1
|
Xie R, Luo Y, Bao B, Wu X, Guo J, Wang J, Qu X, Che X, Zheng C. The Role of Fatty Acid Metabolism, the Related Potential Biomarkers, and Targeted Therapeutic Strategies in Gastrointestinal Cancers. Drug Dev Res 2024; 85:e70014. [PMID: 39527665 DOI: 10.1002/ddr.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/12/2024] [Accepted: 10/13/2024] [Indexed: 11/16/2024]
Abstract
Gastrointestinal cancer has emerged as a significant global health concern due to its high incidence and mortality, limited effectiveness of early detection, suboptimal treatment outcomes, and poor prognosis. Metabolic reprogramming is a prominent feature of cancer, and fatty acid metabolism assumes a pivotal role in bridging glucose metabolism and lipid metabolism. Fatty acids play important roles in cellular structural composition, energy supply, signal transduction, and other lipid-related processes. Changes in the levels of fatty acid metabolite may indicate the malignant transformation of gastrointestinal cells, which have an impact on the prognosis of patients and can be used as a marker to monitor the efficacy of anticancer therapy. Therefore, targeting key enzymes involved in fatty acid metabolism, either as monotherapy or in combination with other agents, is a promising strategy for anticancer treatment. This article reviews the potential mechanisms of fatty acid metabolism disorders in the occurrence and development of gastrointestinal tumors, and summarizes the related potential biomarkers and anticancer strategies.
Collapse
Affiliation(s)
- Ruixi Xie
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Ying Luo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Bowen Bao
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xinshu Wu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jia Guo
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Jin Wang
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiujuan Qu
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaofang Che
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chunlei Zheng
- Department of Medical Oncology, Provincial Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, Clinical Cancer Research Center of Shenyang, The First Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Oncology, Shanghai Electric Power Hospital, Shanghai, China
| |
Collapse
|
2
|
Xia Y, Sun M, Huang H, Jin WL. Drug repurposing for cancer therapy. Signal Transduct Target Ther 2024; 9:92. [PMID: 38637540 PMCID: PMC11026526 DOI: 10.1038/s41392-024-01808-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/05/2024] [Accepted: 03/19/2024] [Indexed: 04/20/2024] Open
Abstract
Cancer, a complex and multifactorial disease, presents a significant challenge to global health. Despite significant advances in surgical, radiotherapeutic and immunological approaches, which have improved cancer treatment outcomes, drug therapy continues to serve as a key therapeutic strategy. However, the clinical efficacy of drug therapy is often constrained by drug resistance and severe toxic side effects, and thus there remains a critical need to develop novel cancer therapeutics. One promising strategy that has received widespread attention in recent years is drug repurposing: the identification of new applications for existing, clinically approved drugs. Drug repurposing possesses several inherent advantages in the context of cancer treatment since repurposed drugs are typically cost-effective, proven to be safe, and can significantly expedite the drug development process due to their already established safety profiles. In light of this, the present review offers a comprehensive overview of the various methods employed in drug repurposing, specifically focusing on the repurposing of drugs to treat cancer. We describe the antitumor properties of candidate drugs, and discuss in detail how they target both the hallmarks of cancer in tumor cells and the surrounding tumor microenvironment. In addition, we examine the innovative strategy of integrating drug repurposing with nanotechnology to enhance topical drug delivery. We also emphasize the critical role that repurposed drugs can play when used as part of a combination therapy regimen. To conclude, we outline the challenges associated with repurposing drugs and consider the future prospects of these repurposed drugs transitioning into clinical application.
Collapse
Affiliation(s)
- Ying Xia
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
- Division of Gastroenterology and Hepatology, Department of Medicine and, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ming Sun
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China
| | - Hai Huang
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, PR China.
