1
|
Peduzzi G, Archibugi L, Farinella R, de Leon Pisani RP, Vodickova L, Vodicka P, Kraja B, Sainz J, Bars-Cortina D, Daniel N, Silvestri R, Uysal-Onganer P, Landi S, Dulińska-Litewka J, Comandatore A, Campa D, Hughes DJ, Rizzato C. The exposome and pancreatic cancer, lifestyle and environmental risk factors for PDAC. Semin Cancer Biol 2025:S1044-579X(25)00061-6. [PMID: 40368260 DOI: 10.1016/j.semcancer.2025.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 04/08/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Pancreatic cancer (PC), particularly pancreatic ductal adenocarcinoma (PDAC), is a significant global health issue with high mortality rates. PDAC, though only 3% of cancer diagnoses, causes 7% of cancer deaths due to its severity and asymptomatic early stages. Risk factors include lifestyle choices, environmental exposures, and genetic predispositions. Conditions like new-onset type 2 diabetes and chronic pancreatitis also contribute significantly. Modifiable risk factors include smoking, alcohol consumption, non-alcoholic fatty pancreatic disease (NAFPD), and obesity. Smoking and heavy alcohol consumption increase PC risk, while NAFPD and obesity, particularly central adiposity, contribute through chronic inflammation and insulin resistance. Refined sugar and sugar-sweetened beverages (SSBs) are also linked to increased PC risk, especially among younger individuals. Hormonal treatments and medications like statins, aspirin, and metformin have mixed results on PC risk, with some showing protective effects. The gut microbiome influences PC through the gut-pancreas axis, with disruptions leading to inflammation and carcinogenesis. Exposure to toxic substances, including heavy metals and chemicals, is associated with increased PC risk. Glycome changes, such as abnormal glycosylation patterns, are significant in PDAC development and offer potential for early diagnosis. Interactions between environmental and genetic factors are crucial in PDAC susceptibility. Genome-wide association studies (GWAS) have identified several single nucleotide polymorphisms (SNPs) linked to PDAC, but gene-environment interactions remain largely unexplored. Future research should focus on polygenic risk scores (PRS) and large-scale studies to better understand these interactions and their impact on PDAC risk.
Collapse
Affiliation(s)
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ruggero Ponz de Leon Pisani
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Ludmila Vodickova
- Biomedical Center Martin, Bioinformatic Center, Comenius University in Bratislava, Jessenius Faculty of Medicine in Martin, Slovakia; Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Pavel Vodicka
- Faculty of Medicine and Biomedical Center in Pilsen, Charles University, Pilsen, Czech Republic
| | - Bledar Kraja
- University Clinic of Gastrohepatology, University Hospital Center Mother Teresa, Tirana, Albania
| | - Juan Sainz
- Department of Biochemistry and Molecular Biology, University of Granada, Granada, Spain; Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain; GENYO. Centre for Genomics and Oncological Research. Genomic Oncology department. Granada, Spain; Instituto de Investigación Biosanitaria Ibs.Granada, Granada, Spain
| | - David Bars-Cortina
- Institut Català d'Oncologia (ICO) IDIBELL, Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain; Institut Català d'Oncologia (ICO) IDIBELL, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | | - Pinar Uysal-Onganer
- Cancer Mechanisms and Biomarkers Research Group, School of Life Sciences, University of Westminster, London, UK
| | - Stefano Landi
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Annalisa Comandatore
- General Surgery Unit, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Italy
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
2
|
Yang H, Chou W, Nguyen P, Nguyen NTH, Phuong NT, Wang C, Hsu JC, Lin M, Huang C. The protective role of anti-parkinsonian drugs in pancreatic cancer risk: A comprehensive case-control study in Taiwan. Cancer Sci 2025; 116:783-791. [PMID: 39629516 PMCID: PMC11875767 DOI: 10.1111/cas.16422] [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: 09/09/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 03/05/2025] Open
Abstract
Pancreatic cancer is among the deadliest cancers, with a grim prognosis despite advances in treatment. We conducted a population-based case-control study from Taiwan, linking Health and Welfare Data Science Center data to the Taiwan Cancer Registry, which offers a promising strategy for its treatment through drug repurposing. The study aims to identify the association of anti-parkinsonian drugs with pancreatic cancer risk across different age groups. The analysis encompassed 18,921 pancreatic cancer cases and 75,684 matched controls, employing conditional logistic regression to assess the impact of anti-parkinsonian drugs on the risk of pancreatic cancer. Key findings revealed a statistically significant association of the administration with specific anti-parkinsonian medications, including anticholinergic agents, tertiary amines, dopa derivatives, and dopamine receptor agonists, with a reduction in pancreatic cancer risk. These associations were represented as adjusted odds ratios (aORs), ranging from 0.620 (95% CI 0.470-0.810) to 0.764 (95% CI 0.655-0.891). Further, age-stratified analysis revealed variations in efficacy across different age groups. Anticholinergic agents and tertiary amines exhibited greater effectiveness in the 40-64-year age group (aOR, 0.653; 95% CI, 0.489-0.872), whereas dopa derivatives and dopamine receptor agonists were particularly efficacious in the cohort aged ≥65 years (aOR, 0.728; 95% CI, 0.624-0.850 and aOR, 0.665; 95% CI, 0.494-0.894, respectively). Notably, specific drugs such as trihexyphenidyl, levodopa/dopa decarboxylase inhibitor (DDCI), and pramipexole demonstrated a significant decrease in cancer risk, especially in the elderly population. These preliminary findings can contribute to the possible therapeutic role of anti-parkinsonian drugs in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Hsuan‐Chia Yang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Research Center of Big Data and Meta‐Analysis, Wan Fang HospitalTaipei Medical UniversityTaipeiTaiwan
| | - Wen‐Chi Chou
- Department of Hematology and Oncology, Chang Gung Memorial Hospital at Linkou and College of MedicineChang Gung UniversityTaoyuanTaiwan
| | - Phung‐Anh Nguyen
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Graduate Institute of Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Nhi Thi Hong Nguyen
- School of Nutrition and Health SciencesTaipei Medical UniversityTaipeiTaiwan
- Health Personnel Training InstituteUniversity of Medicine and Pharmacy, Hue UniversityHueVietnam
| | - Nguyen Thi Phuong
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Ching‐Huan Wang
- Biomedical Informatics and Data Science (BIDS) Section, School of MedicineJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jason C. Hsu
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
- Clinical Data Center, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
- Research Center of Health Care Industry Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- International Ph.D. Program in Biotech and Healthcare Management, College of ManagementTaipei Medical UniversityTaipeiTaiwan
| | - Ming‐Chin Lin
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Department of Neurosurgery, Wang‐Fang HospitalTaipei Medical UniversityTaipeiTaiwan
- Taipei Neuroscience InstituteTaipei Medical UniversityTaipeiTaiwan
| | - Chih‐Wei Huang
- Graduate Institute of Biomedical Informatics, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- International Center for Health Information Technology (ICHIT), College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
- Clinical Big Data Research Center, Taipei Medical University HospitalTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
3
|
Lauri G, Mills K, Majumder S, Capurso G. The exposome as a target for primary prevention and a tool for early detection of pancreatic cancer. Best Pract Res Clin Gastroenterol 2025; 74:101991. [PMID: 40210335 DOI: 10.1016/j.bpg.2025.101991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/04/2025] [Accepted: 02/11/2025] [Indexed: 04/12/2025]
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) is a highly aggressive malignancy with limited survival due to late stage diagnosis and scarce therapeutic options. Emerging evidence highlights the role of the "exposome," which encompasses environmental, lifestyle, and metabolic exposures, as a crucial determinant in PDAC risk and a potential avenue for early detection. This review synthesizes findings on modifiable risk factors, including smoking, obesity, diabetes, diet, and alcohol consumption, and their interplay with genetic and metabolic profiles in PDAC development. Additionally, we explore cutting-edge approaches in exposomic research, such as biobanking, electronic health record analysis, and AI-driven predictive models, to identify early biomarkers and stratify high-risk populations. This integrated framework aims to inform prevention strategies and improve early detection of PDAC.
