. We then selectively knocked out the CCK receptor on the pancreatic cancer cells using clustered regularly interspaced short palindromic repeats technology and showed that without CCK-receptors, dietary fat was unable to stimulate cancer growth. We next demonstrated that dietary fat failed to influence pancreatic cancer xenograft growth in genetically engineered CCK peptide knockout mice. The tumor-associated fibrosis that is so prevalent in the pancreatic cancer microenvironment was significantly decreased with CCK-receptor antagonist therapy because fibroblasts also have CCK receptors. The CCK-receptor antagonist proglumide also altered tumor metalloprotease expression and increased tumor suppressor genes by a PCR array. Our studies confirm that a diet high in saturated fat promotes growth of pancreatic cancer and the action is mediated by the CCK-receptor pathway. NEW & NOTEWORTHY Diets high in long-chain saturated fats promote growth of pancreatic cancer independent of obesity. The mechanism through which dietary fat promotes cancer is mediated through the cholecystokinin (CCK) receptor pathway. Therapy with a CCK-receptor antagonist altered the tumor microenvironment by reducing fibrosis, increasing cluster of differentiation 8+ lymphocytes, increasing tumor suppressor genes, and thus decreasing metastases. Use of CCK-receptor antagonist therapy with standard chemotherapy for pancreatic cancer may improve response by altering the tumor microenvironment.
Collapse
Affiliation(s)
- Sandeep Nadella
- 1Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Julian Burks
- 2Department of Oncology, Georgetown University, Washington, District of Columbia
| | | | - Gloria Inyang
- 3Department of Biochemistry, Georgetown University, Washington, District of Columbia
| | - Juan Wang
- 1Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Robin D. Tucker
- 4Department of Comparative Medicine, Georgetown University, Washington, District of Columbia
| | - Marie E. Zamanis
- 2Department of Oncology, Georgetown University, Washington, District of Columbia
| | - William Bukowski
- 1Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Narayan Shivapurkar
- 1Department of Medicine, Georgetown University, Washington, District of Columbia
| | - Jill P. Smith
- 1Department of Medicine, Georgetown University, Washington, District of Columbia,2Department of Oncology, Georgetown University, Washington, District of Columbia
| |
Collapse
|
34
|
Ulrich CM, Himbert C, Holowatyj AN, Hursting SD. Energy balance and gastrointestinal cancer: risk, interventions, outcomes and mechanisms. Nat Rev Gastroenterol Hepatol 2018; 15:683-698. [PMID: 30158569 PMCID: PMC6500387 DOI: 10.1038/s41575-018-0053-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Obesity increases the risk of multiple gastrointestinal cancers and worsens disease outcomes. Conversely, strong inverse associations have emerged between physical activity and colon cancer and possibly other gastrointestinal malignancies. The effect of weight loss interventions - such as modifications of diet and/or physical activity or bariatric surgery - remains unclear in patients who are obese and have gastrointestinal cancer, although large clinical trials are underway. Human intervention studies have already shed light on potential mechanisms underlying the energy balance-cancer relationship, with preclinical models supporting emerging pathway effects. Central to interventions that reduce obesity or increase physical activity are pluripotent cancer-preventive effects (including reduced systemic and adipose tissue inflammation and angiogenesis, altered adipokine levels and improved insulin resistance) that directly interface with the hallmarks of cancer. Other mechanisms, such as DNA repair, oxidative stress and telomere length, immune function, effects on cancer stem cells and the microbiome, could also contribute to energy balance effects on gastrointestinal cancers. Although some mechanisms are well understood (for instance, systemic effects on inflammation and insulin signalling), other areas remain unclear. The current state of knowledge supports the need to better integrate mechanistic approaches with preclinical and human studies to develop effective, personalized diet and exercise interventions to reduce the burden of obesity on gastrointestinal cancer.