- School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550004, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, PR China.
| |
Collapse
|
3
|
Papadakos SP, Argyrou A, Lekakis V, Arvanitakis K, Kalisperati P, Stergiou IE, Konstantinidis I, Schizas D, Koufakis T, Germanidis G, Theocharis S. Metformin in Esophageal Carcinoma: Exploring Molecular Mechanisms and Therapeutic Insights. Int J Mol Sci 2024; 25:2978. [PMID: 38474224 PMCID: PMC10932447 DOI: 10.3390/ijms25052978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 02/29/2024] [Accepted: 03/03/2024] [Indexed: 03/14/2024] Open
Abstract
Esophageal cancer (EC) remains a formidable malignancy with limited treatment options and high mortality rates, necessitating the exploration of innovative therapeutic avenues. Through a systematic analysis of a multitude of studies, we synthesize the diverse findings related to metformin's influence on EC. This review comprehensively elucidates the intricate metabolic pathways and molecular mechanisms through which metformin may exert its anti-cancer effects. Key focus areas include its impact on insulin signaling, AMP-activated protein kinase (AMPK) activation, and the mTOR pathway, which collectively contribute to its role in mitigating esophageal cancer progression. This review critically examines the body of clinical and preclinical evidence surrounding the potential role of metformin, a widely prescribed anti-diabetic medication, in EC management. Our examination extends to the modulation of inflammation, oxidative stress and angiogenesis, revealing metformin's potential as a metabolic intervention in esophageal cancer pathogenesis. By consolidating epidemiological and clinical data, we assess the evidence that supports metformin's candidacy as an adjuvant therapy for esophageal cancer. By summarizing clinical and preclinical findings, our review aims to enhance our understanding of metformin's role in EC management, potentially improving patient care and outcomes.
Collapse
Affiliation(s)
- Stavros P. Papadakos
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 11527 Athens, Greece;
| | - Alexandra Argyrou
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece; (A.A.); (V.L.)
| | - Vasileios Lekakis
- Academic Department of Gastroenterology, Laikon General Hospital, Athens University Medical School, 11527 Athens, Greece; (A.A.); (V.L.)
| | - Konstantinos Arvanitakis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Polyxeni Kalisperati
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.K.); (I.E.S.)
| | - Ioanna E. Stergiou
- Pathophysiology Department, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.K.); (I.E.S.)
| | | | - Dimitrios Schizas
- First Department of Surgery, Laikon General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Theocharis Koufakis
- Second Propaedeutic Department of Internal Medicine, General Hospital “Hippokration”, Aristotle University of Thessaloniki, 54642 Thessaloniki, Greece;
| | - Georgios Germanidis
- First Department of Internal Medicine, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece;
- Basic and Translational Research Unit (BTRU), Special Unit for Biomedical Research and Education (BRESU), Faculty of Health Sciences, School of Medicine, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, Goudi, 11527 Athens, Greece;
| |
Collapse
|
4
|
Samaddar S, Buckles D, Saha S, Zhang Q, Bansal A. Translating Molecular Biology Discoveries to Develop Targeted Cancer Interception in Barrett's Esophagus. Int J Mol Sci 2023; 24:11318. [PMID: 37511077 PMCID: PMC10379200 DOI: 10.3390/ijms241411318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Esophageal adenocarcinoma (EAC) is a rapidly increasing lethal tumor. It commonly arises from a metaplastic segment known as Barrett's esophagus (BE), which delineates the at-risk population. Ample research has elucidated the pathogenesis of BE and its progression from metaplasia to invasive carcinoma; and multiple molecular pathways have been implicated in this process, presenting several points of cancer interception. Here, we explore the mechanisms of action of various agents, including proton pump inhibitors, non-steroidal anti-inflammatory drugs, metformin, and statins, and explain their roles in cancer interception. Data from the recent AspECT trial are discussed to determine how viable a multipronged approach to cancer chemoprevention would be. Further, novel concepts, such as the repurposing of chemotherapeutic drugs like dasatinib and the prevention of post-ablation BE recurrence using itraconazole, are discussed.