Collapse
Affiliation(s)
- Gaetano Lauri
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Krystal Mills
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Gabriele Capurso
- Pancreatico-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
4
|
Zhan Y, Zhang K, Fan Y, Lin S, Wu J, Xu H. Lipids, lipid-lowering drug target genes and pancreatic cancer: a Mendelian randomization study. Int J Clin Pharm 2025:10.1007/s11096-025-01866-7. [PMID: 39821006 DOI: 10.1007/s11096-025-01866-7] [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: 09/20/2024] [Accepted: 01/04/2025] [Indexed: 01/19/2025]
Abstract
BACKGROUND Pancreatic cancer (PC) is a malignant tumor with a low survival rate. Lipid modifiers show potential for PC therapy, but evidence is lacking. AIM This Mendelian randomization (MR) study aimed to explore the relationship between lipid traits, and lipid-lowering drug target genes with PC risk. METHOD Genetic instrumental variables associated with lipid traits and lipid-lowering drug target genes were used to perform MR analyses of PC risk. MR estimation was based on genome-wide association study data from two large sample sets, and the MR results were meta-analyzed to assess their impact on PC risk. To ensure the reliability of lipid-modifying drug targets, we conducted a Summary Data-based Mendelian Randomization (SMR) analysis. Additionally, a two-step MR analysis was employed to explore potential mediating effects. RESULTS In two independent datasets, HMG-CoA reductase (HMGCR) inhibition was statistically associated with a lower risk of PC (OR 0.50, [95% CI 0.25-1.00]; p = 0.0453). The results were further supported by SMR analysis, which showed a similar association (OR 0.51, [95% CI 0.28-0.96]; p = 0.0369). Mediation analysis revealed that 11.69% of the protective effect of HMGCR inhibitors on PC is mediated through lower BMI levels. No significant effect of lipid traits and the other eight lipid-lowering drug targets on PC risk was found. CONCLUSION This study suggests that HMGCR may be a potential drug target for the treatment or prevention of PC, providing important insights into the use of lipid-targeted drugs in PC therapy.
Collapse
Affiliation(s)
- Yuxuan Zhan
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Kai Zhang
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Yiqun Fan
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Cancer Institute, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Siyi Lin
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Jian Wu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China
| | - Hongxia Xu
- School of Public Health and Institute of Wenzhou and Liangzhu Laboratory, Zhejiang University, Hangzhou, 310058, China.
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| |
Collapse
|
5
|
Mantovani A, Lonardo A, Stefan N, Targher G. Metabolic dysfunction-associated steatotic liver disease and extrahepatic gastrointestinal cancers. Metabolism 2024; 160:156014. [PMID: 39182602 DOI: 10.1016/j.metabol.2024.156014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/09/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) poses a significant and ever-increasing health and economic burden worldwide. Substantial epidemiological evidence shows that MASLD is a multisystem disease that is associated not only with liver-related complications but is also associated with an increased risk of developing cardiometabolic comorbidities and extrahepatic cancers (principally gastrointestinal [GI] cancers). GI cancers account for a quarter of the global cancer incidence and a third of cancer-related deaths. In this narrative review, we provide an overview of the literature on (a) the epidemiological data on the risk of non-liver GI cancers in MASLD, (b) the putative mechanisms by which MASLD (and factors linked with MASLD) may increase this risk, and (c) the possible pharmacotherapies beneficially affecting both MASLD and extrahepatic GI cancer risk. There are multiple potential pathophysiological mechanisms by which MASLD may increase extrahepatic GI cancer risk. Although further studies are needed, the current evidence supports a possible extrahepatic carcinogenic role for MASLD, regardless of obesity and diabetes status, thus highlighting the potential role of tailoring cancer screening for individuals with MASLD. Although there are conflicting data in the literature, aspirin, statins and metformin appear to exert some chemo-preventive effects against GI cancer.
Collapse
Affiliation(s)
- Alessandro Mantovani
- Section of Endocrinology, Diabetes and Metabolism, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Amedeo Lonardo
- Department of Internal Medicine, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Norbert Stefan
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany; Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich, Tübingen, Germany
| | - Giovanni Targher
- Department of Medicine, University of Verona, Italy; Metabolic Diseases Research Unit, IRCCS Sacro Cuore - Don Calabria Hospital, Negrar di Valpolicella, Italy.
| |
Collapse
|
6
|
Joliat GR, Gaspar-Figueiredo S, Labgaa I, Vrochides D, Perinel J, Adham M, Demartines N, Schäfer M. Impact of preoperative statin medication on long-term outcomes after pancreatoduodenectomy for ductal adenocarcinoma: an international multicentric cross-sectional study. J Cancer Res Clin Oncol 2023; 149:17007-17014. [PMID: 37740766 PMCID: PMC10657279 DOI: 10.1007/s00432-023-05426-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023]
Abstract
PURPOSE Statin treatment has been shown in certain population studies and meta-analyses to improve survival of patients with pancreatic ductal adenocarcinoma (PDAC). This study assessed if patients with statin treatment had better overall survival (OS) and disease-free survival (DFS) after upfront pancreatoduodenectomy for PDAC. METHODS Consecutive PDAC patients were retrospectively collected from three centers in Europe and USA (study period: 2000-2017). Adult patients who underwent upfront pancreatoduodenectomy and survived the first 90 postoperative days were included. Patients with metastasis at diagnosis or with macroscopic incomplete resection were excluded. Patients were considered under statin if started at least one month before pancreatoduodenectomy. Survival rates were calculated using Kaplan-Meier method and compared with log-rank test. RESULTS A total of 496 patients were included. Median age was 67 years (IQR 59-75), 48% (n = 236) were women, and 141 patients (28%) received statin treatment already preoperatively. Patients with and without statin treatment were comparable in terms of demographics and pre-/intraoperative characteristics, except for age and pre-existing diabetes. Median OS and DFS were similar in patients with and without statin treatment (OS: 29, 95% CI 22-36 vs. 27 months, 95% CI 22-32, p = 0.370, DFS: 18, 95% CI 14-22 vs. 16 months, 95% CI 14-18, p = 0.430). On multivariable Cox regression, lymph node involvement (HR 1.9, 95% CI 1.6-2.2, p < 0.001), tumor differentiation (HR 1.3, 95% CI 1.1-1.6, p = 0.003), and postoperative chemotherapy (HR 0.5, 95% CI 0.4-0.7, p < 0.001) were predictors of OS, whereas statin treatment was not a prognostic factor (HR 0.9, 95% CI 0.7-1.2, p = 0.376). CONCLUSION In this international cohort of PDAC patients, statin treatment did not influence survival after upfront pancreatoduodenectomy. Nodal involvement, tumor differentiation, and postoperative chemotherapy were independent predictors of OS.
Collapse
Affiliation(s)
- Gaëtan-Romain Joliat
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland.
- Graduate School of Health Sciences, University of Bern, Bern, Switzerland.
| | - Sérgio Gaspar-Figueiredo
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Ismail Labgaa
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Dionisios Vrochides
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Carolinas Medical Center, Charlotte, USA
| | - Julie Perinel
- Department of Surgery, Edouard Herriot Hospital, Lyon, France
| | - Mustapha Adham
- Department of Surgery, Edouard Herriot Hospital, Lyon, France
| | - Nicolas Demartines
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| | - Markus Schäfer
- Department of Visceral Surgery, Lausanne University Hospital CHUV, University of Lausanne (UNIL), Rue du Bugnon 46, 1011, Lausanne, Switzerland
| |
Collapse
|
7
|
Chen Z, Wu P, Wang J, Chen P, Fang Z, Luo F. The association of statin therapy and cancer: a meta-analysis. Lipids Health Dis 2023; 22:192. [PMID: 37950285 PMCID: PMC10636977 DOI: 10.1186/s12944-023-01955-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/24/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND Statins are routinely prescribed to lower cholesterol and have been demonstrated to have significant benefits in atherosclerotic cardiovascular disease. However, whether statin therapy has effects on cancer risk remains controversial. In this study, we investigated the influence of statin therapy on cancer incidence and mortality by conducting a comprehensive meta-analysis of randomized controlled trials. METHODS Systematic searches by Cochrane, Embase, Medline, and PubMed were performed to locate data from eligible randomized controlled trials related to statin therapy and oncology. Our main endpoints were cancer incidence and mortality. Fixed-effects models were used in this study. RESULTS This meta-analysis comprised thirty-five randomized controlled studies. Twenty-eight included studies reported cancer incidence, and eighteen reported cancer mortality. The pooled results indicated no reduction in cancer incidence with statins compared to placebo [OR = 0.99, 95% CI (0.95, 1.03)]. In addition, statins did not decrease cancer mortality [OR = 0.99, 95% CI (0.91, 1.07)]. This study also performed a number of subgroup analyses, which showed no effect of statins on cancer subtypes such as genitourinary and breast cancer. Neither the type of statin nor long-term treatment with statins had an effect on cancer incidence and mortality. CONCLUSION Through comprehensive analysis, we found that statin therapy does not reduce cancer incidence or mortality while protecting the cardiovascular system. TRIAL REGISTRATION Prospero CRD42022377871.
