1
|
Miret Durazo CI, Zachariah Saji S, Rawat A, Motiño Villanueva AL, Bhandari A, Nurjanah T, Ryali N, Zepeda Martínez IG, Cruz Santiago JA. Exploring Aspirin's Potential in Cancer Prevention: A Comprehensive Review of the Current Evidence. Cureus 2024; 16:e70005. [PMID: 39445288 PMCID: PMC11498354 DOI: 10.7759/cureus.70005] [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] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Aspirin, traditionally recognized for its analgesic, anti-inflammatory, antipyretic, and antiplatelet effects, has recently attracted attention for its potential role in cancer prevention. Initially studied for cardiovascular disease prevention, emerging evidence suggests that aspirin may reduce the risk of certain cancers, particularly colorectal cancer (CRC). This narrative review integrates findings from early studies, animal models, epidemiological data, and clinical trials to evaluate aspirin's efficacy as a chemopreventive agent. Aspirin's anticancer effects are primarily attributed to its cyclooxygenase (COX) enzyme inhibition, which decreases prostaglandin E2 (PGE2) levels and disrupts cancer-related signaling pathways. While epidemiological studies support an association between aspirin use and reduced cancer incidence and mortality, especially for CRC and potentially for breast (BC) and prostate cancers (PCa), the risk of adverse effects, such as gastrointestinal (GI) and intracranial bleeding, complicates its use and warrants careful consideration. The decision to use aspirin for cancer prevention should be individualized, balancing its therapeutic benefits against potential adverse effects. It also underscores the necessity for further research to refine dosage guidelines, assess long-term impacts, and explore additional biomarkers to guide personalized cancer prevention strategies.
Collapse
Affiliation(s)
| | | | - Akash Rawat
- Department of General Medicine, Himalayan Institute of Medical Sciences, Swami Rama Himalayan University, Dehradun, IND
| | | | - Amit Bhandari
- Internal Medicine, American University of the Caribbean School of Medicine, Cupecoy, SXM
| | - Tutut Nurjanah
- Department of General Medicine, Universitas Yarsi, Jakarta, IDN
| | - Niharika Ryali
- Department of General Medicine, Gandhi Medical College, Hyderabad, IND
| | | | - Josue A Cruz Santiago
- Department of General Medicine, Universidad Autónoma de Guadalajara, Guadalajara, MEX
| |
Collapse
|
2
|
Park JM, Hahm KB. Dietary Walnuts Prevented Indomethacin-Induced Gastric Damage via AP-1 Transcribed 15-PGDH, Nrf2-Mediated HO-1, and n-3 PUFA-Derived Resolvin E1. Int J Mol Sci 2024; 25:7239. [PMID: 39000345 PMCID: PMC11242660 DOI: 10.3390/ijms25137239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs), the most highly prescribed drugs in the world for the treatment of pain, inflammation, and fever, cause gastric mucosal damage, including ulcers, directly or indirectly, by which the development of GI-safer (-sparing) NSAIDs relates to unmet medical needs. This study aimed to document the preventive effects of walnut polyphenol extracts (WPEs) against NSAID-induced gastric damage along with the molecular mechanisms. RGM-1 gastric mucosal cells were administered with indomethacin, and the expressions of the inflammatory mediators between indomethacin alone or a combination with WPEs were compared. The expressions of the inflammatory mediators, including COX-1 and COX-2, prostaglandin E2, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), and antioxidant capacity, were analyzed by Western blot analysis, RT-PCR, and ELISA, respectively. HO-1, Nrf-2, and keap1 were investigated. The in vivo animal models were followed with in vitro investigations. The NSAIDs increased the expression of COX-2 and decreased COX-1 and 15-PGDH, but the WPEs significantly attenuated the NSAID-induced COX-2 expression. Interestingly, the WPEs induced the expression of 15-PGDH. By using the deletion constructs of the 15-PGDH promoter, we found that c-Jun is the most essential determinant of the WPE-induced up-regulation of 15-PGDH expression. We confirmed that the knockdown of c-Jun abolished the ability of the WPEs to up-regulate the 15-PGDH expression. In addition, the WPEs significantly increased the HO-1 expression. The WPEs increased the nuclear translocation of Nrf2 by Keap-1 degradation, and silencing Nrf2 markedly reduced the WPE-induced HO-1 expression. We found that the WPE-induced HO-1 up-regulation was attenuated in the cells harboring the mutant Keap1, in which the cysteine 151 residue was replaced by serine. These in vitro findings were exactly validated in indomethacin-induced gastric rat models. Daily walnut intake can be a promising nutritional supplement providing potent anti-inflammatory, antioxidative, and mucosa-protective effects against NSAID-induced GI damage.
Collapse
Affiliation(s)
- Jong Min Park
- College of Oriental Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Ki Baik Hahm
- CHA Cancer Preventive Research Center, CHA Bio Complex, Seongnam 13488, Republic of Korea
| |
Collapse
|
3
|
Drew DA, Kim AE, Lin Y, Qu C, Morrison J, Lewinger JP, Kawaguchi E, Wang J, Fu Y, Zemlianskaia N, Díez-Obrero V, Bien SA, Dimou N, Albanes D, Baurley JW, Wu AH, Buchanan DD, Potter JD, Prentice RL, Harlid S, Arndt V, Barry EL, Berndt SI, Bouras E, Brenner H, Budiarto A, Burnett-Hartman A, Campbell PT, Carreras-Torres R, Casey G, Chang-Claude J, Conti DV, Devall MA, Figueiredo JC, Gruber SB, Gsur A, Gunter MJ, Harrison TA, Hidaka A, Hoffmeister M, Huyghe JR, Jenkins MA, Jordahl KM, Kundaje A, Le Marchand L, Li L, Lynch BM, Murphy N, Nassir R, Newcomb PA, Newton CC, Obón-Santacana M, Ogino S, Ose J, Pai RK, Palmer JR, Papadimitriou N, Pardamean B, Pellatt AJ, Peoples AR, Platz EA, Rennert G, Ruiz-Narvaez E, Sakoda LC, Scacheri PC, Schmit SL, Schoen RE, Stern MC, Su YR, Thomas DC, Tian Y, Tsilidis KK, Ulrich CM, Um CY, van Duijnhoven FJ, Van Guelpen B, White E, Hsu L, Moreno V, Peters U, Chan AT, Gauderman WJ. Two genome-wide interaction loci modify the association of nonsteroidal anti-inflammatory drugs with colorectal cancer. SCIENCE ADVANCES 2024; 10:eadk3121. [PMID: 38809988 PMCID: PMC11135391 DOI: 10.1126/sciadv.adk3121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 04/26/2024] [Indexed: 05/31/2024]
Abstract
Regular, long-term aspirin use may act synergistically with genetic variants, particularly those in mechanistically relevant pathways, to confer a protective effect on colorectal cancer (CRC) risk. We leveraged pooled data from 52 clinical trial, cohort, and case-control studies that included 30,806 CRC cases and 41,861 controls of European ancestry to conduct a genome-wide interaction scan between regular aspirin/nonsteroidal anti-inflammatory drug (NSAID) use and imputed genetic variants. After adjusting for multiple comparisons, we identified statistically significant interactions between regular aspirin/NSAID use and variants in 6q24.1 (top hit rs72833769), which has evidence of influencing expression of TBC1D7 (a subunit of the TSC1-TSC2 complex, a key regulator of MTOR activity), and variants in 5p13.1 (top hit rs350047), which is associated with expression of PTGER4 (codes a cell surface receptor directly involved in the mode of action of aspirin). Genetic variants with functional impact may modulate the chemopreventive effect of regular aspirin use, and our study identifies putative previously unidentified targets for additional mechanistic interrogation.
Collapse
Affiliation(s)
- David A. Drew
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Andre E. Kim
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - John Morrison
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Juan Pablo Lewinger
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Eric Kawaguchi
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jun Wang
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yubo Fu
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Natalia Zemlianskaia
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Virginia Díez-Obrero
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Stephanie A. Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Niki Dimou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Baurley
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
- BioRealm LLC, Walnut, CA, USA
| | - Anna H. Wu
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Daniel D. Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, The University of Melbourne, Parkville, Victoria 3010 Australia
- University of Melbourne Centre for Cancer Research, Victorian Comprehensive Cancer Centre, Parkville, Victoria 3010 Australia
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - John D. Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Research Centre for Hauora and Health, Massey University, Wellington, New Zealand
| | - Ross L. Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sophia Harlid
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
| | - Volker Arndt
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elizabeth L. Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Emmanouil Bouras
- Laboratory of Hygiene, Social & Preventive Medicine and Medical Statistics, Department of Medicine, School of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Arif Budiarto
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
| | | | - Peter T. Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert Carreras-Torres
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Digestive Diseases and Microbiota Group, Girona Biomedical Research Institute (IDIBGI), Salt, 17190 Girona, Spain
| | - Graham Casey
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jenny Chang-Claude
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- University Medical Centre Hamburg-Eppendorf, University Cancer Centre Hamburg (UCCH), Hamburg, Germany
| | - David V. Conti
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Matthew A.M. Devall
- Department of Family Medicine, University of Virginia, Charlottesville, VA, USA
| | - Jane C. Figueiredo
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stephen B. Gruber
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, Duarte, CA, USA
- Center for Precision Medicine, City of Hope National Medical Center, Duarte, CA, USA
| | - Andrea Gsur
- Center for Cancer Research, Medical University Vienna, Vienna, Austria
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, UK
| | - Tabitha A. Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Akihisa Hidaka
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jeroen R. Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Mark A. Jenkins
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kristina M. Jordahl
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Anshul Kundaje
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | | | - Li Li
- Department of Family Medicine, University of Virginia, Charlottesville, VA, USA
- UVA Comprehensive Cancer Center, Charlottesville, VA, USA
| | - Brigid M. Lynch
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, Victoria, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Victoria, Australia
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Rami Nassir
- Department of Pathology, School of Medicine, Umm Al-Qura’a University, Mecca, Saudi Arabia
| | - Polly A. Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Mireia Obón-Santacana
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L’Hospitalet del Llobregat, 08908 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jennifer Ose
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Rish K. Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Scottsdale, AZ, USA
| | - Julie R. Palmer
- Slone Epidemiology Center at Boston University, Boston, MA, USA
| | - Nikos Papadimitriou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Bens Pardamean
- Bioinformatics and Data Science Research Center, Bina Nusantara University, Jakarta, Indonesia
| | - Andrew J. Pellatt
- Department of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita R. Peoples
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Gad Rennert
- Department of Community Medicine and Epidemiology, Lady Davis Carmel Medical Center, Haifa, Israel
- Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Clalit National Cancer Control Center, Haifa, Israel
| | - Edward Ruiz-Narvaez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Lori C. Sakoda
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Peter C. Scacheri
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Stephanie L. Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, OH, USA
- Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Robert E. Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Mariana C. Stern
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Yu-Ru Su
- Biostatistics Division, Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Duncan C. Thomas
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Yu Tian
- Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- School of Public Health, Capital Medical University, Beijing, China
| | - Konstantinos K. Tsilidis
- Department of Epidemiology and Biostatistics, Imperial College London, School of Public Health, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina School of Medicine, Ioannina, Greece
| | - Cornelia M. Ulrich
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Caroline Y. Um
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | | | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Victor Moreno
- Colorectal Cancer Group, ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Unit of Biomarkers and Susceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L’Hospitalet del Llobregat, 08908 Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), 28029 Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine and Health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
| | - Andrew T. Chan
- Clinical & Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - W. James Gauderman
- Division of Biostatistics, Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
4
|
Chen GQ, Nan Y, Huang SC, Ning N, Du YH, Lu DD, Yang YT, Meng FD, Yuan L. Research progress of ginger in the treatment of gastrointestinal tumors. World J Gastrointest Oncol 2023; 15:1835-1851. [DOI: 10.4251/wjgo.v15.i11.1835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer seriously endangers human health. Gastrointestinal cancer is the most common and major malignant tumor, and its morbidity and mortality are gradually increasing. Although there are effective treatments such as radiotherapy and chemotherapy for gastrointestinal tumors, they are often accompanied by serious side effects. According to the traditional Chinese medicine and food homology theory, many materials are both food and medicine. Moreover, food is just as capable of preventing and treating diseases as medicine. Medicine and food homologous herbs not only have excellent pharmacological effects and activities but also have few side effects. As a typical medicinal herb with both medicinal and edible uses, some components of ginger have been shown to have good efficacy and safety against cancer. A mass of evidence has also shown that ginger has anti-tumor effects on digestive tract cancers (such as gastric cancer, colorectal cancer, liver cancer, laryngeal cancer, and pancreatic cancer) through a variety of pathways. The aim of this study is to investigate the mechanisms of action of the main components of ginger and their potential clinical applications in treating gastrointestinal tumors.