Collapse
Affiliation(s)
- Sohini Samaddar
- Department of Internal Medicine, University of Kansas Health System, Kansas City, KS 66160, USA
| | - Daniel Buckles
- Department of Gastroenterology and Hepatology, University of Kansas Health System, Kansas City, KS 66160, USA
| | - Souvik Saha
- Department of Internal Medicine, University of Kansas Health System, Kansas City, KS 66160, USA
| | - Qiuyang Zhang
- Center for Esophageal Diseases, Department of Medicine, Baylor University Medical Center, Dallas, TX 75246, USA
- Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, TX 75246, USA
| | - Ajay Bansal
- Department of Gastroenterology and Hepatology, University of Kansas Health System, Kansas City, KS 66160, USA
- University of Kansas Cancer Center, Kansas City, KS 66160, USA
| |
Collapse
|
5
|
Hua Y, Zheng Y, Yao Y, Jia R, Ge S, Zhuang A. Metformin and cancer hallmarks: shedding new lights on therapeutic repurposing. J Transl Med 2023; 21:403. [PMID: 37344841 DOI: 10.1186/s12967-023-04263-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 06/09/2023] [Indexed: 06/23/2023] Open
Abstract
Metformin is a well-known anti-diabetic drug that has been repurposed for several emerging applications, including as an anti-cancer agent. It boasts the distinct advantages of an excellent safety and tolerability profile and high cost-effectiveness at less than one US dollar per daily dose. Epidemiological evidence reveals that metformin reduces the risk of cancer and decreases cancer-related mortality in patients with diabetes; however, the exact mechanisms are not well understood. Energy metabolism may be central to the mechanism of action. Based on altering whole-body energy metabolism or cellular state, metformin's modes of action can be divided into two broad, non-mutually exclusive categories: "direct effects", which induce a direct effect on cancer cells, independent of blood glucose and insulin levels, and "indirect effects" that arise from systemic metabolic changes depending on blood glucose and insulin levels. In this review, we summarize an updated account of the current knowledge on metformin antitumor action, elaborate on the underlying mechanisms in terms of the hallmarks of cancer, and propose potential applications for repurposing metformin for cancer therapeutics.
Collapse
Affiliation(s)
- Yu Hua
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No. 639 Zhizaoju Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, No. 639 Zhizaoju Road, Shanghai, 200011, China.
| |
Collapse
|
6
|
Shah MA, Altorki N, Patel P, Harrison S, Bass A, Abrams JA. Improving outcomes in patients with oesophageal cancer. Nat Rev Clin Oncol 2023; 20:390-407. [PMID: 37085570 DOI: 10.1038/s41571-023-00757-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2023] [Indexed: 04/23/2023]
Abstract
The care of patients with oesophageal cancer or of individuals who have an elevated risk of oesophageal cancer has changed dramatically. The epidemiology of squamous cell and adenocarcinoma of the oesophagus has diverged over the past several decades, with a marked increase in incidence only for oesophageal adenocarcinoma. Only in the past decade, however, have molecular features that distinguish these two forms of the disease been identified. This advance has the potential to improve screening for oesophageal cancers through the development of novel minimally invasive diagnostic technologies predicated on cancer-specific genomic or epigenetic alterations. Surgical techniques have also evolved towards less invasive approaches associated with less morbidity, without compromising oncological outcomes. With improvements in multidisciplinary care, advances in radiotherapy and new tools to detect minimal residual disease, certain patients may no longer even require surgical tumour resection. However, perhaps the most anticipated advance in the treatment of patients with oesophageal cancer is the advent of immune-checkpoint inhibitors, which harness and enhance the host immune response against cancer. In this Review, we discuss all these advances in the management of oesophageal cancer, representing only the beginning of a transformation in our quest to improve patient outcomes.
Collapse
Affiliation(s)
- Manish A Shah
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| | - Nasser Altorki
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Pretish Patel
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebron Harrison
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Adam Bass
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
7
|
Dai J, Ports KD, Corrada MM, Odegaard AO, O’Connell J, Jiang L. Metformin and Dementia Risk: A Systematic Review with Respect to Time Related Biases. J Alzheimers Dis Rep 2022; 6:443-459. [PMID: 36186728 PMCID: PMC9484147 DOI: 10.3233/adr-220002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND When studying drug effects using observational data, time-related biases may exist and result in spurious associations. Numerous observational studies have investigated metformin and dementia risk, but have reported inconsistent findings, some of which might be caused by unaddressed time-related biases. Immortal time bias biases the results toward a "protective" effect, whereas time-lag and time-window biases can lead to either a "detrimental" or "protective" effect. OBJECTIVE To conduct a systematic review examining time-related biases in the literature on metformin and dementia. METHODS The electronic databases PubMed, Web of Science, and ProQuest were searched for the terms "Metformin" AND ("dementia" OR "Alzheimer's Disease" OR "cognitive impairment"). These databases were searched from inception through 09/24/2021. Only English language articles and human research were eligible. RESULTS Seventeen studies were identified: thirteen cohort studies, two case-control studies, and two nested case-control studies. Eleven (64.7%) studies reported a reduced risk of dementia associated with metformin use; two (11.8%) suggested metformin increased dementia risk, while four (23.5%) concluded no significant associations. Eight (61.5%) of thirteen cohort studies had immortal time bias or did not clearly address it. Fifteen (88.2%) of seventeen reviewed studies had time-lag bias or did not clearly address it. Two (50.0%) of four case-control studies did not explicitly address time-window bias. The studies that addressed most biases concluded no associations between metformin and dementia risk. CONCLUSION None of the reviewed studies clearly addressed relevant time-related biases, illustrating time-related biases are common in observational studies investigating the impact of anti-diabetic medications on dementia risk.