Collapse
Affiliation(s)
- Zijian Chen
- Research Institute of Blood Lipid and Atherosclerosis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- School of Medicine, Hunan University of Medicine, Huaihua, 418000, Hunan, China
| | - Panyun Wu
- Research Institute of Blood Lipid and Atherosclerosis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiangang Wang
- Department of Health Management, Central South University, The Third Xiangya Hospital, Changsha, 410013, Hunan, China
| | - Pengfei Chen
- Research Institute of Blood Lipid and Atherosclerosis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Zhenfei Fang
- Research Institute of Blood Lipid and Atherosclerosis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Fei Luo
- Research Institute of Blood Lipid and Atherosclerosis, the Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
8
|
Aguilar-Valdés A, González-Vela F, Sánchez-Vidal H, Martínez-Aguilar J. A proteomic signature and potential pharmacological opportunities in the adaptive resistance to MEK and PI3K kinase inhibition in pancreatic cancer cells. Proteomics 2023; 23:e2300041. [PMID: 37140101 DOI: 10.1002/pmic.202300041] [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: 01/30/2023] [Revised: 03/23/2023] [Accepted: 04/18/2023] [Indexed: 05/05/2023]
Abstract
Pancreatic cancer is one of the most lethal cancer types and is becoming a leading cause of cancer-related deaths. The limited benefit offered by chemotherapy agents has propelled the search for alternative approaches that target specific molecular drivers of cancer growth and progression. Mutant KRas and effector pathways Raf/MEK/ERK and PI3K/Akt are key players in pancreatic cancer; however, preclinical studies have shown adaptive tumour response to combined MEK and PI3K kinase inhibition leading to treatment resistance. There is a critical unmet need to decipher the molecular basis underlying adaptation to this targeted approach. Here, we aimed to identify common protein expression alterations associated with adaptive resistance in KRas-mutant pancreatic cancer cells, and test if it can be overcome by selected already available small molecule drugs. We found a group of 14 proteins with common expression change in resistant cells, including KRas, caveolin-1, filamin-a, eplin, IGF2R and cytokeratins CK-8, -18 and -19. Notably, several proteins have previously been observed in pancreatic cancer cells with intrinsic resistance to the combined kinase inhibition treatment, suggesting a proteomic signature. We also found that resistant cells are sensitive to small molecule drugs ERK inhibitor GDC-0994, S6K1 inhibitor DG2 and statins.
Collapse
Affiliation(s)
- Alain Aguilar-Valdés
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Francisco González-Vela
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Hilda Sánchez-Vidal
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Juan Martínez-Aguilar
- Red de Apoyo a la Investigación, Coordinación de la Investigación Científica, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
9
|
Petrov MS. Fatty change of the pancreas: the Pandora's box of pancreatology. Lancet Gastroenterol Hepatol 2023; 8:671-682. [PMID: 37094599 DOI: 10.1016/s2468-1253(23)00064-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 04/26/2023]
Abstract
Prevention of common diseases of the pancreas or interception of their progression is as attractive in theory as it is elusive in practice. The fundamental challenge has been an incomplete understanding of targets coupled with a multitude of intertwined factors that are associated with the development of pancreatic diseases. Evidence over the past decade has shown unique morphological features, distinctive biomarkers, and complex relationships of intrapancreatic fat deposition. Fatty change of the pancreas has also been shown to affect at least 16% of the global population. This knowledge has solidified the pivotal role of fatty change of the pancreas in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. The pancreatic diseases originating from intrapancreatic fat (PANDORA) hypothesis advanced in this Personal View cuts across traditional disciplinary boundaries with a view to tackling these diseases. New holistic understanding of pancreatic diseases is well positioned to propel pancreatology through lasting research breakthroughs and clinical advances.
Collapse
Affiliation(s)
- Maxim S Petrov
- School of Medicine, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
10
|
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: 0.5] [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
|
11
|
Su CH, Islam MM, Jia G, Wu CC. Statins and the Risk of Gastric Cancer: A Systematic Review and Meta-Analysis. J Clin Med 2022; 11:jcm11237180. [PMID: 36498753 PMCID: PMC9739712 DOI: 10.3390/jcm11237180] [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: 10/20/2022] [Revised: 11/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Previous epidemiological studies have reported that the use of statins is associated with a decreased risk of gastric cancer, although the beneficial effects of statins on the reduction of gastric cancer remain unclear. Therefore, we conducted a systematic review and meta-analysis to investigate the association between the use of statins and the risk of gastric cancer. Electronic databases such as PubMed, EMBASE, Scopus, and Web of Science were searched between 1 January 2000 and 31 August 2022. Two authors used predefined selection criteria to independently screen all titles, abstracts, and potential full texts. Observational studies (cohort and case-control) or randomized control trials that assessed the association between statins and gastric cancer were included in the primary and secondary analyses. The pooled effect sizes were calculated using the random-effects model. The Meta-analysis of Observational Studies in Epidemiology (MOOSE) reporting guidelines were followed to conduct this study. The total sample size across the 20 included studies was 11,870,553. The use of statins was associated with a reduced risk of gastric cancer (RRadjusted: 0.72; 95%CI: 0.64−0.81, p < 0.001). However, the effect size of statin use on the risk of gastric cancer was lower in Asian studies compared to Western studies (RRAsian: 0.62; 95%CI: 0.53−0.73 vs. RRwestern: 0.88; 95%CI: 0.79−0.99). These findings suggest that the use of statins is associated with a reduced risk of gastric cancer. This reverse association was even stronger among Asian people than Western individuals.
Collapse
Affiliation(s)
- Chun-Hsien Su
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 111396, Taiwan
- Graduate Institute of Sports Coaching Science, College of Kinesiology and Health, Chinese Culture University, Taipei 111396, Taiwan
| | - Md. Mohaimenul Islam
- International Center for Health Information Technology (ICHIT), Taipei Medical University, Taipei 111396, Taiwan
| | - Guhua Jia
- Sports Teaching Department, Shanghai University of Medicine & Health Sciences, Shanghai 201318, China
| | - Chieh-Chen Wu
- Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, Taipei 111396, Taiwan
- Correspondence:
| |
Collapse
|
12
|
Ejam SS, Saleh RO, Catalan Opulencia MJ, Najm MA, Makhmudova A, Jalil AT, Abdelbasset WK, Al-Gazally ME, Hammid AT, Mustafa YF, Sergeevna SE, Karampoor S, Mirzaei R. Pathogenic role of 25-hydroxycholesterol in cancer development and progression. Future Oncol 2022; 18:4415-4442. [PMID: 36651359 DOI: 10.2217/fon-2022-0819] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/06/2022] [Indexed: 01/19/2023] Open
Abstract
Cholesterol is an essential lipid that serves several important functions, including maintaining the homeostasis of cells, acting as a precursor to bile acid and steroid hormones and preserving the stability of membrane lipid rafts. 25-hydroxycholesterol (25-HC) is a cholesterol derivative that may be formed from cholesterol. 25-HC is a crucial component in various biological activities, including cholesterol metabolism. In recent years, growing evidence has shown that 25-HC performs a critical function in the etiology of cancer, infectious diseases and autoimmune disorders. This review will summarize the latest findings regarding 25-HC, including its biogenesis, immunomodulatory properties and role in innate/adaptive immunity, inflammation and the development of various types of cancer.