Collapse
Affiliation(s)
- Guo-Qing Chen
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yi Nan
- Key Laboratory of Ningxia Ethnomedicine Modernization, Ministry of Education, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Shi-Cong Huang
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Na Ning
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Yu-Hua Du
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Dou-Dou Lu
- School of Clinical Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ya-Ting Yang
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Fan-Di Meng
- Traditional Chinese Medicine College, Ningxia Medical University, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| | - Ling Yuan
- College of Pharmacy, Ningxia Medical College, Yinchuan 750004, Ningxia Hui Autonomous Region, China
| |
Collapse
|
5
|
Davies JR, Mell T, Fuller H, Harland M, Saleh RN, Race AD, Rees CJ, Brown LC, Loadman PM, Downing A, Minihane AM, Williams EA, Hull MA. Polymorphisms in Cyclooxygenase, Lipoxygenase, and TP53 Genes Predict Colorectal Polyp Risk Reduction by Aspirin in the seAFOod Polyp Prevention Trial. Cancer Prev Res (Phila) 2023; 16:621-629. [PMID: 37756582 PMCID: PMC10618644 DOI: 10.1158/1940-6207.capr-23-0111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 07/13/2023] [Accepted: 09/25/2023] [Indexed: 09/29/2023]
Abstract
Aspirin and eicosapentaenoic acid (EPA) reduce colorectal adenomatous polyp risk and affect synthesis of oxylipins including prostaglandin E2. We investigated whether 35 SNPs in oxylipin metabolism genes such as cyclooxygenase (PTGS) and lipoxygenase (ALOX), as well as 7 SNPs already associated with colorectal cancer risk reduction by aspirin (e.g., TP53; rs104522), modified the effects of aspirin and EPA on colorectal polyp recurrence in the randomized 2 × 2 factorial seAFOod trial. Treatment effects were reported as the incidence rate ratio (IRR) and 95% confidence interval (CI) by stratifying negative binomial and Poisson regression analyses of colorectal polyp risk on SNP genotype. Statistical significance was reported with adjustment for the false discovery rate as the P and q value. 542 (of 707) trial participants had both genotype and colonoscopy outcome data. Reduction in colorectal polyp risk in aspirin users compared with nonaspirin users was restricted to rs4837960 (PTGS1) common homozygotes [IRR, 0.69; 95% confidence interval (CI), 0.53-0.90); q = 0.06], rs2745557 (PTGS2) compound heterozygote-rare homozygotes [IRR, 0.60 (0.41-0.88); q = 0.06], rs7090328 (ALOX5) rare homozygotes [IRR 0.27 (0.11-0.64); q = 0.05], rs2073438 (ALOX12) common homozygotes [IRR, 0.57 (0.41-0.80); q = 0.05], and rs104522 (TP53) rare homozygotes [IRR, 0.37 (0.17-0.79); q = 0.06]. No modification of colorectal polyp risk in EPA users was observed. In conclusion, genetic variants relevant to the proposed mechanism of action on oxylipins are associated with differential colorectal polyp risk reduction by aspirin in individuals who develop multiple colorectal polyps. SNP genotypes should be considered during development of personalized, predictive models of colorectal cancer chemoprevention by aspirin. PREVENTION RELEVANCE Single-nucleotide polymorphisms in genes controlling lipid mediator signaling may modify the colorectal polyp prevention activity of aspirin. Further investigation is required to determine whether testing for genetic variants can be used to target cancer chemoprevention by aspirin to those who will benefit most.
Collapse
Affiliation(s)
- John R. Davies
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Tracey Mell
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Harriett Fuller
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Mark Harland
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Rasha N.M. Saleh
- Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- Department of Clinical and Chemical Pathology, Faculty of Medicine, Alexandria University, Egypt
| | - Amanda D. Race
- Institute of Cancer Therapeutics, University of Bradford, United Kingdom
| | - Colin J. Rees
- Population Health Science Institute, Newcastle University, United Kingdom
| | - Louise C. Brown
- MRC Clinical Trials Unit at University College, London, United Kingdom
| | - Paul M. Loadman
- Institute of Cancer Therapeutics, University of Bradford, United Kingdom
| | - Amy Downing
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| | - Anne Marie Minihane
- Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia, Norwich, United Kingdom
- Norwich Institute of Health Ageing, Norwich, United Kingdom
| | | | - Mark A. Hull
- Leeds Institute of Medical Research, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
6
|
Lepore Signorile M, Grossi V, Fasano C, Simone C. Colorectal Cancer Chemoprevention: A Dream Coming True? Int J Mol Sci 2023; 24:ijms24087597. [PMID: 37108756 PMCID: PMC10140862 DOI: 10.3390/ijms24087597] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Colorectal cancer (CRC) is one of the deadliest forms of cancer worldwide. CRC development occurs mainly through the adenoma-carcinoma sequence, which can last decades, giving the opportunity for primary prevention and early detection. CRC prevention involves different approaches, ranging from fecal occult blood testing and colonoscopy screening to chemoprevention. In this review, we discuss the main findings gathered in the field of CRC chemoprevention, focusing on different target populations and on various precancerous lesions that can be used as efficacy evaluation endpoints for chemoprevention. The ideal chemopreventive agent should be well tolerated and easy to administer, with low side effects. Moreover, it should be readily available at a low cost. These properties are crucial because these compounds are meant to be used for a long time in populations with different CRC risk profiles. Several agents have been investigated so far, some of which are currently used in clinical practice. However, further investigation is needed to devise a comprehensive and effective chemoprevention strategy for CRC.
Collapse
Affiliation(s)
- Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology-IRCCS "Saverio de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology-IRCCS "Saverio de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology-IRCCS "Saverio de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology-IRCCS "Saverio de Bellis" Research Hospital, Castellana Grotte, 70013 Bari, Italy
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
7
|
Jiang J, Li A, Lai X, Zhang H, Wang C, Wang H, Li L, Liu Y, Xie L, Yang C, Zhang C, Lu S, Li Y. Correlation between Metabolite of Prostaglandin E2 and the incidence of colorectal adenomas. Front Oncol 2023; 13:1068469. [PMID: 36923425 PMCID: PMC10009184 DOI: 10.3389/fonc.2023.1068469] [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: 10/13/2022] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
Colorectal cancer is a common malignancy, and the incidence and mortality rates continue to rise. An important factor in the emergence of inflammation-induced colorectal carcinogenesis is elevated cyclooxygenase-2. Prostaglandin E2 (PGE2) over-production is frequently equated with cyclooxygenase-2 gene over-expression. PGE2 can be assessed by measuring the level of prostaglandin's main metabolite, PGE-M, in urine. Colorectal adenoma is a precancerous lesion that can lead to colorectal cancer. We conducted research to evaluate the association between urinary levels of the PGE-M and the risk of colorectal adenomas. In a western Chinese population, we identified 152 cases of adenoma and 152 controls patients without polyps. Adenoma cases were categorized into control, low-risk and high-risk groups. There was no significant change in PGE-M levels, between the control group and the low-risk adenoma group. In the high-risk group, the PGE-M levels were 23% higher than the control group. When compared to people with the lowest urine PGE-M levels (first quartile), people with greater urinary PGE-M levels had a higher chance of developing high-risk colorectal adenomas, with an adjusted odds ratio (95% CI) of 1.65 (0.76-3.57) in the fourth quartile group, (p= 0.013). We conclude urinary PGE-M is associated with the risk of developing high-risk adenomas. Urinary PGE-M level may be used as a non-invasive indicator for estimating cancer risk.
Collapse
Affiliation(s)
- Jia Jiang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Anjie Li
- Department of Medicine-Cardiovascular, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiaolian Lai
- Department of Digestive, People's Hospital of Songtao Miao Autonomous County, Tongren, China
| | - Hanqun Zhang
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Chonghong Wang
- Department of Digestive, People's Hospital of Songtao Miao Autonomous County, Tongren, China
| | - Huimin Wang
- Department of Digestive, People's Hospital of Songtao Miao Autonomous County, Tongren, China
| | - Libo Li
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Yuncong Liu
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Lu Xie
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Can Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Cui Zhang
- Zunyi Medical University, Zunyi, China
| | - Shuoyan Lu
- Department of Digestive, People's Hospital of Songtao Miao Autonomous County, Tongren, China
| | - Yong Li
- Department of Oncology, Guizhou Provincial People’s Hospital, Guiyang, China
| |
Collapse
|
8
|
Grancher A, Michel P, Di Fiore F, Sefrioui D. Colorectal cancer chemoprevention: is aspirin still in the game? Cancer Biol Ther 2022; 23:446-461. [PMID: 35905195 PMCID: PMC9341367 DOI: 10.1080/15384047.2022.2104561] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Screening strategies have demonstrated their potential for decreasing the incidence and mortality of cancers, particularly that of colorectal cancer (CRC). Another strategy that has been developed to reduce CRC occurrence is the use of chemoprevention agents. Among them, aspirin is the most promising. Aspirin acts in colorectal tumourigenesis through several mechanisms, either directly in tumor cells or in their microenvironment, such as through its anti-inflammatory activity or its effect on the modulation of platelet function. Many retrospective studies, as well as follow-up of large cohorts from trials with primary cardiovascular end points, have shown that long-term treatment with daily low-dose aspirin decreases the incidence of adenomas and colorectal cancers. Therefore, aspirin is currently recommended by the United States Preventive Services Task Force (USPSTF) for primary prevention of CRC in all patients aged 50 to 59 with a 10-y risk of cardiovascular events greater than 10%. Furthermore, several studies have also reported that long-term aspirin treatment taking after CRC resection decreases recurrence risk and increases overall survival, especially in patients with PIK3CA-mutated tumors. This review summarizes current knowledge on the pathophysiological mechanisms of aspirin chemoprevention, discusses the primary clinical results on CRC prevention and highlights the potential biomarkers identified to predict aspirin efficacy.
Collapse
Affiliation(s)
- Adrien Grancher
- Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Normandie Univ, Iron Group, Rouen University Hospital, Rouen, France
| | - Pierre Michel
- Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Normandie Univ, Iron Group, Rouen University Hospital, Rouen, France
| | - Frederic Di Fiore
- Normandy Centre for Genomic and Personalized Medicine, Department of Hepatogastroenterology and Department of Medical Oncology, Henri Becquerel Centre, Normandie Univ, IRON group, Rouen University Hospital, Rouen, France
| | - David Sefrioui
- Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Normandie Univ, Iron Group, Rouen University Hospital, Rouen, France
| |
Collapse
|
9
|
Yang R, Liu Z, Cao H, Shi Y. LINC01089, suppressed by YY1, inhibits lung cancer progression by targeting miR-301b-3p/HPDG axis. Cell Biol Toxicol 2022; 38:1063-1077. [PMID: 34561789 DOI: 10.1007/s10565-021-09643-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 08/09/2021] [Indexed: 01/25/2023]
Abstract
PURPOSE LINC01089 is a newly identified lncRNA and rarely reported in human cancers. Our study aimed to investigate its role in lung cancer. METHODS YY1, LINC01089, and miR-301b-3p levels in lung cancer tissues and cells were assessed using qRT-PCR. Bioinformatics analysis and luciferase reporter, ChIP, and RIP assays were carried out for determining the relationships among YY1, LINC01089, miR-301b-3p, and HPGD. Gain- and loss-of-function assays were carried out to confirm the impacts of LINC01089 and HPDG in lung cancer cells. CCK-8 assay was used to assess cell proliferation rate, and Transwell assay was applied to measure cell invasion and migration. An in vivo tumor model was applied for validating the role of LINC01089. RESULTS LINC01089 was decreased in lung cancer tissues and cells, and low LINC01089 level predicted a poor clinical outcome. YY1 directly bound to LINC01089 promoter region and inhibited its transcription. LINC01089 knockdown thwarted the proliferation, invasion, and migration capacity of H1299 and A549 cells and aggravated tumor growth. Specifically, LINC01089 functioned as a competing endogenous RNA of miR-301b-3p to modulate HPGD and thereby affected lung cancer progression. CONCLUSION Our data revealed that LINC01089, directly suppressed by YY1, inhibited lung cancer progression by targeting the miR-301b-3p/HPGD axis. Graphical abstract 1. LINC01089 expression was downregulated in lung cancer tisuues and cell lines, and low LINC01089 levels predicted a poor clinical outcome. 2. LINC01089 knockdown enhanced proliferation, invasion, and migration of H1299 and A549 cells in vitro and promoted lung cancer cell tumorigenesis and metastasis in vivo. 3. LINC01089, directly suppressed by YY1, functioned as a competing endogenous RNA against miR-301b-3p to increase HPGD expression.