Collapse
Affiliation(s)
- Cornelia M. Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT, USA.,Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA.,
| | - Caroline Himbert
- Huntsman Cancer Institute, Salt Lake City, UT, USA.,Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Andreana N. Holowatyj
- Huntsman Cancer Institute, Salt Lake City, UT, USA.,Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Stephen D. Hursting
- Department of Nutrition, University of North Carolina, Chapel Hill, NC, USA.,UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC, USA
| |
Collapse
|
35
|
Ligibel JA. Is It Time to Give Breast Cancer Patients a Prescription for a Low-Fat Diet? JNCI Cancer Spectr 2018; 2:pky066. [PMID: 31360881 PMCID: PMC6649733 DOI: 10.1093/jncics/pky066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/22/2018] [Indexed: 11/27/2022] Open
|
36
|
Botteri E, de Lange T, Tonstad S, Berstad P. Exploring the effect of a lifestyle intervention on cancer risk: 43-year follow-up of the randomized Oslo diet and antismoking study. J Intern Med 2018; 284:282-291. [PMID: 29790221 DOI: 10.1111/joim.12765] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND/OBJECTIVES The Oslo diet and antismoking study showed that counselling for a healthy lifestyle reduced lifelong coronary mortality in high-risk men. We explored whether the same counselling reduced also cancer risk. METHODS The study randomly allocated males at high coronary risk to either a 5-year intervention for lifestyle changes (cholesterol-lowering dietary changes, weight loss and stopping smoking) or a control group (1 : 1) in 1972/73. We explored the incidence and mortality of all cancers and cancer forms related to smoking, BMI or diet up to 43 years after randomization. RESULTS A total of 595 men in the intervention and 621 in the control group were included. At inclusion median age was 45 years, 588 (48.4%) subjects were overweight (BMI > 25 kg m-2 ) and 925 (76.1%) current smokers. The intervention did not reduce the risk of cancer after 43 years (adjusted hazard ratio (HR) 0.96, 95% confidence interval (CI) 0.80-1.15). In the first 25 years of follow-up, among the 1088 (89.5%) men who were overweight/obese and/or smokers, the intervention reduced the incidence of those cancer forms related to smoking, BMI or diet (including carcinoma of the respiratory, digestive and urinary tracts; adjusted HR 0.69; 95% CI 0.49-0.99). The intervention had no significant effect on incidence beyond 25 years, or on mortality. CONCLUSIONS The 5-year counselling for a healthy lifestyle did not reduce the overall cancer risk in the very long term. However, in the first 25 years, the counselling reduced the risk of relevant cancer types in overweight/obese subjects and smokers.
Collapse
Affiliation(s)
- E Botteri
- Department of Bowel cancer screening, Cancer Registry of Norway, Oslo, Norway.,Norwegian National Advisory Unit for Women's Health, Women's Clinic, Oslo University Hospital, Oslo, Norway
| | - T de Lange
- Department of Bowel cancer screening, Cancer Registry of Norway, Oslo, Norway.,Unit for Research, Innovation and Education, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - S Tonstad
- Department of Preventive Cardiology, Oslo University Hospital, Oslo, Norway.,School of Public Health, Loma Linda University, Loma Linda, CA, USA
| | - P Berstad
- Department of Bowel cancer screening, Cancer Registry of Norway, Oslo, Norway
| |
Collapse
|
37
|
Abstract
Commensal microbes engineered to convert natural compounds found in cruciferous vegetables into molecules with anticancer properties prevent carcinogenesis and cause the regression of colorectal cancer in mice fed with a vegetable diet.
Collapse
Affiliation(s)
- David A Drew
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
- Broad Institute, Cambridge, MA, USA.
| |
Collapse
|
38
|
Stolzenberg-Solomon RZ, Katki HA. Is the Women’s Health Initiative (WHI) Dietary Modification Associated With a Reduced Risk of Pancreatic Cancer? J Natl Cancer Inst 2018; 110:4085223. [DOI: 10.1093/jnci/djx139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/06/2017] [Indexed: 11/14/2022] Open
Affiliation(s)
- Rachael Z Stolzenberg-Solomon
- Metabolic Epidemiology Branch and Biostatics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville MD
| | - Hormuzd A Katki
- Metabolic Epidemiology Branch and Biostatics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville MD
| |
Collapse
|