Collapse
Affiliation(s)
| | - Raed Obaid Saleh
- Department of Pharmacy, Al-Maarif University College, Al-Anbar, Iraq
| | | | - Mazin Aa Najm
- Pharmaceutical Chemistry Department, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Aziza Makhmudova
- Department of Social Sciences & Humanities, Samarkand State Medical Institute, Samarkand, Uzbekistan
- Department of Scientific Affairs, Tashkent State Dental Institute, Makhtumkuli Street 103, Tashkent, 100047, Uzbekistan
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq
| | - Walid Kamal Abdelbasset
- Department of Health & Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia
- Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | | | - Ali Thaeer Hammid
- Computer Engineering Techniques Department, Faculty of Information Technology, Imam Ja'afar Al-Sadiq University, Baghdad, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Sergushina Elena Sergeevna
- National Research Ogarev Mordovia State University, 68 Bolshevitskaya Street, Republic of Mordovia, Saransk, 430005, Russia
| | - Sajad Karampoor
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Venom & Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
13
|
Miyaki C, Lynch LM. An Update on Common Pharmaceuticals in the Prevention of Pancreatic Cancer. Cureus 2022; 14:e25496. [PMID: 35800820 PMCID: PMC9246430 DOI: 10.7759/cureus.25496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 01/03/2023] Open
Abstract
In this review, we aim to update readers about the most recent studies on common pharmaceuticals and their association with pancreatic cancer risk. The use of prophylactic aspirin, metformin, beta-blockers, and statins has been studied in the past but showed inconclusive results in the reduction of pancreatic cancer incidence. However, in recent studies, these medications along with combination therapy of aspirin and metformin were found to have a more significant association with decreasing risk. Given the poor prognosis of pancreatic cancer despite treatment, medication prophylaxis prevention should be considered. In this review, we hope to encourage future case-control or prospective studies on common medications that have shown great potential in delaying pancreatic cancer development.
Collapse
|
14
|
Li PC, Huang RY, Yang YC, Hsieh KP, Yang YH. Prognostic impact of angiotensin-converting enzyme inhibitors and angiotensin receptors blockers in esophageal or gastric cancer patients with hypertension - a real-world study. BMC Cancer 2022; 22:430. [PMID: 35443635 PMCID: PMC9022235 DOI: 10.1186/s12885-022-09513-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 01/18/2022] [Indexed: 11/10/2022] Open
Abstract
Background Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) are used in treating cardiovascular diseases. Previous studies indicated that ACEIs/ARBs may benefit cancer patients by inhibiting tumor angiogenesis and proliferation. The effect of ACEIs/ARBs on cancer survival in esophageal and gastric cancer is still unclear. This study is to investigate the association between ACEIs/ARBs usage and esophageal and gastric cancer prognosis. Methods This retrospective cohort study identified esophageal and gastric cancer patients during 2008–2016 from the Taiwan Cancer Registry, and obtained medication usage and follow-up information from the National Health Insurance Research Database and Death Registry. Analysis groups were defined as ACEIs/ARBs user or non-user based on the usage of ACEIs/ARBs within the 6 months after cancer diagnosis. The stabilized inverse probability of treatment weighting using propensity scores was applied to balance covariates between study groups. We also used Kaplan-Meier estimates and Cox regression to compare survival outcome and estimate hazard ratios (HRs). Results We identified 14,463 and 21,483 newly-diagnosed esophageal and gastric cancer patients during 2008–2016. ACEIs/ARBs users were associated with lower risk of cancer-specific mortality, although only significantly in gastric cancer (gastric: adjusted HR = 0.87, 95% CI = 0.78–0.97; esophageal: adjusted HR =0.88, 95% CI = 0.76–1.02). A better survival outcome was observed among patients who received higher cumulative defined daily dose of ACEIs/ARBs. Conclusions We found that using ACEIs/ARBs after cancer diagnosis were associated with lower risk of mortality. Our results add to the knowledge of the benefit of ACEIs/ARBs against mortality in individuals with esophageal/gastric cancer patients with hypertension. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09513-4.
Collapse
Affiliation(s)
- Po-Chih Li
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, Taiwan.,Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Ru-Yu Huang
- National Institute of Cancer Research, National Health Research Institutes, No.367, Sheng-Li Rd., North District, Tainan, 70456, Taiwan
| | - Yu-Chien Yang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, Taiwan.,Department of Pharmacy, Chi Mei Medical Center, Tainan, Taiwan
| | - Kun-Pin Hsieh
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, Taiwan. .,Department of Pharmacy, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| | - Yi-Hsin Yang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, 100, Shih-Chuan 1st Road, Kaohsiung, Taiwan. .,National Institute of Cancer Research, National Health Research Institutes, No.367, Sheng-Li Rd., North District, Tainan, 70456, Taiwan.
| |
Collapse
|
15
|
Uemura N, Hayashi H, Baba H. Statin as a therapeutic agent in gastroenterological cancer. World J Gastrointest Oncol 2022; 14:110-123. [PMID: 35116106 PMCID: PMC8790423 DOI: 10.4251/wjgo.v14.i1.110] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/19/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023] Open
Abstract
Statins inhibit 3-hydroxy-3-methylglutaryl-CoA reductase, the rate-limiting enzyme of the mevalonate pathway, and are widely used as an effective and safe approach handle hypercholesterolemia. The mevalonate pathway is a vital metabolic pathway that uses acetyl-CoA to generate isoprenoids and sterols that are crucial to tumor growth and progression. Multiple studies have indicated that statins improve patient prognosis in various carcinomas. Basic research on the mechanisms underlying the antitumor effects of statins is underway. The development of new anti-cancer drugs is progressing, but increasing medical costs from drug development have become a major obstacle. Readily available, inexpensive and well-tolerated drugs like statins have not yet been successfully repurposed for cancer treatment. Identifying the cancer patients that may benefit from statins is key to improved patient treatment. This review summarizes recent advances in statin research in cancer and suggests important considerations for the clinical use of statins to improve outcomes for cancer patients.
Collapse
Affiliation(s)
- Norio Uemura
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hiromitsu Hayashi
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Hideo Baba
- Department of Gastroenterological Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| |
Collapse
|
16
|
Dorsch M, Kowalczyk M, Planque M, Heilmann G, Urban S, Dujardin P, Forster J, Ueffing K, Nothdurft S, Oeck S, Paul A, Liffers ST, Kaschani F, Kaiser M, Schramm A, Siveke JT, Winslow MM, Fendt SM, Nalbant P, Grüner BM. Statins affect cancer cell plasticity with distinct consequences for tumor progression and metastasis. Cell Rep 2021; 37:110056. [PMID: 34818551 PMCID: PMC8640221 DOI: 10.1016/j.celrep.2021.110056] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/21/2021] [Accepted: 11/03/2021] [Indexed: 12/12/2022] Open
Abstract
Statins are among the most commonly prescribed drugs, and around every fourth person above the age of 40 is on statin medication. Therefore, it is of utmost clinical importance to understand the effect of statins on cancer cell plasticity and its consequences to not only patients with cancer but also patients who are on statins. Here, we find that statins induce a partial epithelial-to-mesenchymal transition (EMT) phenotype in cancer cells of solid tumors. Using a comprehensive STRING network analysis of transcriptome, proteome, and phosphoproteome data combined with multiple mechanistic in vitro and functional in vivo analyses, we demonstrate that statins reduce cellular plasticity by enforcing a mesenchymal-like cell state that increases metastatic seeding ability on one side but reduces the formation of (secondary) tumors on the other due to heterogeneous treatment responses. Taken together, we provide a thorough mechanistic overview of the consequences of statin use for each step of cancer development, progression, and metastasis.