Collapse
Affiliation(s)
- Rusong Yang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, 210011, People's Republic of China.
| | - Zhengcheng Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, 210011, People's Republic of China
| | - Hui Cao
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, 210011, People's Republic of China
| | - Ye Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Nanjing Medical University, No.121 Jiangjiayuan Road, Gulou District, Nanjing, Jiangsu, 210011, People's Republic of China
| |
Collapse
|
10
|
Fan Y, Yang L, Ren Y, Wu Y, Li L, Li L. Sp1-Induced SETDB1 Overexpression Transcriptionally Inhibits HPGD in a β-Catenin-Dependent Manner and Promotes the Proliferation and Metastasis of Gastric Cancer. J Gastric Cancer 2022; 22:319-338. [PMID: 36316108 PMCID: PMC9633935 DOI: 10.5230/jgc.2022.22.e26] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 08/20/2024] Open
Abstract
PURPOSE Gastric cancer (GC) has high morbidity and mortality, the cure rate of surgical treatment and drug chemotherapy is not ideal. Therefore, development of new treatment strategies is necessary. We aimed to identify the mechanism underlying Sp1 regulation of GC progression. METHODS AND METHODS The levels of Sp1, β-catenin, SET domain bifurcated 1 (SETDB1), and 15-hydroxyprostaglandin dehydrogenase (HPGD) were detected by quantitative reverse transcription polymerase chain reaction and western blot analysis. The targets of SETDB1 were predicted by AnimalTFDB, and dual-luciferase reporter assay was used for confirming the combination of Sp1, β-catenin, and SETDB1. HGC27 or AGS cells (1×106 cells/mouse) were injected into mice via the caudal vein for GC model establishment. The level of Ki67 was detected using immunohistochemistry, and hematoxylin and eosin staining was performed for evaluating tumor metastasis in mice with GC. RESULTS HPGD was inhibited, while the protein levels of Sp1, β-catenin, and SETDB1 were up-regulated in GC tissues and cell lines. HPGD overexpression or SETDB1 silencing inhibited the proliferation, invasion, and migration of GC cells, and Sp1 regulated the proliferation, invasion, and migration of GC cells in a β-catenin-dependent manner. Furthermore, HPGD served as a target of SETDB1, and it was negatively regulated by SETDB1; additionally, Sp1 and β-catenin bound to the SETDB1 promoter and negatively regulated HPGD expression. We proved that Sp1 regulated GC progression via the SETDB1/HPGD axis. CONCLUSIONS Our findings revealed that Sp1 transcriptionally inhibited HPGD via SETDB1 in a β-catenin-dependent manner and promoted the proliferation and metastasis of GC cells.
Collapse
Affiliation(s)
- Yaguan Fan
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of Medical Oncology, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China
| | - Libo Yang
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of Medical Oncology, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China
| | - Yi Ren
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of Medical Oncology, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China
| | - Yunhua Wu
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of Medical Oncology, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China
| | - Linhai Li
- Department of General Surgery, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of General Surgery, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China.
| | - Lihua Li
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, Kunming, P.R. China
- Department of Medical Oncology, Kunming University of Science and Technology School of Medicine, Kunming, P.R. China.
| |
Collapse
|
11
|
Shureiqi I. Aspirin for Colorectal Cancer Prevention: Age Matters. Cancer Prev Res (Phila) 2022; 15:565-567. [PMID: 36047054 DOI: 10.1158/1940-6207.capr-22-0176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 06/23/2022] [Accepted: 06/30/2022] [Indexed: 11/16/2022]
Abstract
Further data from the Aspirin in Reducing Events in the Elderly (ASPREE) trial heightens the concern regarding aspirin use for colorectal cancer prevention in elderly subjects. A 95-variant colorectal cancer polygenic risk score (PRS) failed to identify a subset of elderly individuals who could have benefited from aspirin preventive activity. Further research to define predictive biomarkers of aspirin preventive activity is needed. Meanwhile, the use of aspirin for colorectal cancer prevention in the elderly becomes more questionable. See Cancer Prev Res 15(7):447-53.
Collapse
Affiliation(s)
- Imad Shureiqi
- Department of Internal Medicine, Division of Hematology and Oncology, University of Michigan, Ann Arbor, Michigan.,Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
12
|
Abumustafa W, Zamer BA, Khalil BA, Hamad M, Maghazachi AA, Muhammad JS. Protein arginine N-methyltransferase 5 in colorectal carcinoma: Insights into mechanisms of pathogenesis and therapeutic strategies. Biomed Pharmacother 2021; 145:112368. [PMID: 34794114 DOI: 10.1016/j.biopha.2021.112368] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/20/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
Protein arginine N-methyltransferase 5 (PRMT5) enzyme is one of the eight canonical PRMTs, classified as a type II PRMT, induces arginine monomethylation and symmetric dimethylation. PRMT5 is known to be overexpressed in multiple cancer types, including colorectal cancer (CRC), where its overexpression is associated with poor survival. Recent studies have shown that upregulation of PRMT5 induces tumor growth and metastasis in CRC. Moreover, various novel PRMT5 inhibitors tested on CRC cell lines showed promising anticancer effects. Also, it was suggested that PRMT5 could be a valid biomarker for CRC diagnosis and prognosis. Hence, a deeper understanding of PRMT5-mediated CRC carcinogenesis could provide new avenues towards developing a targeted therapy. In this study, we started with in silico analysis correlating PRMT5 expression in CRC patients as a prelude to further our investigation of its role in CRC. We then carried out a comprehensive review of the scientific literature that dealt with the role(s) of PRMT5 in CRC pathogenesis, diagnosis, and prognosis. Also, we have summarized key findings from in vitro research using various therapeutic agents and strategies directly targeting PRMT5 or disrupting its function. In conclusion, PRMT5 seems to play a significant role in the pathogenesis of CRC; therefore, its prognostic and therapeutic potential merits further investigation.
Collapse
Affiliation(s)
- Wafaa Abumustafa
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Batoul Abi Zamer
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Bariaa A Khalil
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Mawieh Hamad
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Azzam A Maghazachi
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Jibran Sualeh Muhammad
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates.
| |
Collapse
|
13
|
Zhang Y, Chan AT, Meyerhardt JA, Giovannucci EL. Timing of Aspirin Use in Colorectal Cancer Chemoprevention: A Prospective Cohort Study. J Natl Cancer Inst 2021; 113:841-851. [PMID: 33528007 PMCID: PMC8246825 DOI: 10.1093/jnci/djab009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Prior epidemiological and intervention studies have not been able to separate independent effects of dose, timing, and duration of aspirin use in colorectal cancer (CRC) chemoprevention. We examined aspirin-based CRC chemoprevention according to timing in the Nurses' Health Study and Health Professionals Follow-Up Study. METHODS The exposures include cumulative average dose and total duration of aspirin use in more than 10 years before follow-up started (remote period) and in the immediate 10 years before follow-up started (recent period). Cox models were used to estimate hazard ratios (HRs) and 95% confidence intervals (CIs) for exposures and CRC risk. RESULTS Aspirin use of longer than 10 years before follow-up started (HR = 0.88, 95% CI = 0.83 to 0.94) per 5-year increment and the immediate 10 years before follow-up started (HR = 0.90, 95% CI = 0.84 to 0.96) were similarly important in CRC chemoprevention, though a 5-year lag was required for a clear benefit in the recent period. In the remote period, the association was not dose dependent; compared with less than 0.5 standard-dose (325 mg) tablets per week; hazard ratios were 0.78 (95% CI = 0.63 to 0.98), 0.81 (95% CI = 0.72 to 0.91), and 0.74 (95% CI = 0.64 to 0.86) for doses of 0.5 to less than 1.5, 1.5 to less than 5, and 5 and more tablets per week, respectively. However, there was dose dependency in the recent period (with respective HR = 0.91, 95% CI = 0.79 to 1.06; HR = 0.87, 95% CI = 0.77 to 0.98; and HR = 0.76, 95% CI = 0.64 to 0.91). CONCLUSIONS A suggestive benefit necessitates at least 6-10 years and most clearly after approximately 10 years since initiation of aspirin. Remote use and use within the previous 10 years both contribute independently to decrease risk, though a lower dose may be required for a benefit with longer term use.
Collapse
Affiliation(s)
- Yin Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
14
|
Bai J, Chen H, Bai X. Relationship between microsatellite status and immune microenvironment of colorectal cancer and its application to diagnosis and treatment. J Clin Lab Anal 2021; 35:e23810. [PMID: 33938589 PMCID: PMC8183910 DOI: 10.1002/jcla.23810] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 12/13/2022] Open
Abstract
Due to advances in understanding the immune microenvironment of colorectal cancer (CRC), microsatellite classification (dMMR/MSI-H and pMMR/MSS) has become a key biomarker for the diagnosis and treatment of CRC patients and therefore has important clinical value. Microsatellite status is associated with a variety of clinicopathological features and affects drug resistance and the prognosis of patients. CRC patients with different microsatellite statuses have different compositions and distributions of immune cells and cytokines within their tumor microenvironments (TMEs). Therefore, there is great interest in reversing or reshaping CRC TMEs to transform immune tolerant "cold" tumors into immune sensitive "hot" tumors. This requires a thorough understanding of differences in the immune microenvironments of MSI-H and MSS type tumors. This review focuses on the relationship between CRC microsatellite status and the immune microenvironment. It focuses on how this relationship has value for clinical application in diagnosis and treatment, as well as exploring the limitations of its current application.
Collapse
Affiliation(s)
- Junge Bai
- The Fourth Hospital of Harbin Medical UniversityHarbinChina
| | - Hongsheng Chen
- Department of General SurgeryThe Fourth Hospital of Harbin Medical UniversityHarbinChina
| | - Xuefeng Bai
- Department of Colorectal SurgeryHarbin Medical University Cancer HospitalHarbinChina
| |
Collapse
|
15
|
Feng Y, Tao L, Wang G, Li Z, Yang M, He W, Zhong X, Zhang Y, Yang J, Cheung S, McDonald F, Chen L. Aspirin inhibits prostaglandins to prevents colon tumor formation via down-regulating Wnt production. Eur J Pharmacol 2021; 906:174173. [PMID: 34033814 DOI: 10.1016/j.ejphar.2021.174173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/15/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023]
Abstract
According to numerous epidemiological studies, aspirin is a non-steroidal anti-inflammatory drug (NSAID) that reduces the occurrence and mortality of colorectal cancer (CRC). However, the underlying mechanisms are not well identified. In an effort to fill these gaps, we administered aspirin on mice one day before induction in an azoxymethane (AOM)/dextran sulfate sodium (DSS) induced CRC model. In this study, we assessed the effects of aspirin on tumorigenesis and tumor cell proliferation. Multi-layer analyses were carried out to identify changes in cytokines, metabolites, level of gene expressions, and proteins associated with tumorigenesis and aspirin treatment. The results showed that aspirin-treated mice developed fewer colon tumors in response to AOM/DSS, and aspirin can actively block cyclooxygenase (COX) metabolism and reduce levels of pro-inflammatory cytokines. In addition, the transcriptomic and proteomic analyses both indicated that aspirin has an inhibitory effect on the Wnt pathway. The in vitro results further indicated that aspirin inhibits WNT6 production, possibly by suppressing its transcription factor NR4A2, which in turn is regulated by prostaglandin E2, thereby ultimately inhibiting the Wnt pathway. These findings improve our understanding of the mechanisms behind aspirin's chemoprevention effect on CRC.
Collapse
Affiliation(s)
- Yaqian Feng
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Lei Tao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China; Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Guoqiang Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Zhen Li
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Mingming Yang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Weishen He
- Boston College, 140 Commonwealth Ave, Chestnut Hill, MA 02467, USA
| | - Xincheng Zhong
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China
| | - Yong Zhang
- Advanced Biotechnology and Application Research Center, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing, 100024, China
| | - Jinliang Yang
- Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | | | - Fiona McDonald
- Bayer AG, Research & Development Pharmaceuticals, Berlin, 13342, Germany.
| | - Ligong Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Tsinghua University, Beijing, 100084, China; Collaborative Innovation Center for Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China.
| |
Collapse
|
16
|
Abstract
Colorectal cancer (CRC) is characterized by genetic-environmental interplay leading to diffuse changes in the entire colonic mucosa (field carcinogenesis or field of injury) and to a pro-neoplastic genetic/epigenetic/physiological milieu. The clinical consequences are increased risk of synchronous and metachronous neoplasia. Factors such as genetics, race, ethnicity, age, and socioeconomic status are thought to influence neoplasia development. Here, we explore the potential improvement to CRC screening through exploiting field carcinogenesis, with particular focus on racial disparities and chemoprevention strategies. Also, we discuss future directions for field carcinogenesis/risk stratification using molecular and novel biophotonic techniques for personalized CRC screening.
Collapse
|
17
|
Abstract
High-quality evidence indicates that regular use of aspirin is effective in reducing the risk for precancerous colorectal neoplasia and colorectal cancer (CRC). This has led to US and international guidelines recommending aspirin for the primary prevention of CRC in specific populations. In this review, we summarize key questions that require addressing prior to broader adoption of aspirin-based chemoprevention, review recent evidence related to the benefits and harms of aspirin use among specific populations, and offer a rationale for precision prevention approaches. We specifically consider the mechanistic implications of evidence showing differences in aspirin's effects according to age, the potential role of modifiable mechanistic biomarkers for personalizing prevention, and emerging evidence that the gut microbiota may offer novel aspirin-associated preventive targets to reduce high-risk neoplasia.