Collapse
Affiliation(s)
- Madeleine Dorsch
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Manuela Kowalczyk
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Mélanie Planque
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Geronimo Heilmann
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Sebastian Urban
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Philip Dujardin
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Jan Forster
- Department of Genome Informatics, Institute for Human Genetics, University of Duisburg-Essen, Duisburg, Germany; German Cancer Consortium (DKTK) partner site Essen, Essen, Germany
| | - Kristina Ueffing
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Silke Nothdurft
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Sebastian Oeck
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Annika Paul
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Sven T Liffers
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany
| | - Farnusch Kaschani
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Markus Kaiser
- Department of Chemical Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Alexander Schramm
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy, West German Cancer Center, University Medicine Essen, Essen, Germany; Division of Solid Tumor Translational Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), partner site Essen, Heidelberg, Germany
| | - Monte M Winslow
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah-Maria Fendt
- Laboratory of Cellular Metabolism and Metabolic Regulation, VIB-KU Leuven Center for Cancer Biology, VIB, Leuven, Belgium; Laboratory of Cellular Metabolism and Metabolic Regulation, Department of Oncology, KU Leuven and Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Perihan Nalbant
- Department of Molecular Cell Biology, Center for Medical Biotechnology, University of Duisburg-Essen, Duisburg, Germany
| | - Barbara M Grüner
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen at the University Duisburg-Essen, Duisburg, Germany; German Cancer Consortium (DKTK) partner site Essen, Essen, Germany.
| |
Collapse
|
17
|
Dent P, Booth L, Poklepovic A, Von Hoff D, Martinez J, Zhou Y, Hancock JF. Osimertinib-resistant NSCLC cells activate ERBB2 and YAP/TAZ and are killed by neratinib. Biochem Pharmacol 2021; 190:114642. [PMID: 34077739 PMCID: PMC11082938 DOI: 10.1016/j.bcp.2021.114642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/26/2021] [Accepted: 05/28/2021] [Indexed: 11/15/2022]
Abstract
We performed additional mechanistic analyses to redefine neratinib biology and determined the mechanisms by which the multi-kinase inhibitor neratinib interacted with the thymidylate synthase inhibitor pemetrexed to kill NSCLC cells expressing either mutant KRAS (G12S; Q61H; G12A; G12C) or mutant NRAS (Q61K) or mutant ERBB1 (L858R; L858R T790M; exon 19 deletion). Neratinib rapidly reduced KRASG12V and RAC1G12V nanoclustering which was followed by KRASG12V, but not RAC1G12V, being extensively mislocalized away from the plasma membrane. This correlated with reduced levels of, and reorganized membrane localization of phosphatidylserine and cholesterol. Reduced nanoclustering was not associated with inactivation of ERBB1, Merlin or Ezrin. The drug combination killed cells expressing mutant KRAS, NRAS or mutant ERBB1 proteins. Afatinib or osimertinib resistant cells were killed with a similar efficacy to non-resistant cells. Compared to osimertinib-resistant cells, sensitive cells had less ERBB2 Y1248 phosphorylation. In osimertinib resistant H1975 cells, the drug combination was less capable of inactivating AKT, mTOR, STAT3, STAT5, ERK1/2 whereas it gained the ability to inactivate ERBB3. In resistant H1650 cells, the drug combination was less capable of inactivating JAK2 and STAT5. Sensitive cells exhibited elevated basal phosphorylation of YAP and TAZ. In resistant cells, portions of YAP and TAZ were localized in the nucleus. [Neratinib + pemetrexed] increased phosphorylation of YAP and TAZ, caused their nuclear exit, and enhanced ERBB2 degradation. Thus, neratinib targets an unidentified protein whose functional inhibition directly results in RAS inactivation and tumor cell killing. Our data prove that, albeit indirectly, oncogenic RAS proteins are druggable by neratinib.
Collapse
Affiliation(s)
- Paul Dent
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States.
| | - Laurence Booth
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| | - Andrew Poklepovic
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| | - Daniel Von Hoff
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| | - Jennifer Martinez
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| | - Yong Zhou
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| | - John F Hancock
- Department of Biochemistry and Molecular Biology, Medicine, Virginia Commonwealth University, Richmond, VA 23298, United States; Translational Genomics Research Institute (TGEN), Phoenix, AZ 85004, United States; Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, United States; Inflammation & Autoimmunity Group, National Institute of Environmental Health Sciences, Triangle Park, NC 27709, United States
| |
Collapse
|
18
|
Saito K, Sato Y, Nakatani E, Kaneda H, Yamamoto S, Miyachi Y, Itoh H. Statin Exposure and Pancreatic Cancer Incidence: A Japanese Regional Population-Based Cohort Study, the Shizuoka Study. Cancer Prev Res (Phila) 2021; 14:863-872. [PMID: 34244151 DOI: 10.1158/1940-6207.capr-21-0123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/07/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022]
Abstract
Preclinical studies suggest that statins contribute to the prevention of pancreatic cancer; however, the results of epidemiologic studies are inconsistent. Furthermore, sufficient data are unavailable for the general population of Asia. Here, we conducted an observational study using a comprehensive patient-linked, longitudinal health insurance database comprising the records of 2,230,848 individuals residing in Shizuoka Prefecture, Japan, from April 2012 to September 2018. We included individuals older than 40 years with data for medical examinations and statin exposure (≥365 statin prescription days). To balance baseline characteristics between the statin exposure and statin nonexposure groups, we used inverse probability of treatment propensity score weighting method. We estimated hazard ratios for associations with pancreatic cancer using the Cox proportional hazards regression model. Among 2,230,848 individuals, we included 100,537 in the statin exposure group (24%) and 326,033 in the statin nonexposure group (76%). Among the statin exposure group (352,485 person-years) and the statin nonexposure group (1,098,463 person-years), 394 (1.12 per 1,000 person-years) and 1176 (1.07 per 1,000 person-years) developed pancreatic cancer, respectively (P = 0.464). After adjustments using inverse probability of treatment weighting, the statin exposure group was associated with a decreased incidence of pancreatic cancer (hazard ratio, 0.84; 95% confidence intervals, 0.72-0.99; P = 0.036). In conclusion, the current Japanese regional population-based cohort study shows that statin exposure was associated with a lower incidence of pancreatic cancer. PREVENTION RELEVANCE: This study may support the possible role of statins in preventing pancreatic cancer in the general population in Japan.
Collapse
Affiliation(s)
- Kohei Saito
- Shizuoka General Hospital, Shizuoka City, Shizuoka, Japan.,Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Tokyo, Japan.,Shizuoka Graduate University of Public Health, Shizuoka City, Shizuoka, Japan
| | - Yoko Sato
- Shizuoka General Hospital, Shizuoka City, Shizuoka, Japan.,Shizuoka Graduate University of Public Health, Shizuoka City, Shizuoka, Japan
| | - Eiji Nakatani
- Shizuoka General Hospital, Shizuoka City, Shizuoka, Japan. .,Shizuoka Graduate University of Public Health, Shizuoka City, Shizuoka, Japan
| | - Hideaki Kaneda
- Translational Research Center for Medical Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe city, Hyogo, Japan
| | - Seiichiro Yamamoto
- Shizuoka Graduate University of Public Health, Shizuoka City, Shizuoka, Japan.,Center for Cancer Control and Information Services, National Cancer Center, Tokyo, Japan
| | - Yoshiki Miyachi
- Shizuoka Graduate University of Public Health, Shizuoka City, Shizuoka, Japan
| | - Hiroshi Itoh
- Department of Endocrinology, Metabolism and Nephrology, School of Medicine, Keio University, Tokyo, Japan
| |
Collapse
|
19
|
Tamburrino D, Guarneri G, Pagnanelli M, Crippa S, Partelli S, Belfiori G, Capurso G, Falconi M. Chemopreventive Agents After Pancreatic Resection for Ductal Adenocarcinoma: Legend or Scientific Evidence? Ann Surg Oncol 2021; 28:2312-2322. [PMID: 32920722 DOI: 10.1245/s10434-020-09097-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 08/16/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is currently the fourth leading cause of cancer-related death in the USA. A wealth of evidence has demonstrated the chemopreventive activity of aspirin, statins, and metformin against PDAC. The aim of this study is to investigate the effect of aspirin, statins, and metformin on disease-free survival (DFS) and disease-specific survival (DSS) in a large population of PDAC patients undergoing pancreatic resection. PATIENTS AND METHODS All patients who underwent pancreatic resections between January 2015 and September 2018 were retrospectively reviewed. The potentially "chemopreventive agents" considered for the analysis were aspirin, statins, and metformin. Drug use was defined in case of regular assumption at least 6 months before diagnosis and regularly after surgery along the follow-up period. RESULTS A total of 430 patients were enrolled in this study, with median DFS and DSS of 21 months (IQR 13-30) months and 34 (IQR 26-52) months, respectively. On multivariable analysis, use of aspirin was associated with better DFS (HR: 0.62; p = 0.038). Metformin was associated with better DFS, without reaching statistical significance (p = 0.083). Use of statins did not influence DFS in the studied population. Aspirin, metformin, and statins were not associated with better DSS on multivariable analysis. Factors influencing DSS were pT3/pT4, N1, N2, no adjuvant treatment, G3, and ASA score > 3. CONCLUSIONS The results suggest that chronic use of aspirin is associated with increased DFS but not with better DSS after surgical resection in patients with PDAC.