Collapse
Affiliation(s)
- David A Drew
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; ,
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA; ,
- Department of Immunology and Infectious Disease, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
18
|
Gibbs DC, Fedirko V, Baron JA, Barry EL, Flanders WD, McCullough ML, Yacoub R, Raavi T, Rutherford RE, Seabrook ME, Bostick RM. Inflammation Modulation by Vitamin D and Calcium in the Morphologically Normal Colorectal Mucosa of Patients with Colorectal Adenoma in a Clinical Trial. Cancer Prev Res (Phila) 2021; 14:65-76. [PMID: 32917645 PMCID: PMC7947029 DOI: 10.1158/1940-6207.capr-20-0140] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/29/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022]
Abstract
Increased COX-2 and decreased 15-hydroxyprostaglandin dehydrogenase (15-HPGD) expression promote prostaglandin-mediated inflammation and colorectal carcinogenesis. Experimental studies suggest that vitamin D and calcium may inhibit these pathways, but their effects on colorectal tissue COX-2 and 15-HPGD expression in humans are unknown. We tested the effects of supplemental vitamin D (1,000 IU/day) and/or calcium (1,200 mg/day) on COX-2 and 15-HPGD expression in the morphologically normal rectal mucosa from 62 paients with colorectal adenoma in a placebo-controlled chemoprevention trial. We measured biomarker expression using automated IHC and quantitative image analysis at baseline and 1-year follow-up, and assessed treatment effects using mixed linear models. The primary outcome was the COX-2/15-HPGD expression ratio, because these enzymes function as physiologic antagonists. After 1 year of treatment, the mean COX-2/15-HPGD expression ratio in full-length crypts proportionately decreased 47% in the vitamin D group (P = 0.001), 46% in the calcium group (P = 0.002), and 34% in the calcium + vitamin D group (P = 0.03), relative to the placebo group. Among individuals with the functional vitamin D-binding protein isoform DBP2 (GC rs4588*A), the COX-2/15-HPDG ratio decreased 70% (P = 0.0006), 75% (P = 0.0002), and 60% (P = 0.006) in the vitamin D, calcium, and combined supplementation groups, respectively, relative to placebo. These results show that vitamin D and calcium favorably modulate the balance of expression of COX-2 and 15-HPGD-biomarkers of inflammation that are strongly linked to colorectal carcinogenesis-in the normal-appearing colorectal mucosa of patients with colorectal adenoma (perhaps especially those with the DBP2 isoform). PREVENTION RELEVANCE: Supplemental calcium and vitamin D reduce indicators of cancer-promoting inflammation in normal colorectal tissue in humans, thus furthering our understanding of how they may help prevent colorectal cancer.
Collapse
Affiliation(s)
- David Corley Gibbs
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - John A Baron
- University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Elizabeth L Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | - W Dana Flanders
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | | | - Rami Yacoub
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Tapasya Raavi
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Robin E Rutherford
- Division of Digestive Diseases, Department of Medicine, Emory University, Atlanta, Georgia
| | | | - Roberd M Bostick
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia. .,Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
19
|
Curcumin induces expression of 15-hydroxyprostaglandin dehydrogenase in gastric mucosal cells and mouse stomach in vivo: AP-1 as a potential target. J Nutr Biochem 2020; 85:108469. [DOI: 10.1016/j.jnutbio.2020.108469] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023]
|
20
|
Usha T, Middha SK, Kukanur AA, Shravani RV, Anupama MN, Harshitha N, Rahamath A, Kukanuri SA, Goyal AK. Drug Repurposing Approaches: Existing Leads For Novel Threats And Drug Targets. Curr Protein Pept Sci 2020; 22:CPPS-EPUB-110124. [PMID: 32957901 DOI: 10.2174/1389203721666200921152853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 11/22/2022]
Abstract
Drug Repurposing (DR) is an alternative to the traditional drug discovery process. It is cost and time effective, with high returns and low risk process that can tackle the increasing need for interventions for varied diseases and new outbreaks. Repurposing of old drugs for other diseases has gained a wider attention, as there have been several old drugs approved by FDA for new diseases. In the global emergency of COVID19 pandemic, this is one of the strategies implemented in repurposing of old anti-infective, anti-rheumatic and anti-thrombotic drugs. The goal of the current review is to elaborate the process of DR, its advantages, repurposed drugs for a plethora of disorders, and the evolution of related academic publications. Further, detailed are the computational approaches: literature mining and semantic inference, network-based drug repositioning, signature matching, retrospective clinical analysis, molecular docking and experimental phenotypic screening. We discuss the legal and economical potential barriers in DR, existent collaborative models and recommendations for overcoming these hurdles and leveraging the complete potential of DR in finding new indications.
Collapse
Affiliation(s)
- Talambedu Usha
- Department of Biochemistry, Bangalore University, Bengaluru, Karnataka. India
| | - Sushil K Middha
- DBT-BIF Centre, Department of Biotechnology, Maharani Lakshmi Ammanni College for Women(mLAC), Bengaluru, Karnataka. India
| | | | | | | | | | - Ameena Rahamath
- Department of Biochemistry, mLAC, Bengaluru, Karnataka. India
| | | | - Arvind K Goyal
- Department of Biotechnology, Bodoland University, Kokrajhar783370, BTAD, Assam. India
| |
Collapse
|
21
|
Barry EL, Fedirko V, Baron JA. NSAIDs and Colorectal Cancer Phenotypes: What Now? J Natl Cancer Inst 2020; 111:440-441. [PMID: 30388268 DOI: 10.1093/jnci/djy174] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Accepted: 08/24/2018] [Indexed: 02/07/2023] Open
Affiliation(s)
- Elizabeth L Barry
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Veronika Fedirko
- Department of Epidemiology, Rollins School of Public Health, Winship Cancer Institute, Emory University, Atlanta, GA
| | - John A Baron
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Hanover, NH.,Department of Medicine, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC
| |
Collapse
|
22
|
Zarrinpar A, David Cheng TY, Huo Z. What Can We Learn About Drug Safety and Other Effects in the Era of Electronic Health Records and Big Data That We Would Not Be Able to Learn From Classic Epidemiology? J Surg Res 2019; 246:599-604. [PMID: 31653413 DOI: 10.1016/j.jss.2019.09.053] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/16/2019] [Accepted: 09/19/2019] [Indexed: 02/07/2023]
Abstract
As more and more health systems have converted to the use of electronic health records, the amount of searchable and analyzable data is exploding. This includes not just provider or laboratory created data but also data collected by instruments, personal devices, and patients themselves, among others. This has led to more attention being paid to the analysis of these data to answer previously unaddressed questions. This is especially important given the number of therapies previously found to be beneficial in clinical trials that are currently being re-scrutinized. Because there are orders of magnitude more information contained in these data sets, a fundamentally different approach needs to be taken to their processing and analysis and the generation of knowledge. Health care and medicine are drivers of this phenomenon and will ultimately be the main beneficiaries. Concurrently, many different types of questions can now be asked using these data sets. Research groups have become increasingly active in mining large data sets, including nationwide health care databases, to learn about associations of medication use and various unrelated diseases such as cancer. Given the recent increase in research activity in this area, its promise to radically change clinical research, and the relative lack of widespread knowledge about its potential and advances, we surveyed the available literature to understand the strengths and limitations of these new tools. We also outline new databases and techniques that are available to researchers worldwide, with special focus on work pertaining to the broad and rapid monitoring of drug safety and secondary effects.
Collapse
Affiliation(s)
- Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida.
| | - Ting-Yuan David Cheng
- Department of Epidemiology, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health & Health Professions and College of Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
23
|
Hybiak J, Broniarek I, Kiryczyński G, Los LD, Rosik J, Machaj F, Sławiński H, Jankowska K, Urasińska E. Aspirin and its pleiotropic application. Eur J Pharmacol 2019; 866:172762. [PMID: 31669590 DOI: 10.1016/j.ejphar.2019.172762] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/21/2019] [Accepted: 10/25/2019] [Indexed: 12/31/2022]
Abstract
Aspirin (acetylsalicylic acid), the oldest synthetic drug, was originally used as an anti-inflammatory medication. Being an irreversible inhibitor of COX (prostaglandin-endoperoxide synthase) enzymes that produce precursors for prostaglandins and thromboxanes, it has gradually found several other applications. Sometimes these applications are unrelated to its original purpose for example its use as an anticoagulant. Applications such as these have opened opportunities for new treatments. In this case, it has been tested in patients with cardiovascular disease to reduce the risk of myocardial infarct. Its function as an anticoagulant has also been explored in the prophylaxis and treatment of pre-eclampsia, where due to its anti-inflammatory properties, aspirin intake may be used to reduce the risk of colorectal cancer. It is important to always consider both the risks and benefits of aspirin's application. This is especially important for proposed use in the prevention and treatment of neurologic ailments like Alzheimer's disease, or in the prophylaxis of myocardial infarct. In such cases, the decision if aspirin should be applied, and at what dose may be guided by specific molecular markers. In this revived paper, the pleiotropic application of aspirin is summarized.
Collapse
Affiliation(s)
- Jolanta Hybiak
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland.
| | - Izabela Broniarek
- Department of Gene Expression, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University Poznan, Poland
| | - Gerard Kiryczyński
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Laura D Los
- Faculty of Science, University of Manitoba, Winnipeg, Canada
| | - Jakub Rosik
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Filip Machaj
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Hubert Sławiński
- Wellcome Centre for Human Genetics, University of Oxford, United Kingdom
| | - Kornelia Jankowska
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Elżbieta Urasińska
- Department of Pathology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
24
|
Hachim MY, Hachim IY, Elemam NM, Hamoudi RA. Toxicogenomic analysis of publicly available transcriptomic data can predict food, drugs, and chemical-induced asthma. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:181-199. [PMID: 31692590 PMCID: PMC6717055 DOI: 10.2147/pgpm.s217535] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/30/2019] [Indexed: 02/04/2023]
Abstract
Background : With the increasing incidence of asthma, more attention is focused on the diverse and complex nutritional and environmental triggers of asthma exacerbations. Currently, there are no established risk assessment tools to evaluate asthma triggering potentials of most of the nutritional and environmental triggers encountered by asthmatic patients. Purpose The objective of this study is to devise a reliable workflow, capable of estimating the toxicogenomic effect of such factors on key player genes in asthma pathogenesis. Methods Gene expression extracted from publicly available datasets of asthmatic bronchial epithelium were subjected to a comprehensive analysis of differential gene expression to identify significant genes involved in asthma development and progression. The identified genes were subjected to Gene Set Enrichment Analysis using a total of 31,826 gene sets related to chemical, toxins, and drugs to identify common agents that share similar asthma-related targets genes and signaling pathways. Results Our analysis identified 225 differentially expressed genes between severe asthmatic and healthy bronchial epithelium. Gene Set Enrichment Analysis of the identified genes showed that they are involved in response to toxic substances and organic cyclic compounds and are targeted by 41 specific diets, plants products, and plants related toxins (eg adenine, arachidonic acid, baicalein, caffeic acid, corilagin, curcumin, ellagic acid, luteolin, microcystin-RR, phytoestrogens, protoporphyrin IX, purpurogallin, rottlerin, and salazinic acid). Moreover, the identified chemicals share interesting inflammation-related pathways like NF-κB. Conclusion Our analysis was able to explain and predict the toxicity in terms of stimulating the differentially expressed genes between severe asthmatic and healthy epithelium. Such an approach can pave the way to generate a cost-effective and reliable source for asthma-specific toxigenic reports thus allowing the asthmatic patients, physicians, and medical researchers to be aware of the potential triggering factors with fatal consequences.
Collapse
Affiliation(s)
- Mahmood Yaseen Hachim
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Ibrahim Yaseen Hachim
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Noha M Elemam
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rifat A Hamoudi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates.,Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
25
|
Sajiki Y, Konnai S, Okagawa T, Nishimori A, Maekawa N, Goto S, Watari K, Minato E, Kobayashi A, Kohara J, Yamada S, Kaneko MK, Kato Y, Takahashi H, Terasaki N, Takeda A, Yamamoto K, Toda M, Suzuki Y, Murata S, Ohashi K. Prostaglandin E 2-Induced Immune Exhaustion and Enhancement of Antiviral Effects by Anti-PD-L1 Antibody Combined with COX-2 Inhibitor in Bovine Leukemia Virus Infection. THE JOURNAL OF IMMUNOLOGY 2019; 203:1313-1324. [PMID: 31366713 DOI: 10.4049/jimmunol.1900342] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/28/2019] [Indexed: 01/09/2023]
Abstract
Bovine leukemia virus (BLV) infection is a chronic viral infection of cattle and endemic in many countries, including Japan. Our previous study demonstrated that PGE2, a product of cyclooxygenase (COX) 2, suppresses Th1 responses in cattle and contributes to the progression of Johne disease, a chronic bacterial infection in cattle. However, little information is available on the association of PGE2 with chronic viral infection. Thus, we analyzed the changes in plasma PGE2 concentration during BLV infection and its effects on proviral load, viral gene transcription, Th1 responses, and disease progression. Both COX2 expression by PBMCs and plasma PGE2 concentration were higher in the infected cattle compared with uninfected cattle, and plasma PGE2 concentration was positively correlated with the proviral load. BLV Ag exposure also directly enhanced PGE2 production by PBMCs. Transcription of BLV genes was activated via PGE2 receptors EP2 and EP4, further suggesting that PGE2 contributes to disease progression. In contrast, inhibition of PGE2 production using a COX-2 inhibitor activated BLV-specific Th1 responses in vitro, as evidenced by enhanced T cell proliferation and Th1 cytokine production, and reduced BLV proviral load in vivo. Combined treatment with the COX-2 inhibitor meloxicam and anti-programmed death-ligand 1 Ab significantly reduced the BLV proviral load, suggesting a potential as a novel control method against BLV infection. Further studies using a larger number of animals are required to support the efficacy of this treatment for clinical application.
Collapse
Affiliation(s)
- Yamato Sajiki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan; .,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Asami Nishimori
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Shinya Goto
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Kei Watari
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Erina Minato
- Department of Veterinary Clinical Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Atsushi Kobayashi
- Department of Veterinary Clinical Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Junko Kohara
- Animal Research Center, Agriculture Research Department, Hokkaido Research Organization, Shintoku 081-0038, Japan
| | - Shinji Yamada
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Hirofumi Takahashi
- Shibecha Branch, Kushiro Central Office, Hokkaido Higashi Agricultural Mutual Aid Association, Shibecha 088-2311, Japan
| | - Nobuhiro Terasaki
- Shibecha Branch, Kushiro Central Office, Hokkaido Higashi Agricultural Mutual Aid Association, Shibecha 088-2311, Japan
| | - Akira Takeda
- Shibecha Branch, Kushiro Central Office, Hokkaido Higashi Agricultural Mutual Aid Association, Shibecha 088-2311, Japan
| | - Keiichi Yamamoto
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.,Research and Development Center, Fuso Pharmaceutical Industries, Ltd., Osaka 536-8523, Japan
| | - Mikihiro Toda
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.,New Business and International Business Development, Fuso Pharmaceutical Industries, Ltd., Osaka 536-8523, Japan
| | - Yasuhiko Suzuki
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.,Division of Bioresources, Research Center for Zoonosis Control, Hokkaido University, Sapporo 001-0019, Japan; and.,Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education, Hokkaido University, Sapporo 001-0019, Japan
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
26
|
Zhang X, Feng Y, Liu X, Ma J, Li Y, Wang T, Li X. Beyond a chemopreventive reagent, aspirin is a master regulator of the hallmarks of cancer. J Cancer Res Clin Oncol 2019; 145:1387-1403. [PMID: 31037399 DOI: 10.1007/s00432-019-02902-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 03/22/2019] [Indexed: 12/14/2022]
Abstract
PURPOSE Aspirin, one of the most commonly used nonsteroidal anti-inflammatory drugs (NAIDS), not only shows cancer chemoprevention effects but also improves cancer therapeutic effects when combined with other therapies. Studies that focus on aspirin regulation of the hallmarks of cancer and the associated molecular mechanisms facilitate a more thorough understanding of aspirin in mediating chemoprevention and may supply additional information for the development of novel cancer therapeutic agents. METHODS The relevant literatures from PubMed have been reviewed in this article. RESULTS Current studies have revealed that aspirin regulates almost all the hallmarks of cancer. Within tumor tissue, aspirin suppresses the bioactivities of cancer cells themselves and deteriorates the tumor microenvironment that supports cancer progression. In addition to tumor tissues, blocking of platelet activation also contributes to the ability of aspirin to inhibit cancer progression. In terms of the molecular mechanism, aspirin targets oncogenes and cancer-related signaling pathways and activates certain tumor suppressors. CONCLUSION Beyond a chemopreventive agent, aspirin is a master regulator of the hallmarks of cancer.
Collapse
Affiliation(s)
- Xiao Zhang
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Yukuan Feng
- Key Laboratory of Heilongjiang Province for Cancer Prevention and Control, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xi Liu
- Center of Cardiovascular Disease, Inner Mongolia People's Hospital, Hohhot, 010017, Inner Mongolia, China
| | - Jianhui Ma
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Yafei Li
- Department of Pathology, Harbin Medical University, Harbin, 150086, China
| | - Tianzhen Wang
- Department of Pathology, Harbin Medical University, Harbin, 150086, China.
| | - Xiaobo Li
- Department of Pathology, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
27
|
Kanikarla-Marie P, Kopetz S, Hawk ET, Millward SW, Sood AK, Gresele P, Overman M, Honn K, Menter DG. Bioactive lipid metabolism in platelet "first responder" and cancer biology. Cancer Metastasis Rev 2019; 37:439-454. [PMID: 30112590 DOI: 10.1007/s10555-018-9755-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Platelets can serve as "first responders" in cancer and metastasis. This is partly due to bioactive lipid metabolism that drives both platelet and cancer biology. The two primary eicosanoid metabolites that maintain platelet rapid response homeostasis are prostacyclin made by endothelial cells that inhibits platelet function, which is counterbalanced by thromboxane produced by platelets during activation, aggregation, and platelet recruitment. Both of these arachidonic acid metabolites are inherently unstable due to their chemical structure. Tumor cells by contrast predominantly make more chemically stable prostaglandin E2, which is the primary bioactive lipid associated with inflammation and oncogenesis. Pharmacological, clinical, and epidemiologic studies demonstrate that non-steroidal anti-inflammatory drugs (NSAIDs), which target cyclooxygenases, can help prevent cancer. Much of the molecular and biological impact of these drugs is generally accepted in the field. Cyclooxygenases catalyze the rate-limiting production of substrate used by all synthase molecules, including those that produce prostaglandins along with prostacyclin and thromboxane. Additional eicosanoid metabolites include lipoxygenases, leukotrienes, and resolvins that can also influence platelets, inflammation, and carcinogenesis. Our knowledge base and technology are now progressing toward identifying newer molecular and cellular interactions that are leading to revealing additional targets. This review endeavors to summarize new developments in the field.
Collapse
Affiliation(s)
- Preeti Kanikarla-Marie
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Scott Kopetz
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Ernest T Hawk
- Office of the Vice President Cancer Prevention and Population Science, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Steven W Millward
- Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Anil K Sood
- Gynocologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.,Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Paolo Gresele
- Department of Medicine, Section of Internal and Cardiovascular Medicine, University of Perugia, Via E. Dal Pozzo, 06126, Perugia, Italy
| | - Michael Overman
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA
| | - Kenneth Honn
- Bioactive Lipids Research Program, Department of Pathology, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA.,Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA.,Department of Gastrointestinal Medical Oncology, M. D. Anderson Cancer Center, 1515 Holcombe Boulevard--Unit 0426, Houston, TX, 77030, USA
| | - David G Menter
- Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77054, USA.
| |
Collapse
|
28
|
Ogino S, Nowak JA, Hamada T, Milner DA, Nishihara R. Insights into Pathogenic Interactions Among Environment, Host, and Tumor at the Crossroads of Molecular Pathology and Epidemiology. ANNUAL REVIEW OF PATHOLOGY 2019; 14:83-103. [PMID: 30125150 PMCID: PMC6345592 DOI: 10.1146/annurev-pathmechdis-012418-012818] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Evidence indicates that diet, nutrition, lifestyle, the environment, the microbiome, and other exogenous factors have pathogenic roles and also influence the genome, epigenome, transcriptome, proteome, and metabolome of tumor and nonneoplastic cells, including immune cells. With the need for big-data research, pathology must transform to integrate data science fields, including epidemiology, biostatistics, and bioinformatics. The research framework of molecular pathological epidemiology (MPE) demonstrates the strengths of such an interdisciplinary integration, having been used to study breast, lung, prostate, and colorectal cancers. The MPE research paradigm not only can provide novel insights into interactions among environment, tumor, and host but also opens new research frontiers. New developments-such as computational digital pathology, systems biology, artificial intelligence, and in vivo pathology technologies-will further transform pathology and MPE. Although it is necessary to address the rarity of transdisciplinary education and training programs, MPE provides an exemplary model of integrative scientific approaches and contributes to advancements in precision medicine, therapy, and prevention.
Collapse
Affiliation(s)
- Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02215, USA; , ,
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, Massachusetts 02215, USA;
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02215, USA; , ,
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, and Harvard Medical School, Boston, Massachusetts 02215, USA;
| | - Danny A Milner
- American Society for Clinical Pathology, Chicago, Illinois 60603, USA;
| | - Reiko Nishihara
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02215, USA; , ,
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02142, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
29
|
Lordan R, Tsoupras A, Zabetakis I. The Potential Role of Dietary Platelet-Activating Factor Inhibitors in Cancer Prevention and Treatment. Adv Nutr 2019; 10:148-164. [PMID: 30721934 PMCID: PMC6370273 DOI: 10.1093/advances/nmy090] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/11/2018] [Accepted: 10/10/2018] [Indexed: 12/11/2022] Open
Abstract
Cancer is the second leading cause of mortality worldwide. The role of unresolved inflammation in cancer progression and metastasis is well established. Platelet-activating factor (PAF) is a key proinflammatory mediator in the initiation and progression of cancer. Evidence suggests that PAF is integral to suppression of the immune system and promotion of metastasis and tumor growth by altering local angiogenic and cytokine networks. Interactions between PAF and its receptor may have a role in various digestive, skin, and hormone-dependent cancers. Diet plays a critical role in the prevention of cancer and its treatment. Research indicates that the Mediterranean diet may reduce the incidence of several cancers in which dietary PAF inhibitors have a role. Dietary PAF inhibitors such as polar lipids have demonstrated inhibitory effects against the physiological actions of PAF in cancer and other chronic inflammatory conditions in vitro and in vivo. In addition, experimental models of radiotherapy and chemotherapy demonstrate that inhibition of PAF as adjuvant therapy may lead to more favorable outcomes. Although promising, there is limited evidence on the potential benefits of dietary PAF inhibitors on cancer prevention or treatment. Therefore, further extensive research is required to assess the effects of various dietary factors and PAF inhibitors and to elucidate the mechanisms in prevention of cancer progression and metastasis at a molecular level.
Collapse
Affiliation(s)
- Ronan Lordan
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Alexandros Tsoupras
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| | - Ioannis Zabetakis
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
| |
Collapse
|
30
|
van Elsland D, Neefjes J. Bacterial infections and cancer. EMBO Rep 2018; 19:embr.201846632. [PMID: 30348892 PMCID: PMC6216254 DOI: 10.15252/embr.201846632] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/10/2018] [Accepted: 09/24/2018] [Indexed: 12/30/2022] Open
Abstract
Infections are estimated to contribute to 20% of all human tumours. These are mainly caused by viruses, which explains why a direct bacterial contribution to cancer formation has been largely ignored. While epidemiological data link bacterial infections to particular cancers, tumour formation is generally assumed to be solely caused by the ensuing inflammation responses. Yet, many bacteria directly manipulate their host cell in various phases of their infection cycle. Such manipulations can affect host cell integrity and can contribute to cancer formation. We here describe how bacterial surface moieties, bacterial protein toxins and bacterial effector proteins can induce host cell DNA damage, and thereby can interfere with essential host cell signalling pathways involved in cell proliferation, apoptosis, differentiation and immune signalling.
Collapse
Affiliation(s)
- Daphne van Elsland
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| | - Jacques Neefjes
- Oncode Institute and Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Leiden, The Netherlands
| |
Collapse
|
31
|
Abstract
In recent years, the reports on using nonsteroidal anti-inflammatory drugs (NSAIDs) for cancer prevention and treatment have been on the rise. In 2017, the US Preventive Services Working Group issued primary prevention guidelines on the use of NSAIDs, especially aspirin, for cardiovascular disease and colorectal cancer, and formally established the role and status of aspirin in cancer prevention. However, the mechanism of NSAIDs on preventing cancer is still not clear. In this paper, the progress of the application of NSAIDs, especially aspirin, in the prevention and treatment of tumors in recent years is summarized, and new ideas and directions for the follow-up study are also discussed.
Collapse
Affiliation(s)
- Zhen Zhang
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, , .,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, ,
| | - Fulin Chen
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, , .,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, ,
| | - Lijun Shang
- Laboratory of Tissue Engineering, Faculty of Life Science, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, , .,Provincial Key Laboratory of Biotechnology of Shaanxi, Northwest University, Xi'an, Shaanxi Province 710069, People's Republic of China, , .,School of Chemistry and Biosciences, Faculty of Life Sciences, University of Bradford, Bradford, UK,
| |
Collapse
|
32
|
Wang J, Cho NL, Zauber AG, Hsu M, Dawson D, Srivastava A, Mitchell-Richards KA, Markowitz SD, Bertagnolli MM. Chemopreventive Efficacy of the Cyclooxygenase-2 (Cox-2) Inhibitor, Celecoxib, Is Predicted by Adenoma Expression of Cox-2 and 15-PGDH. Cancer Epidemiol Biomarkers Prev 2018; 27:728-736. [PMID: 29769213 PMCID: PMC6519921 DOI: 10.1158/1055-9965.epi-17-0573] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 12/05/2017] [Accepted: 04/23/2018] [Indexed: 12/19/2022] Open
Abstract
Background: The Adenoma Prevention with Celecoxib (APC) Trial showed that cyclooxygenase-2 (Cox-2) inhibitor, celecoxib, decreased adenoma development in patients at high risk for colorectal cancer. A prospectively planned analysis of the APC Trial tested the hypothesis that expression of target enzymes in adenomas removed before beginning study treatment would identify individuals at high risk of adenoma development, and/or predict response to Cox-2 inhibition.Methods: Pre-treatment adenomas were examined using immunohistochemistry to assess expression of Cox-2 (high vs. low) and 15-prostaglandin dehydrogenase (15-PGDH, presence vs. loss). The Mantel-Cox test evaluated whether these markers predicted benefit from celecoxib for reduction of adenoma detection.Results: Patients whose pre-treatment adenomas demonstrated elevated Cox-2 achieved the greatest adenoma reduction with celecoxib treatment [RR, 0.37; 95% confidence interval (CI), 0.22-0.61; P = 0.0001]. This reduction was less in the low Cox-2 category (RR, 0.64; 95% CI, 0.56-0.73). Patients whose pre-treatment adenomas showed 15-PGDH loss had a similar treatment-associated reduction in adenoma detection (RR, 0.60; 95% CI, 0.52-0.69; P < 0.0001). In contrast, patients with intact tumor 15-PGDH expression did not significantly benefit from celecoxib (RR, 0.73; 95% CI, 0.47-1.12; P = 0.15). However, subset analysis suggested that this lack of response to celecoxib was confined to those patients with 15-PGDH intact tumors who were also using cardioprotective aspirin.Conclusions: The expression of Cox-2 and 15-PGDH in pre-treatment adenomas provides predictive information in patients treated with celecoxib for prevention of colorectal adenomas.Impact: The results of this study show that Cox-2 and 15-PGDH are characteristics of colorectal adenomas that may be used to predict nonsteroidal anti-inflammatory drug chemoprevention efficacy. Cancer Epidemiol Biomarkers Prev; 27(7); 728-36. ©2018 AACR.