Collapse
Affiliation(s)
- Domenico Tamburrino
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Giovanni Guarneri
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Michele Pagnanelli
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Crippa
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giulio Belfiori
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Vita-Salute San Raffaele University, Milan, Italy.
- Vita-Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
20
|
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: 31] [Impact Index Per Article: 7.8] [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
|
21
|
Chen C, Wu H, Kong D, Xu Y, Zhang Z, Chen F, Zou L, Li Z, Shui J, Luo H, Liu SH, Yu J, Wang K, Brunicardi FC. Transcriptome sequencing analysis reveals unique and shared antitumor effects of three statins in pancreatic cancer. Oncol Rep 2020; 44:2569-2580. [PMID: 33125137 PMCID: PMC7640361 DOI: 10.3892/or.2020.7810] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/15/2020] [Indexed: 12/20/2022] Open
Abstract
Statins, a class of commonly prescribed cholesterol‑lowering medications, have been revealed to influence the risk of multiple types of cancer. However, the antitumor effects of statins on pancreatic cancer and their differential efficacy among a variety of statins are not currently well‑defined. The aim of the present study was therefore to identify and compare the genes and related biological pathways that were affected by each individual statin on pancreatic cancer. Two human pancreatic cancer cell lines, MiaPaCa2 and PANC1, were exposed to three statins, lovastatin, fluvastatin and simvastatin. The inhibitory effect of statins on pancreatic cancer cell proliferation was first validated. Next, RNA‑seq analysis was used to determine the gene expression alterations in either low (2 µM) or high (20 µM) statin concentration‑treated cancer cells. Marked differences in gene transcription profiles of both pancreatic cancer cell lines exposed to high concentration statins were observed. Notably, the high concentration statins significantly suppressed core‑gene CCNA2‑associated cell cycle and DNA replication pathways and upregulated genes involved in ribosome and autophagy pathways. However, the low concentration statin‑induced gene expression alterations were only detected in MiaPaCa2 cells. In conclusion, a marked difference in the intra and inter cell‑type performance of pancreatic cancer cells exposed to a variety of statins at low or high concentrations was reported herein, which may provide insights for the potential clinical use of statins in future pancreatic cancer therapeutics.
Collapse
Affiliation(s)
- Cheng Chen
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Hongjin Wu
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Deshengyue Kong
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Yu Xu
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Zunyue Zhang
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Fengrong Chen
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Lei Zou
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Ziwei Li
- Shanghai International Travel Healthcare Center, Shanghai 200000, P.R. China
| | - Jin Shui
- Shanghai International Travel Healthcare Center, Shanghai 200000, P.R. China
| | - Huayou Luo
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Shi-He Liu
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Juehua Yu
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Kunhua Wang
- The NHC Key Laboratory of Drug Addiction Medicine, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
- Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - F. Charles Brunicardi
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
22
|
Abstract
Cellular plasticity contributes to intratumoral heterogeneity, metastatic spread, and treatment resistance of cancers. In this issue of Cancer Cell, Gabitova-Cornell et al. identify the potential to inadvertently develop an undifferentiated and more aggressive pancreas cancer with agents commonly prescribed to manage heart disease risk.
Collapse
Affiliation(s)
- Sita Kugel
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA.
| | - Sunil R Hingorani
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA; Division of Medical Oncology, University of Washington School of Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
23
|
Archibugi L, Graglia B, Valente R, Stigliano S, Roberto M, Capalbo C, Marchetti P, Nigri G, Capurso G. Gynecological and reproductive factors and the risk of pancreatic cancer: A case-control study. Pancreatology 2020; 20:1149-1154. [PMID: 32800645 DOI: 10.1016/j.pan.2020.07.398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/28/2020] [Accepted: 07/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND /Objectives: Pancreatic ductal adenocarcinoma (PDAC) has a higher incidence in men compared to women, although the difference in known risk factors cannot explain this disparity completely. Reproductive and hormonal factors have been demonstrated in pre-clinical studies to influence pancreatic carcinogenesis, but the few published data on the topic are inconsistent. The aim was to investigate the role of reproductive and hormonal factors on PDAC occurrence in women. METHODS We conducted a unicenter case-control study; PDAC cases were matched to controls by age with a 1:2 ratio. Risk factors were screened through questionnaires about gynecologic and medical history. Comparisons were made using Chi-square and Fisher's exact tests where appropriate for categorical variables and Student's t-test for continuous variables. Logistic regression was used to calculate Odds Ratios (ORs) and their 95% confidence intervals (CI). Multivariable logistic regression models were adjusted for potential confounders. RESULTS 253 PDAC and 506 matched controls were enrolled. At logistic regression multivariable analysis adjusted for confounding factors, older age at menopause (OR:0.95 per year; 95% CI:0.91-0.98; p = 0.007), use of Oral Contraceptives (OR:0.52; 95% CI:0.30-0.89; p = 0.018), use of Hormonal Replacement Therapy (OR:0.31; 95% CI:0.15-0.64; p = 0.001), and having had two children (OR:0.57; 95% CI:0.38-0.84; p = 0.005) were significant, independent protective factors for the onset of PDAC. CONCLUSIONS These data confirm some previous findings on menopause age and number of births while, to our knowledge, this is the first study to show a protective effect of HRT and OC use. The results collectively support the hypothesis that exposure to estrogens plays a protective role towards PDAC.
Collapse
Affiliation(s)
- Livia Archibugi
- Digestive and Liver Disease Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy; Pancreatobiliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Benedetta Graglia
- Digestive and Liver Disease Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy
| | - Roberto Valente
- Digestive and Liver Disease Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy; Department of Surgery, Umeå University Hospital, Umeå, Sweden; Department of Surgery, Anschutz Medical Campus, University of Colorado, Denver, USA
| | - Serena Stigliano
- Digestive and Liver Disease Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy; Department of Internal Medicine and Medical Specialties, Gastroenterology Unit, Sapienza University of Rome, Milan, Italy
| | - Michela Roberto
- Oncology Unit, S. Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy
| | - Carlo Capalbo
- Oncology Unit, S. Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy
| | - Paolo Marchetti
- Oncology Unit, S. Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy
| | - Giuseppe Nigri
- General Surgery and Hepato-pancreato-biliary Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy
| | - Gabriele Capurso
- Digestive and Liver Disease Unit, Sant'Andrea Hospital, Faculty of Medicine and Psychology, Sapienza University of Rome, Milan, Italy; Pancreatobiliary Endoscopy and Endosonography Division, Pancreas Translational and Clinical Research Center, San Raffaele Hospital, San Raffaele Scientific Institute IRCCS, Milan, Italy.
| |
Collapse
|
24
|
Oni TE, Biffi G, Baker LA, Hao Y, Tonelli C, Somerville TD, Deschênes A, Belleau P, Hwang CI, Sánchez-Rivera FJ, Cox H, Brosnan E, Doshi A, Lumia RP, Khaledi K, Park Y, Trotman LC, Lowe SW, Krasnitz A, Vakoc CR, Tuveson DA. SOAT1 promotes mevalonate pathway dependency in pancreatic cancer. J Exp Med 2020; 217:151922. [PMID: 32633781 PMCID: PMC7478739 DOI: 10.1084/jem.20192389] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 03/28/2020] [Accepted: 05/12/2020] [Indexed: 12/31/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a dismal prognosis, and new therapies are needed. Altered metabolism is a cancer vulnerability, and several metabolic pathways have been shown to promote PDAC. However, the changes in cholesterol metabolism and their role during PDAC progression remain largely unknown. Here we used organoid and mouse models to determine the drivers of altered cholesterol metabolism in PDAC and the consequences of its disruption on tumor progression. We identified sterol O-acyltransferase 1 (SOAT1) as a key player in sustaining the mevalonate pathway by converting cholesterol to inert cholesterol esters, thereby preventing the negative feedback elicited by unesterified cholesterol. Genetic targeting of Soat1 impairs cell proliferation in vitro and tumor progression in vivo and reveals a mevalonate pathway dependency in p53 mutant PDAC cells that have undergone p53 loss of heterozygosity (LOH). In contrast, pancreatic organoids lacking p53 mutation and p53 LOH are insensitive to SOAT1 loss, indicating a potential therapeutic window for inhibiting SOAT1 in PDAC.