Collapse
Affiliation(s)
- Jiping Wang
- Department of Surgery, Division of Surgical Oncology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Nancy L Cho
- Department of Surgery, Division of Surgical Oncology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Ann G Zauber
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Meier Hsu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Dawn Dawson
- Department of Pathology, and Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - Amitabh Srivastava
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | | | - Sanford D Markowitz
- Departments of Medicine and Case Comprehensive Cancer Center, Case Western Reserve University, and University Hospitals Seidman Cancer Center, Cleveland, Ohio
| | - Monica M Bertagnolli
- Department of Surgery, Division of Surgical Oncology, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
33
|
Leenders EKSM, Westdorp H, Brüggemann RJ, Loeffen J, Kratz C, Burn J, Hoogerbrugge N, Jongmans MCJ. Cancer prevention by aspirin in children with Constitutional Mismatch Repair Deficiency (CMMRD). Eur J Hum Genet 2018; 26:1417-1423. [PMID: 29904176 DOI: 10.1038/s41431-018-0197-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 05/18/2018] [Accepted: 05/22/2018] [Indexed: 12/16/2022] Open
Abstract
Constitutional MisMatch Repair Deficiency (CMMRD) is caused by homozygous or compound heterozygous germline variants in one of the mismatch repair (MMR) genes (MSH2, MSH6, PMS2, MLH1). This syndrome results in early onset colorectal cancer, leukemia and lymphoma, brain tumors and other malignancies. Children with CMMRD are at high risk of developing multiple cancers and cancer surveillance does not guarantee detection of cancer at a curable stage. The development of a preventive treatment strategy would be a major step forward. Long-term daily use of acetylsalicylic acid (ASA) has been shown to reduce cancer risk in individuals with Lynch syndrome (LS). LS is caused by heterozygous germline variants of MSH2, MSH6, PMS2 and MLH1 and characterized by an increased risk of developing colorectal and endometrial cancer at adult age. Here we discuss the potential use of ASA for cancer prevention in patients with CMMRD.
Collapse
Affiliation(s)
- Erika K S M Leenders
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Harm Westdorp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Roger J Brüggemann
- Department of Pharmacy, Radboud University Nijmegen Medical Centre, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Jan Loeffen
- Department of Pediatric Oncology, Erasmus MC - Sophia Children's Hospital, Rotterdam, The Netherlands.,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Christian Kratz
- Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - John Burn
- Institute of Genetic Medicine Newcastle University, Newcastle upon Tyne, UK
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands
| | - Marjolijn C J Jongmans
- Department of Human Genetics, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Nijmegen, The Netherlands. .,Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands. .,Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
34
|
Ogino S, Nowak JA, Hamada T, Phipps AI, Peters U, Milner DA, Giovannucci EL, Nishihara R, Giannakis M, Garrett WS, Song M. Integrative analysis of exogenous, endogenous, tumour and immune factors for precision medicine. Gut 2018; 67:1168-1180. [PMID: 29437869 PMCID: PMC5943183 DOI: 10.1136/gutjnl-2017-315537] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/02/2018] [Accepted: 01/05/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy strategies targeting immune checkpoints such as the CTLA4 and CD274 (programmed cell death 1 ligand 1, PD-L1)/PDCD1 (programmed cell death 1, PD-1) T-cell coreceptor pathways are revolutionising oncology. The approval of pembrolizumab use for solid tumours with high-level microsatellite instability or mismatch repair deficiency by the US Food and Drug Administration highlights promise of precision immuno-oncology. However, despite evidence indicating influences of exogenous and endogenous factors such as diet, nutrients, alcohol, smoking, obesity, lifestyle, environmental exposures and microbiome on tumour-immune interactions, integrative analyses of those factors and immunity lag behind. Immune cell analyses in the tumour microenvironment have not adequately been integrated into large-scale studies. Addressing this gap, the transdisciplinary field of molecular pathological epidemiology (MPE) offers research frameworks to integrate tumour immunology into population health sciences, and link the exposures and germline genetics (eg, HLA genotypes) to tumour and immune characteristics. Multilevel research using bioinformatics, in vivo pathology and omics (genomics, epigenomics, transcriptomics, proteomics and metabolomics) technologies is possible with use of tissue, peripheral blood circulating cells, cell-free plasma, stool, sputum, urine and other body fluids. This immunology-MPE model can synergise with experimental immunology, microbiology and systems biology. GI neoplasms represent exemplary diseases for the immunology-MPE model, given rich microbiota and immune tissues of intestines, and the well-established carcinogenic role of intestinal inflammation. Proof-of-principle studies on colorectal cancer provided insights into immunomodulating effects of aspirin, vitamin D, inflammatory diets and omega-3 polyunsaturated fatty acids. The integrated immunology-MPE model can contribute to better understanding of environment-tumour-immune interactions, and effective immunoprevention and immunotherapy strategies for precision medicine.
Collapse
Affiliation(s)
- Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA,Department of Epidemiology, University of Washington, Seattle, Washington, USA
| | - Danny A Milner
- American Society for Clinical Pathology, Chicago, Illinois, USA
| | - Edward L Giovannucci
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Reiko Nishihara
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Marios Giannakis
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA,Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Wendy S Garrett
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA,Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Mingyang Song
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, Massachusetts, USA,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
35
|
Abstract
Chronic inflammation is a risk factor for gastrointestinal cancer and other diseases. Most studies have focused on cytokines and chemokines as mediators connecting chronic inflammation to cancer, whereas the involvement of lipid mediators, including prostanoids, has not been extensively investigated. Prostanoids are among the earliest signaling molecules released in response to inflammation. Multiple lines of evidence suggest that prostanoids are involved in gastrointestinal cancer. In this Review, we discuss how prostanoids impact gastrointestinal cancer development. In particular, we highlight recent advances in our understanding of how prostaglandin E2 induces the immunosuppressive microenvironment in gastrointestinal cancers.
Collapse
Affiliation(s)
- Dingzhi Wang
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Raymond N DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina, USA.,Department of Research and Division of Gastroenterology, Mayo Clinic, Scottsdale, Arizona, USA
| |
Collapse
|
36
|
Vestibular Schwannoma Growth With Aspirin and Other Nonsteroidal Anti-inflammatory Drugs. Otol Neurotol 2018; 38:1158-1164. [PMID: 28692590 DOI: 10.1097/mao.0000000000001506] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate whether the use of aspirin or other non-steroidal anti-inflammatory drugs (NSAIDs) impact the growth of vestibular schwannoma (VS). STUDY DESIGN Retrospective case series. SETTING Single academic, tertiary care center. PATIENTS Patients with VS who underwent at least two magnetic resonance imaging (MRI) studies before intervention. INTERVENTION(S) Serial MRI studies. MAIN OUTCOME MEASURE(S) VS tumor growth, defined as more than or equal to 2 mm increase in the maximum tumor diameter between consecutive MRI studies, or between the first and last available study. Mean growth rate was also calculated, defined as the change in tumor size divided by length of follow-up. RESULTS A total of 564 VS patients met inclusion criteria, with 234 (41.2%) taking some type of NSAID. Aspirin use was not associated with VS tumor growth, presenting tumor diameter, or mean VS growth rate. Further, aspirin dosage did not impact growth outcomes or presenting tumor diameter. A total of 96 (17.0%) patients took an NSAID other than aspirin. Neither non-aspirin NSAID use nor degree of cyclooxygenase-2 (COX-2) selectivity, including aspirin, was significantly associated with VS tumor growth, presenting tumor diameter, or mean VS growth rate. CONCLUSIONS While previous studies have suggested a relationship between aspirin usage and VS growth, we found no significant association in our series of 564 observed VS. Furthermore, there was no apparent relationship between aspirin dosage, non-aspirin NSAID use, and COX-2 selectivity with VS growth, presenting tumor diameter at presentation, or mean VS growth rate.
Collapse
|
37
|
Hu Y, Yu K, Wang G, Zhang D, Shi C, Ding Y, Hong D, Zhang D, He H, Sun L, Zheng JN, Sun S, Qian F. Lanatoside C inhibits cell proliferation and induces apoptosis through attenuating Wnt/β-catenin/c-Myc signaling pathway in human gastric cancer cell. Biochem Pharmacol 2018; 150:280-292. [DOI: 10.1016/j.bcp.2018.02.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 02/16/2018] [Indexed: 02/06/2023]
|
38
|
Siena L, Cipollina C, Di Vincenzo S, Ferraro M, Bruno A, Gjomarkaj M, Pace E. Electrophilic derivatives of omega-3 fatty acids counteract lung cancer cell growth. Cancer Chemother Pharmacol 2018; 81:705-716. [PMID: 29435611 DOI: 10.1007/s00280-018-3538-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/03/2018] [Indexed: 12/22/2022]
Abstract
PURPOSE 17-oxo-DHA is an electrophilic keto-derivative of the omega-3 fatty acid docosahexaenoic acid (DHA) endogenously generated by cyclooxygenase-2 and a cellular dehydrogenase. 17-oxo-DHA displays anti-inflammatory and cytoprotective actions. DHA, alone or in combination with standard chemotherapy, displays antitumor activity. However, the effects of electrophilic keto-derivatives of DHA on cancer growth have never been evaluated. We investigated whether 17-oxo-DHA, alone or in combination with gemcitabine, displayed antitumor effects. Furthermore, we evaluated whether the enzyme 15-prostaglandin dehydrogenase (15-PGDH) was required for transducing the antitumor effects of DHA. METHODS A panel of five histologically different human non-small cell lung cancer (NSCLC) cell lines was used. Cells were treated with 17-oxo-DHA and gemcitabine, alone or in combination, and apoptosis, proliferation, Fas and FasL expression (mRNA and protein) and active caspase-3/7 and -8 were assessed. Furthermore, an inhibitor of 15-PGDH was used to test the involvement of this enzyme in mediating the antitumor effects of DHA. RESULTS 17-oxo-DHA (50 µM, 72 h) significantly reduced proliferation, increased cell apoptosis, Fas and FasL expression as well as active caspase-8 and -3/7. When 17-oxo-DHA was given in combination with gemcitabine, stronger effects were observed compared to gemcitabine alone. The enzyme 15-PGDH was required for DHA to promote its full anti-apoptotic effect suggesting that enzymatically generated keto-derivatives of DHA mediate its antitumor actions. CONCLUSIONS Data herein provided, demonstrate that 17-oxo-DHA displays antitumor effects in NSCLC cell lines. Of note, the combination of 17-oxo-DHA plus gemcitabine, resulted in stronger anticancer effects compared to gemcitabine alone.
Collapse
Affiliation(s)
- Liboria Siena
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy
| | - Chiara Cipollina
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy.,Fondazione Ri.MED, Palermo, Italy
| | - Serena Di Vincenzo
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy
| | - Maria Ferraro
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy
| | - Andreina Bruno
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy
| | - Mark Gjomarkaj
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy
| | - Elisabetta Pace
- Istituto di Biomedicina e Immunologia Molecolare-Consiglio Nazionale delle Ricerche, Via Ugo La Malfa, 153, 90146, Palermo, Italy.
| |
Collapse
|
39
|
Review Article: The Role of Molecular Pathological Epidemiology in the Study of Neoplastic and Non-neoplastic Diseases in the Era of Precision Medicine. Epidemiology 2018; 27:602-11. [PMID: 26928707 DOI: 10.1097/ede.0000000000000471] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Molecular pathology diagnostics to subclassify diseases based on pathogenesis are increasingly common in clinical translational medicine. Molecular pathological epidemiology (MPE) is an integrative transdisciplinary science based on the unique disease principle and the disease continuum theory. While it has been most commonly applied to research on breast, lung, and colorectal cancers, MPE can investigate etiologic heterogeneity in non-neoplastic diseases, such as cardiovascular diseases, obesity, diabetes mellitus, drug toxicity, and immunity-related and infectious diseases. This science can enhance causal inference by linking putative etiologic factors to specific molecular biomarkers as outcomes. Technological advances increasingly enable analyses of various -omics, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, metagenomics, microbiome, immunomics, interactomics, etc. Challenges in MPE include sample size limitations (depending on availability of biospecimens or biomedical/radiological imaging), need for rigorous validation of molecular assays and study findings, and paucities of interdisciplinary experts, education programs, international forums, and standardized guidelines. To address these challenges, there are ongoing efforts such as multidisciplinary consortium pooling projects, the International Molecular Pathological Epidemiology Meeting Series, and the Strengthening the Reporting of Observational Studies in Epidemiology-MPE guideline project. Efforts should be made to build biorepository and biobank networks, and worldwide population-based MPE databases. These activities match with the purposes of the Big Data to Knowledge (BD2K), Genetic Associations and Mechanisms in Oncology (GAME-ON), and Precision Medicine Initiatives of the United States National Institute of Health. Given advances in biotechnology, bioinformatics, and computational/systems biology, there are wide open opportunities in MPE to contribute to public health.