Collapse
Affiliation(s)
- Tobiloba E. Oni
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY
| | - Giulia Biffi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Lindsey A. Baker
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Claudia Tonelli
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Astrid Deschênes
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Chang-il Hwang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Department of Microbiology and Molecular Genetics, University of California, Davis, Davis, CA
| | | | - Hilary Cox
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Erin Brosnan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Abhishek Doshi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Rebecca P. Lumia
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Kimia Khaledi
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY
| | | | - Scott W. Lowe
- Department of Cancer Biology and Genetics, Memorial Sloan-Kettering Cancer Center, New York, NY,Howard Hughes Medical Institute, Memorial Sloan-Kettering Cancer Center, New York, NY
| | | | | | - David A. Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY,Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY,Correspondence to David A. Tuveson:
| |
Collapse
|
25
|
Kirtonia A, Gala K, Fernandes SG, Pandya G, Pandey AK, Sethi G, Khattar E, Garg M. Repurposing of drugs: An attractive pharmacological strategy for cancer therapeutics. Semin Cancer Biol 2020; 68:258-278. [PMID: 32380233 DOI: 10.1016/j.semcancer.2020.04.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/20/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Human malignancies are one of the major health-related issues though out the world and anticipated to rise in the future. The development of novel drugs/agents requires a huge amount of cost and time that represents a major challenge for drug discovery. In the last three decades, the number of FDA approved drugs has dropped down and this led to increasing interest in drug reposition or repurposing. The present review focuses on recent concepts and therapeutic opportunities for the utilization of antidiabetics, antibiotics, antifungal, anti-inflammatory, antipsychotic, PDE inhibitors and estrogen receptor antagonist, Antabuse, antiparasitic and cardiovascular agents/drugs as an alternative approach against human malignancies. The repurposing of approved non-cancerous drugs is an effective strategy to develop new therapeutic options for the treatment of cancer patients at an affordable cost in clinics. In the current scenario, most of the countries throughout the globe are unable to meet the medical needs of cancer patients because of the high cost of the available cancerous drugs. Some of these drugs displayed potential anti-cancer activity in preclinic and clinical studies by regulating several key molecular mechanisms and oncogenic pathways in human malignancies. The emerging pieces of evidence indicate that repurposing of drugs is crucial to the faster and cheaper discovery of anti-cancerous drugs.
Collapse
Affiliation(s)
- Anuradha Kirtonia
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Kavita Gala
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India; Equal contribution
| | - Gouri Pandya
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India; Equal contribution
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Haryana, 122413, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India.
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida, 201313, India.
| |
Collapse
|
26
|
Tamburrino D, Crippa S, Partelli S, Archibugi L, Arcidiacono PG, Falconi M, Capurso G. Statin use improves survival in patients with pancreatic ductal adenocarcinoma: A meta-analysis. Dig Liver Dis 2020; 52:392-399. [PMID: 32113888 DOI: 10.1016/j.dld.2020.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/21/2020] [Accepted: 01/23/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Previous studies on statins' effect on survival of patients with pancreatic ductal adenocarcinoma (PDAC) report conflicting results. AIMS To evaluate the association between statin use and PDAC patients' survival. METHODS A systematic review and meta-analysis was performed including case-control, cohort studies and randomized controlled trials assessing the association between statin use and survival in PDAC patients. Pooled HRs with 95%CIs were calculated using random effects model; publication bias was assessed through Begg and Mazumdar test and heterogeneity by I2 value. RESULTS 14 studies with 33,137 PDAC patients, 40% under statins, were included. Statins use was associated to a reduced death risk (HR 0.871; 95%CI: 0.819; 0.927; p = 0.0001) suggesting a protective effect, homogeneous for different geographic areas. This effect was significant in surgically resected patients (HR 0.50; 95%CI: 0.32; 0.76; p = 0.001) but not in those with advanced disease (HR 0.78; 95%CI: 0.59; 1.02; p = 0.07). In studies providing information on statin type, only rosuvastatin resulted associated to a reduced risk of death (HR 0.88; 95%CI: 0.81; 0.96; p = 0.004). CONCLUSIONS Statins use is significantly associated with a reduced risk of death in resected PDAC patients. This finding has to be considered with caution due to publication bias and the availability of only few studies for sensitivity analyses.
Collapse
Affiliation(s)
- Domenico Tamburrino
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; PhD Candidate in Digestive Oncology, "La Sapienza University" Rome, Italy
| | - Stefano Crippa
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita Salute University, Milan, Italy
| | - Stefano Partelli
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita Salute University, Milan, Italy
| | - Livia Archibugi
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Paolo Giorgio Arcidiacono
- Vita Salute University, Milan, Italy; Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| | - Massimo Falconi
- Pancreatic Surgery Unit, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute, Milan, Italy; Vita Salute University, Milan, Italy
| | - Gabriele Capurso
- Pancreato-Biliary Endoscopy and Endosonography Division, Pancreas Translational & Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy.
| |
Collapse
|
27
|
Tran KT, McMenamin ÚC, Coleman HG, Cardwell CR, Murchie P, Iversen L, Lee AJ, Thrift AP. Statin use and risk of liver cancer: Evidence from two population-based studies. Int J Cancer 2020; 146:1250-1260. [PMID: 31112291 DOI: 10.1002/ijc.32426] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 04/30/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
Epidemiological studies of statin use and liver cancer risk have produced conflicting results. We examined the association between statin use and risk of primary liver cancer in two large independent study populations taking account of important covariates and main indications of statins such as high cholesterol and chronic liver disease. We performed a nested case-control study within the Scottish Primary Care Clinical Informatics Unit (PCCIU) database. Five controls were matched to cases with primary liver cancer and we used conditional logistic regression to calculate odds ratios (ORs) and 95% confidence intervals (CIs) for associations with statin use. We also conducted a prospective cohort study within the UK Biobank using self-reported statin use and cancer-registry recorded primary liver cancer outcomes. Cox regression was used to calculate hazard ratios (HRs) and 95% CIs. In the PCCIU case-control analysis, 434 liver cancer cases were matched to 2,103 controls. In the UK Biobank cohort, 182 out of 475,768 participants developed incident liver cancer. Statin use was associated with 39% lower risk of liver cancer in the PCCIU (adjusted OR 0.61, 95% CI 0.43-0.87). When we examined specific subtypes of liver cancer in the UK Biobank, statin use was associated with lower risk of hepatocellular carcinoma (HCC; adjusted HR, 0.48; 95% CI, 0.24-0.94) but not intrahepatic bile duct carcinoma (IBDC; adjusted HR, 1.09; 95% CI, 0.45-2.64). In conclusion, we found a consistent inverse relationship between statin use and risk of primary liver cancer which was only seen for HCC but not IBDC.
Collapse
Affiliation(s)
- Kim Tu Tran
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Úna C McMenamin
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Helen G Coleman
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Chris R Cardwell
- Cancer Epidemiology and Health Services Research Group, Centre for Public Health, Queen's University Belfast, Belfast, Northern Ireland, United Kingdom
| | - Peter Murchie
- Academic Primary Care, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Lisa Iversen
- Academic Primary Care, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Amanda J Lee
- Medical Statistics Team, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
28
|
Paternoster S, Falasca M. The intricate relationship between diabetes, obesity and pancreatic cancer. Biochim Biophys Acta Rev Cancer 2019; 1873:188326. [PMID: 31707038 DOI: 10.1016/j.bbcan.2019.188326] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/28/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is one of the leading determinants of global cancer mortality, and its incidence is predicted to increase, to become in 2030 the second most common cause of cancer-related death. Obesity and diabetes are recognized risk factors for the development of pancreatic cancer. In the last few decades an epidemic of diabetes and obesity has been spreading worldwide, forewarning an increase in incidence of pancreatic cancer. This review considers the most recent literature, covering the multiple molecular axis linking these three pathologies, aiming to draw a more comprehensive view of pancreatic cancer for a better theragnostic stratification of the population.