Collapse
|
40
|
Inamura K. Colorectal Cancers: An Update on Their Molecular Pathology. Cancers (Basel) 2018; 10:cancers10010026. [PMID: 29361689 PMCID: PMC5789376 DOI: 10.3390/cancers10010026] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancers (CRCs) are the third leading cause of cancer-related mortality worldwide. Rather than being a single, uniform disease type, accumulating evidence suggests that CRCs comprise a group of molecularly heterogeneous diseases that are characterized by a range of genomic and epigenomic alterations. This heterogeneity slows the development of molecular-targeted therapy as a form of precision medicine. Recent data regarding comprehensive molecular characterizations and molecular pathological examinations of CRCs have increased our understanding of the genomic and epigenomic landscapes of CRCs, which has enabled CRCs to be reclassified into biologically and clinically meaningful subtypes. The increased knowledge of the molecular pathological epidemiology of CRCs has permitted their evolution from a vaguely understood, heterogeneous group of diseases with variable clinical courses to characteristic molecular subtypes, a development that will allow the implementation of personalized therapies and better management of patients with CRC. This review provides a perspective regarding recent developments in our knowledge of the molecular and epidemiological landscapes of CRCs, including results of comprehensive molecular characterizations obtained from high-throughput analyses and the latest developments regarding their molecular pathologies, immunological biomarkers, and associated gut microbiome. Advances in our understanding of potential personalized therapies for molecularly specific subtypes are also reviewed.
Collapse
Affiliation(s)
- Kentaro Inamura
- Division of Pathology, The Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan.
| |
Collapse
|
41
|
Barker EC, Kim BG, Yoon JH, Tochtrop GP, Letterio JJ, Choi SH. Potent suppression of both spontaneous and carcinogen-induced colitis-associated colorectal cancer in mice by dietary celastrol supplementation. Carcinogenesis 2018; 39:36-46. [PMID: 29069290 PMCID: PMC5862246 DOI: 10.1093/carcin/bgx115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/22/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022] Open
Abstract
Celastrol is an anti-inflammatory natural triterpenoid, isolated from the herb Tripterygium wilfordii or thunder god vine. Here, we define mechanisms mediating anti-inflammatory activity of celastrol and demonstrate efficacy of a dietary celastrol supplement for chemoprevention of inflammation-driven carcinogenesis in mice. Dietary celastrol (31.25 ppm in rodent diet from 8 weeks to 25 weeks of age) is well tolerated and protects against LPS-induced acute inflammation in C57BL/6 mice, potently suppressing LPS-induction of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, Interleukin (IL)-6 and IL-1β. To test whether dietary celastrol suppresses inflammation-driven colorectal cancer (CRC), we employed a unique model of spontaneous, inflammation-driven CRC in mice harboring a germ line deletion of the p27Kip1 gene and a T cell-specific deletion of Smad4 gene (Smad4co/co;Lck-crep27Kip1-/-or DKO), which develop severe intestinal inflammation and carcinogenesis as early as 3 months of age. Exposure of DKO mice to daily dietary celastrol (12.5 ppm in diet) from 6 weeks of age significantly suppressed development of colitis-associated CRC (CAC). Celastrol chemoprevention of CAC in this new model of intestinal neoplasia was associated with significant suppression of iNOS at 4 months of age, and iNOS, COX-2 and NFκB at 6 months of age, with significant reduction in inflammatory cytokines, IL-6 and IL-1β. Chemoprevetion of CAC by dietary celastrol was further confirmed in the model of azoxymethane (AOM) plus dextran sodium sulfate (DSS)-induced carcinogenesis in C57BL/6 mice. These data suggest the potential for celastrol as a safe and effective dietary supplement in the chemoprevention of CAC in humans.
Collapse
Affiliation(s)
- Emily C Barker
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
| | - Byung-Gyu Kim
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Ji Hee Yoon
- University of Notre Dame, Notre Dame, Indiana
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
- The Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - John J Letterio
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
- The Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
- The Angie Fowler Adolescent & Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children’s Hospital, Cleveland, Ohio
| | - Sung Hee Choi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
42
|
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths. Understanding its pathophysiology is essential for developing efficient strategies to treat this disease. Lipidome, the sum of total lipids, related enzymes, receptors and signaling pathways, plays crucial roles in multiple cellular processes, such as metabolism, energy storage, proliferation and apoptosis. Dysregulation of lipid metabolism and function contributes to the development of CRC, and can be used towards the evaluation of prognosis. The strategies targeting lipidome have been applied in clinical trails and showed promising results. Here we discuss recent advances in abnormal lipid metabolism in CRC, the mechanisms by which the lipidome regulates tumorigenesis and tumor progression, and suggest potential therapeutic targets for clinical trials.
Collapse
|
43
|
Ogino S, Jhun I, Mata DA, Soong TR, Hamada T, Liu L, Nishihara R, Giannakis M, Cao Y, Manson JE, Nowak JA, Chan AT. Integration of pharmacology, molecular pathology, and population data science to support precision gastrointestinal oncology. NPJ Precis Oncol 2017; 1. [PMID: 29552640 PMCID: PMC5856171 DOI: 10.1038/s41698-017-0042-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Precision medicine has a goal of customizing disease prevention and treatment strategies. Under the precision medicine paradigm, each patient has unique pathologic processes resulting from cellular genomic, epigenomic, proteomic, and metabolomic alterations, which are influenced by pharmacological, environmental, microbial, dietary, and lifestyle factors. Hence, to realize the promise of precision medicine, multi-level research methods that can comprehensively analyze many of these variables are needed. In order to address this gap, the integrative field of molecular pathology and population data science (i.e., molecular pathological epidemiology) has been developed to enable such multi-level analyses, especially in gastrointestinal cancer research. Further integration of pharmacology can improve our understanding of drug effects, and inform decision-making of drug use at both the individual and population levels. Such integrative research demonstrated potential benefits of aspirin in colorectal carcinoma with PIK3CA mutations, providing the basis for new clinical trials. Evidence also suggests that HPGD (15-PDGH) expression levels in normal colon and the germline rs6983267 polymorphism that relates to tumor CTNNB1 (β-catenin)/WNT signaling status may predict the efficacy of aspirin for cancer chemoprevention. As immune checkpoint blockade targeting the CD274 (PD-L1)/PDCD1 (PD-1) pathway for microsatellite instability-high (or mismatch repair-deficient) metastatic gastrointestinal or other tumors has become standard of care, potential modifying effects of dietary, lifestyle, microbial, and environmental factors on immunotherapy need to be studied to further optimize treatment strategies. With its broad applicability, our integrative approach can provide insights into the interactive role of medications, exposures, and molecular pathology, and guide the development of precision medicine.
Collapse
Affiliation(s)
- Shuji Ogino
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Iny Jhun
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Douglas A Mata
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Thing Rinda Soong
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tsuyoshi Hamada
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Li Liu
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Reiko Nishihara
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marios Giannakis
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yin Cao
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - JoAnn E Manson
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA.,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.,Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
44
|
Grancher A, Michel P, Di Fiore F, Sefrioui D. [Aspirin and colorectal cancer]. Bull Cancer 2017; 105:171-180. [PMID: 29153543 DOI: 10.1016/j.bulcan.2017.09.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 08/18/2017] [Accepted: 09/08/2017] [Indexed: 12/17/2022]
Abstract
Colorectal cancer is a worldwide public health problem. Aspirin has been identified as a protective factor against the apparition of colorectal cancer. There are several mechanisms about the actions by aspirin on colorectal tumorogenesis. These are not perfectly known nowadays. On one hand, there are direct mechanisms on colorectal mucosa, on the other hand there are indirect mechanisms through platelet functions. Aspirin also plays a role by its anti-inflammatory action and the stimulation of antitumor immunity. Several studies show that long-term treatment with low-doses of aspirin decreases the incidence of adenomas and colorectal cancers. In the United States, aspirin is currently recommended for primary prevention of the risk of colorectal cancer in all patients aged 50 to 59, with a 10-year risk of cardiovascular event greater than 10 %. However, primary prevention with aspirin should not be a substitute for screening in colorectal cancer. Furthermore, aspirin seems to be beneficial when used in post-diagnosis of colorectal cancer. It could actually decrease the risk of metastasis in case of a localized colorectal cancer, and increase the survival in particular, concerning PIK3CA mutated tumors. The association of aspirin with neoadjuvant treatment of colorectal cancer by radiochimiotherapy seems to have beneficial effects. French prospective randomized study is currently being conducted to investigate postoperative aspirin in colorectal cancers with a PIK3CA mutation.
Collapse
Affiliation(s)
- Adrien Grancher
- Normandie université, UNIROUEN, hôpital universitaire de Rouen, service d'hépato-gastroenterologie, 76000 Rouen, France
| | - Pierre Michel
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, service d'hépato-gastroentérologie, 76000 Rouen, France.
| | - Frédéric Di Fiore
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, centre Henri-Becquerel, département d'oncolgie médicale, service d'hépato-gastroentérologie, 76000 Rouen, France
| | - David Sefrioui
- Normandie université, UNIROUEN, Inserm 1245, IRON group, hôpital universitaire de Rouen, service d'hépato-gastroentérologie, 76000 Rouen, France
| |
Collapse
|
45
|
Jaffee EM, Dang CV, Agus DB, Alexander BM, Anderson KC, Ashworth A, Barker AD, Bastani R, Bhatia S, Bluestone JA, Brawley O, Butte AJ, Coit DG, Davidson NE, Davis M, DePinho RA, Diasio RB, Draetta G, Frazier AL, Futreal A, Gambhir SS, Ganz PA, Garraway L, Gerson S, Gupta S, Heath J, Hoffman RI, Hudis C, Hughes-Halbert C, Ibrahim R, Jadvar H, Kavanagh B, Kittles R, Le QT, Lippman SM, Mankoff D, Mardis ER, Mayer DK, McMasters K, Meropol NJ, Mitchell B, Naredi P, Ornish D, Pawlik TM, Peppercorn J, Pomper MG, Raghavan D, Ritchie C, Schwarz SW, Sullivan R, Wahl R, Wolchok JD, Wong SL, Yung A. Future cancer research priorities in the USA: a Lancet Oncology Commission. Lancet Oncol 2017; 18:e653-e706. [PMID: 29208398 PMCID: PMC6178838 DOI: 10.1016/s1470-2045(17)30698-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 12/12/2022]
Abstract
We are in the midst of a technological revolution that is providing new insights into human biology and cancer. In this era of big data, we are amassing large amounts of information that is transforming how we approach cancer treatment and prevention. Enactment of the Cancer Moonshot within the 21st Century Cures Act in the USA arrived at a propitious moment in the advancement of knowledge, providing nearly US$2 billion of funding for cancer research and precision medicine. In 2016, the Blue Ribbon Panel (BRP) set out a roadmap of recommendations designed to exploit new advances in cancer diagnosis, prevention, and treatment. Those recommendations provided a high-level view of how to accelerate the conversion of new scientific discoveries into effective treatments and prevention for cancer. The US National Cancer Institute is already implementing some of those recommendations. As experts in the priority areas identified by the BRP, we bolster those recommendations to implement this important scientific roadmap. In this Commission, we examine the BRP recommendations in greater detail and expand the discussion to include additional priority areas, including surgical oncology, radiation oncology, imaging, health systems and health disparities, regulation and financing, population science, and oncopolicy. We prioritise areas of research in the USA that we believe would accelerate efforts to benefit patients with cancer. Finally, we hope the recommendations in this report will facilitate new international collaborations to further enhance global efforts in cancer control.
Collapse
Affiliation(s)
| | - Chi Van Dang
- Ludwig Institute for Cancer Research New York, NY; Wistar Institute, Philadelphia, PA, USA.
| | - David B Agus
- University of Southern California, Beverly Hills, CA, USA
| | - Brian M Alexander
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Alan Ashworth
- University of California San Francisco, San Francisco, CA, USA
| | | | - Roshan Bastani
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Sangeeta Bhatia
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeffrey A Bluestone
- University of California San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | | | - Atul J Butte
- University of California San Francisco, San Francisco, CA, USA
| | - Daniel G Coit
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center and University of Washington, Seattle, WA, USA
| | - Mark Davis
- California Institute for Technology, Pasadena, CA, USA
| | | | | | - Giulio Draetta
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - A Lindsay Frazier
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Andrew Futreal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Patricia A Ganz
- Fielding School of Public Health and the Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Levi Garraway
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; The Broad Institute, Cambridge, MA, USA; Eli Lilly and Company, Boston, MA, USA
| | | | - Sumit Gupta
- Division of Haematology/Oncology, Hospital for Sick Children, Faculty of Medicine and IHPME, University of Toronto, Toronto, Canada
| | - James Heath
- California Institute for Technology, Pasadena, CA, USA
| | - Ruth I Hoffman
- American Childhood Cancer Organization, Beltsville, MD, USA
| | - Cliff Hudis
- Breast Cancer Medicine Service, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Chanita Hughes-Halbert
- Medical University of South Carolina and the Hollings Cancer Center, Charleston, SC, USA
| | - Ramy Ibrahim
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Hossein Jadvar
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brian Kavanagh
- Department of Radiation Oncology, University of Colorado, Denver, CO, USA
| | - Rick Kittles
- College of Medicine, University of Arizona, Tucson, AZ, USA; University of Arizona Cancer Center, University of Arizona, Tucson, AZ, USA
| | | | - Scott M Lippman
- University of California San Diego Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - David Mankoff
- Department of Radiology and Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elaine R Mardis
- The Institute for Genomic Medicine at Nationwide Children's Hospital Columbus, OH, USA; College of Medicine, Ohio State University, Columbus, OH, USA
| | - Deborah K Mayer
- University of North Carolina Lineberger Cancer Center, Chapel Hill, NC, USA
| | - Kelly McMasters
- The Hiram C Polk Jr MD Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | | | | | - Peter Naredi
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dean Ornish
- University of California San Francisco, San Francisco, CA, USA
| | - Timothy M Pawlik
- Department of Surgery, Wexner Medical Center, Ohio State University, Columbus, OH, USA
| | | | - Martin G Pomper
- The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Derek Raghavan
- Levine Cancer Institute, Carolinas HealthCare, Charlotte, NC, USA
| | | | - Sally W Schwarz
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | | | - Richard Wahl
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Jedd D Wolchok
- Ludwig Center for Cancer Immunotherapy, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Sandra L Wong
- Department of Surgery, The Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Alfred Yung
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
46
|
Chalouhi N, Jabbour P, Zanaty M, Starke RM, Torner J, Nakagawa D, Hasan DM. Sex Differential in 15-Hydroxyprostaglandin Dehydrogenase Levels in the Lumen of Human Intracranial Aneurysms. J Am Heart Assoc 2017; 6:e006639. [PMID: 29042428 PMCID: PMC5721867 DOI: 10.1161/jaha.117.006639] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 08/14/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND Aspirin is a promising medical therapy for the prevention of intracranial aneurysm (IA) rupture. Recently, we found that men have a better response to aspirin than women. The purpose of this study was to determine whether a sex differential exists in the level of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in the lumen of human IAs. METHODS AND RESULTS Consecutive patients undergoing coiling or stent-assisted coiling for a saccular IA at our institution were enrolled. Two samples (A and B) were collected from IA lumens, and the plasma level of 15-PGDH was measured using an ELISA-based method. The study included 38 patients, with 20 women and 18 men. Women and men were comparable on baseline characteristics. The mean plasma concentration of 15-PGDH did not differ statistically between sample A (62.8±16.2 ng/mL) and sample B (61.8±17.9 ng/mL; 95% confidence interval -6.6 to 9.4). The mean plasma concentration of 15-PGDH in IA lumens of samples A and B was significantly higher in men (73.8±13.5 ng/mL) than women (49.6±7.8 ng/mL; P<0.0001). CONCLUSIONS Higher enzyme levels of 15-PGDH exist in the lumen of IAs of men compared with women. This observation could explain why aspirin confers better protection against IA rupture in men than in women. The susceptibility of an individual to aspirin may differ according to the level of 15-PGDH.
Collapse
Affiliation(s)
- Nohra Chalouhi
- Department of Neurosurgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, PA
| | - Pascal Jabbour
- Department of Neurosurgery, Thomas Jefferson University and Jefferson Hospital for Neuroscience, Philadelphia, PA
| | - Mario Zanaty
- Department of Neurosurgery, University of Iowa, Iowa City, IA
| | | | - James Torner
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, IA
| | | | - David M Hasan
- Department of Neurosurgery, University of Iowa, Iowa City, IA
| |
Collapse
|
47
|
Correlation Between Aspirin Intake and Reduced Growth of Human Vestibular Schwannoma: Volumetric Analysis. Otol Neurotol 2017; 37:1428-34. [PMID: 27631829 DOI: 10.1097/mao.0000000000001180] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To determine whether people with sporadic vestibular schwannoma (VS) who take aspirin for unrelated medical reasons exhibit less tumor growth than nonaspirin users. We previously demonstrated the efficacy of salicylates in inhibiting VS growth in vitro, corroborating the results of our retrospective clinical study, which found halted VS growth (based on linear tumor measurements) in aspirin users. The current study evaluates this association using more accurate tumor volumetric measurements, and quantifies the degree of frequency-specific, VS-induced hearing loss. STUDY DESIGN Retrospective analysis. SETTING Tertiary care hospital. PATIENTS Diagnosed with VS between 1980 and 2012, followed by serial magnetic resonance imaging for at least 1 year. MAIN OUTCOME MEASURES Patient history of aspirin intake; change in VS volume over time of observation; frequency-specific, VS-induced audiometric threshold shifts. RESULTS Of the 347 patients followed by serial magnetic resonance imaging scans, 86 had sequential scans available for 3D-segmented volumetric analysis for up to 11 years of follow-up (median 53 mo). Twenty-five (29%) had documented history of aspirin intake; 8 (32%) of these demonstrated VS growth. Of the 61 (71%) nonusers, 36 (59%) demonstrated growth. A significant inverse association was found among aspirin users and VS growth: odds ratio 0.32, 95% confidence interval 0.11 to 0.91. VS-induced audiometric thresholds shifts were larger above than below 2000 Hz. CONCLUSION Our volumetric analysis of VS growth reaffirms the results of our linear analysis and suggests that aspirin may inhibit VS growth. The audiometric findings are consistent with the previously reported VS-induced predominantly high-frequency sensorineural hearing loss.
Collapse
|
48
|
Abstract
Cancer is a major health issue worldwide, and the global burden of cancer is expected to increase in the coming years. Whereas the limited success with current therapies has driven huge investments into drug development, the average number of FDA approvals per year has declined since the 1990s. This unmet need for more effective anti-cancer drugs has sparked a growing interest for drug repurposing, i.e. using drugs already approved for other indications to treat cancer. As such, data both from pre-clinical experiments, clinical trials and observational studies have demonstrated anti-tumor efficacy for compounds within a wide range of drug classes other than cancer. Whereas some of them induce cancer cell death or suppress various aspects of cancer cell behavior in established tumors, others may prevent cancer development. Here, we provide an overview of promising candidates for drug repurposing in cancer, as well as studies describing the biological mechanisms underlying their anti-neoplastic effects.
Collapse
Affiliation(s)
- Linda Sleire
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Hilde Elise Førde
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Inger Anne Netland
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Lina Leiss
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway
| | - Bente Sandvei Skeie
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway; Department of Neurosurgery, Haukeland University Hospital, Jonas Lies vei, 71, 5021 Bergen, Norway
| | - Per Øyvind Enger
- Oncomatrix Research Group, Department of Biomedicine, University of Bergen, Jonas Lies vei 91 5009 Bergen, Norway; Department of Neurosurgery, Haukeland University Hospital, Jonas Lies vei, 71, 5021 Bergen, Norway.
| |
Collapse
|
49
|
Westdorp H, Kolders S, Hoogerbrugge N, de Vries IJM, Jongmans MCJ, Schreibelt G. Immunotherapy holds the key to cancer treatment and prevention in constitutional mismatch repair deficiency (CMMRD) syndrome. Cancer Lett 2017. [PMID: 28645564 DOI: 10.1016/j.canlet.2017.06.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Monoallelic germline mutations in one of the DNA mismatch repair (MMR) genes cause Lynch syndrome, with a high lifetime risks of colorectal and endometrial cancer at adult age. Less well known, is the constitutional mismatch repair deficiency (CMMRD) syndrome caused by biallelic germline mutations in MMR genes. This syndrome is characterized by the development of childhood cancer. Patients with CMMRD are at extremely high risk of developing multiple cancers including hematological, brain and intestinal tumors. Mutations in MMR genes impair DNA repair and therefore most tumors of patients with CMMRD are hypermutated. These mutations lead to changes in the translational reading frame, which consequently result in neoantigen formation. Neoantigens are recognized as foreign by the immune system and can induce specific immune responses. The growing evidence on the clinical efficacy of immunotherapies, such as immune checkpoint inhibitors, offers the prospect for treatment of patients with CMMRD. Combining neoantigen-based vaccination strategies and immune checkpoint inhibitors could be an effective way to conquer CMMRD-related tumors. Neoantigen-based vaccines might also be a preventive treatment option in healthy biallelic MMR mutation carriers. Future studies need to reveal the safety and efficacy of immunotherapies for patients with CMMRD.
Collapse
Affiliation(s)
- Harm Westdorp
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sigrid Kolders
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Nicoline Hoogerbrugge
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marjolijn C J Jongmans
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Gerty Schreibelt
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
50
|
Vrijens K, Winckelmans E, Tsamou M, Baeyens W, De Boever P, Jennen D, de Kok TM, Den Hond E, Lefebvre W, Plusquin M, Reynders H, Schoeters G, Van Larebeke N, Vanpoucke C, Kleinjans J, Nawrot TS. Sex-Specific Associations between Particulate Matter Exposure and Gene Expression in Independent Discovery and Validation Cohorts of Middle-Aged Men and Women. ENVIRONMENTAL HEALTH PERSPECTIVES 2017; 125:660-669. [PMID: 27740511 PMCID: PMC5381989 DOI: 10.1289/ehp370] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 08/12/2016] [Accepted: 08/22/2016] [Indexed: 05/05/2023]
Abstract
BACKGROUND Particulate matter (PM) exposure leads to premature death, mainly due to respiratory and cardiovascular diseases. OBJECTIVES Identification of transcriptomic biomarkers of air pollution exposure and effect in a healthy adult population. METHODS Microarray analyses were performed in 98 healthy volunteers (48 men, 50 women). The expression of eight sex-specific candidate biomarker genes (significantly associated with PM10 in the discovery cohort and with a reported link to air pollution-related disease) was measured with qPCR in an independent validation cohort (75 men, 94 women). Pathway analysis was performed using Gene Set Enrichment Analysis. Average daily PM2.5 and PM10 exposures over 2-years were estimated for each participant's residential address using spatiotemporal interpolation in combination with a dispersion model. RESULTS Average long-term PM10 was 25.9 (± 5.4) and 23.7 (± 2.3) μg/m3 in the discovery and validation cohorts, respectively. In discovery analysis, associations between PM10 and the expression of individual genes differed by sex. In the validation cohort, long-term PM10 was associated with the expression of DNAJB5 and EAPP in men and ARHGAP4 (p = 0.053) in women. AKAP6 and LIMK1 were significantly associated with PM10 in women, although associations differed in direction between the discovery and validation cohorts. Expression of the eight candidate genes in the discovery cohort differentiated between validation cohort participants with high versus low PM10 exposure (area under the receiver operating curve = 0.92; 95% CI: 0.85, 1.00; p = 0.0002 in men, 0.86; 95% CI: 0.76, 0.96; p = 0.004 in women). CONCLUSIONS Expression of the sex-specific candidate genes identified in the discovery population predicted PM10 exposure in an independent cohort of adults from the same area. Confirmation in other populations may further support this as a new approach for exposure assessment, and may contribute to the discovery of molecular mechanisms for PM-induced health effects.
Collapse
Affiliation(s)
- Karen Vrijens
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Ellen Winckelmans
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Maria Tsamou
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Willy Baeyens
- Department of Analytical and Environmental Chemistry, Free University of Brussels, Brussels, Belgium
| | - Patrick De Boever
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Danyel Jennen
- Department of Toxicogenomics, Maastricht University, Maastricht, Netherlands
| | - Theo M. de Kok
- Department of Toxicogenomics, Maastricht University, Maastricht, Netherlands
| | - Elly Den Hond
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Mol, Belgium
- Provincial Institute for Hygiene, Antwerp, Belgium
| | - Wouter Lefebvre
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Mol, Belgium
| | - Michelle Plusquin
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
| | - Hans Reynders
- Environment, Nature and Energy Department, Flemish Government, Brussels, Belgium
| | - Greet Schoeters
- Environmental Risk and Health Unit, Flemish Institute for Technological Research (VITO), Mol, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
- University of Southern Denmark, Institute of Public Health, Department of Environmental Medicine, Odense, Denmark
| | - Nicolas Van Larebeke
- Department of Radiotherapy and Nuclear Medicine, Ghent University, Ghent, Belgium
| | | | - Jos Kleinjans
- Department of Toxicogenomics, Maastricht University, Maastricht, Netherlands
| | - Tim S. Nawrot
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Department of Public Health and Primary Care, Leuven University, Leuven, Belgium
- Address correspondence to T.S. Nawrot, Centre for Environmental Sciences, Hasselt University, Agoralaan gebouw D, B-3590 Diepenbeek, Belgium. Telephone: 0032/11-26.83.82. E-mail:
| |
Collapse
|