Collapse
Affiliation(s)
- Silvano Paternoster
- Metabolic Signalling Group, School Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia.
| | - Marco Falasca
- Metabolic Signalling Group, School Pharmacy and Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Bentley 6102, Perth, Western Australia, Australia.
| |
Collapse
|
29
|
Choi JH, Lee SH, Huh G, Chun JW, You MS, Paik WH, Ryu JK, Kim YT. The association between use of statin or aspirin and pancreatic ductal adenocarcinoma: A nested case-control study in a Korean nationwide cohort. Cancer Med 2019; 8:7419-7430. [PMID: 31637875 PMCID: PMC6885885 DOI: 10.1002/cam4.2617] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022] Open
Abstract
Background Although several studies have suggested that aspirin and statins may help prevent pancreatic ductal adenocarcinoma (PDAC), this concept has been controversial. This study aimed to evaluate the association between use of statin or aspirin and PDAC in a nationwide large cohort. Methods In this nested case‐control study, we used data from a 12‐year nationwide longitudinal cohort in Korea. Cases with PDAC and controls who were matched to cases by age, sex, income, and index year at a 1:5 ratio were established. We used multivariate logistic regression analyses to identify the independent risk factors of PDAC. Results We identified a total of 827 patients with PDAC between 2007 and 2013, and included 4135 matched controls. Diabetes mellitus, chronic and acute pancreatitis, pancreatic cystic lesions, and cholelithiasis were independent risk factors for PDAC. Statin use (odds ratio [OR], 0.92; 95% confidence interval [CI] 0.76‐1.11; P = .344; adjusted OR [aOR], 0.70; 95% CI 0.56‐0.87; P = .001) was associated with a reduced risk of PDAC after correction of the confounding factors, but aspirin use (OR, 0.98; 95% CI 0.84‐1.15; P = .838; aOR 0.84; 95% CI 0.70‐1.01, P = .068) was not associated with PDAC. Among the patients with risk factors, both statin use (OR, 0.50; 95% CI 0.38‐0.66; P < .001; aOR, 0.62; 95% CI 0.45‐0.84; P = .002) and aspirin use (OR, 0.48; 95% CI 0.31‐0.67; P < .001; aOR 0.67; 95% CI 0.50‐0.89, P = .006) were associated with a reduced risk of PDAC. Conclusion This study suggests that statin use was associated with a reduced risk of PDAC incidence but aspirin use was not. Both statin use and aspirin use were associated a reduced risk of PDAC incidence for patients with risk factors.
Collapse
Affiliation(s)
- Jin Ho Choi
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sang Hyub Lee
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Gunn Huh
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jung Won Chun
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Min Su You
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Woo Hyun Paik
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Ji Kon Ryu
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Yong-Tae Kim
- Department of Internal Medicine, Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
30
|
Capurso G. Deprescription during last year of life in patients with pancreatic cancer: Optimization or nihilism? Cancer 2019; 125:3470-3471. [PMID: 31290998 DOI: 10.1002/cncr.32389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Gabriele Capurso
- Pancreato-biliary Endoscopy and EUS Division, Pancreas Translational and Clinical Research Center, San Raffaele Scientific Institute IRCCS, Milan, Italy
| |
Collapse
|
31
|
Neratinib inhibits Hippo/YAP signaling, reduces mutant K-RAS expression, and kills pancreatic and blood cancer cells. Oncogene 2019; 38:5890-5904. [PMID: 31253872 DOI: 10.1038/s41388-019-0849-8] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 05/20/2019] [Accepted: 05/30/2019] [Indexed: 12/29/2022]
Abstract
Prior studies demonstrated that the irreversible ERBB1/2/4 inhibitor neratinib caused plasma membrane-associated mutant K-RAS to localize in intracellular vesicles, concomitant with its degradation. Herein, we discovered that neratinib interacted with the chemically distinct irreversible ERBB1/2/4 inhibitor afatinib to reduce expression of ERBB1, ERBB2, K-RAS and N-RAS; this was associated with greater-than-additive cell killing of pancreatic tumor cells. Knock down of Beclin1, ATG16L1, Rubicon or cathepsin B significantly lowered the ability of neratinib to reduce ERBB1 and K-RAS expression, and to cause tumor cell death. Knock down of ATM-AMPK suppressed vesicle formation and knock down of cathepsin B-AIF significantly reduced neratinib lethality. PKG phosphorylates K-RAS and HMG CoA reductase inhibitors reduce K-RAS farnesylation both of which remove K-RAS from the plasma membrane, abolishing its activity. Neratinib interacted with the PKG activator sildenafil and the HMG CoA reductase inhibitor atorvastatin to further reduce K-RAS expression, and to further enhance cell killing. Neratinib is also a Ste20 kinase family inhibitor and in carcinoma cells, and hematopoietic cancer cells lacking ERBB1/2/4, it reduced K-RAS expression and the phosphorylation of MST1/3/4/Ezrin by ~ 30%. Neratinib increased LATS1 phosphorylation as well as that of YAP and TAZ also by ~ 30%, caused the majority of YAP to translocate into the cytosol and reduced YAP/TAZ protein levels. Neratinib lethality was enhanced by knock down of YAP. Neratinib, in a Rubicon-dependent fashion, reduced PAK1 phosphorylation and that of its substrate Merlin. Our data demonstrate that neratinib coordinately suppresses both mutant K-RAS and YAP function to kill pancreatic tumor cells.
Collapse
|
32
|
Zhang H, Wu J, Zhang Z, Qian H, Wang Y, Yang M, Cheng Y, Tang S. Association of atorvastatin with the risk of hepatotoxicity: a pilot prescription sequence symmetry analysis. Ther Clin Risk Manag 2019; 15:803-810. [PMID: 31417267 PMCID: PMC6602299 DOI: 10.2147/tcrm.s204860] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/27/2019] [Indexed: 01/01/2023] Open
Abstract
Purpose This study aimed to evaluate Atorvastatin (ATO)-associated hepatotoxicity using prescription sequence symmetry analysis (PSSA), based on a health insurance database of a Chinese population living in Jiangsu Province, China. Methods Patients prescribed ATO and hepatoprotective drugs in 2017 were identified, and the run-in period was determined based on the "waiting-time" distribution. Adjusted sequence ratio (ASR) and 95% confidence interval (95% CI) were calculated to estimate the risk of ATO-associated hepatotoxicity under different time intervals or based on gender and age stratification. Results A total of 2,549 patients, with 1,518 filling the ATO prescription first and 1,031 filling the ATO prescription second, were analyzed. After setting the run-in period as 30 days and the time interval as 15, 30, 60, 90, 120, and 180 days, the ASRs were 1.492 (95% CI: 1.367-1.652), 1.399 (95% CI: 1.308-1.508), 1.280 (95% CI: 1.213-1.357), 1.292 (95% CI: 1.234-1.356), 1.278 (95% CI: 1.226-1.336), and 1.274 (95% CI: 1.229-1.323), respectively. No significant difference was observed between different genders and ages (χ2=0.161, P=0.688; χ2=1.565, P=0.211, respectively). Conclusion This is the first study conducted in a real-world setting to evaluate the relationship between ATO and hepatotoxicity using the PSSA in a Chinese population. We found a 1.3- to 1.5-fold increase in risk of hepatotoxicity following ATO, with the greater risk occurring within the first 30 days of treatment.
Collapse
Affiliation(s)
- Haiping Zhang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Jiani Wu
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Zhuolin Zhang
- Department of Clinical Pharmacy, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Haisheng Qian
- Department of Internal Medicine, The First Clinical Medical College, Nanjing Medical University, Nanjing, Jiangsu 210029, People's Republic of China
| | - Yifan Wang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Miaomiao Yang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| | - Yinchu Cheng
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, People's Republic of China
| | - Shaowen Tang
- Department of Epidemiology, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, People's Republic of China
| |
Collapse
|
33
|
Primary and Secondary Prevention of Pancreatic Cancer. CURR EPIDEMIOL REP 2019. [DOI: 10.1007/s40471-019-00189-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
34
|
Kawada T. Statin use and pancreatic cancer: A risk assessment. Dig Liver Dis 2019; 51:749. [PMID: 30926286 DOI: 10.1016/j.dld.2019.02.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Tomoyuki Kawada
- Department of Hygiene and Public Health, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-Ku, Tokyo 113-8602, Japan.
| |
Collapse
|
35
|
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: 3.3] [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
|