Collapse
Affiliation(s)
- Jiahui Dai
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Kayleen Deanna Ports
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Maria M. Corrada
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
- Department of Neurology, University of California Irvine, Irvine, CA, USA
- Institute for Memory Impairments and Neurological Disorders (UCI MIND), University of California Irvine, Irvine, CA, USA
| | - Andrew O. Odegaard
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| | - Joan O’Connell
- Centers for American Indian and Alaska Native Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Luohua Jiang
- Department of Epidemiology & Biostatistics, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
8
|
Triggle CR, Mohammed I, Bshesh K, Marei I, Ye K, Ding H, MacDonald R, Hollenberg MD, Hill MA. Metformin: Is it a drug for all reasons and diseases? Metabolism 2022; 133:155223. [PMID: 35640743 DOI: 10.1016/j.metabol.2022.155223] [Citation(s) in RCA: 139] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/22/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Metformin was first used to treat type 2 diabetes in the late 1950s and in 2022 remains the first-choice drug used daily by approximately 150 million people. An accumulation of positive pre-clinical and clinical data has stimulated interest in re-purposing metformin to treat a variety of diseases including COVID-19. In polycystic ovary syndrome metformin improves insulin sensitivity. In type 1 diabetes metformin may help reduce the insulin dose. Meta-analysis and data from pre-clinical and clinical studies link metformin to a reduction in the incidence of cancer. Clinical trials, including MILES (Metformin In Longevity Study), and TAME (Targeting Aging with Metformin), have been designed to determine if metformin can offset aging and extend lifespan. Pre-clinical and clinical data suggest that metformin, via suppression of pro-inflammatory pathways, protection of mitochondria and vascular function, and direct actions on neuronal stem cells, may protect against neurodegenerative diseases. Metformin has also been studied for its anti-bacterial, -viral, -malaria efficacy. Collectively, these data raise the question: Is metformin a drug for all diseases? It remains unclear as to whether all of these putative beneficial effects are secondary to its actions as an anti-hyperglycemic and insulin-sensitizing drug, or result from other cellular actions, including inhibition of mTOR (mammalian target for rapamycin), or direct anti-viral actions. Clarification is also sought as to whether data from ex vivo studies based on the use of high concentrations of metformin can be translated into clinical benefits, or whether they reflect a 'Paracelsus' effect. The environmental impact of metformin, a drug with no known metabolites, is another emerging issue that has been linked to endocrine disruption in fish, and extensive use in T2D has also raised concerns over effects on human reproduction. The objectives for this review are to: 1) evaluate the putative mechanism(s) of action of metformin; 2) analyze the controversial evidence for metformin's effectiveness in the treatment of diseases other than type 2 diabetes; 3) assess the reproducibility of the data, and finally 4) reach an informed conclusion as to whether metformin is a drug for all diseases and reasons. We conclude that the primary clinical benefits of metformin result from its insulin-sensitizing and antihyperglycaemic effects that secondarily contribute to a reduced risk of a number of diseases and thereby enhancing healthspan. However, benefits like improving vascular endothelial function that are independent of effects on glucose homeostasis add to metformin's therapeutic actions.
Collapse
Affiliation(s)
- Chris R Triggle
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar.
| | - Ibrahim Mohammed
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Khalifa Bshesh
- Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Isra Marei
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Kevin Ye
- Department of Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Hong Ding
- Department of Pharmacology, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar; Department of Medical Education, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Ross MacDonald
- Distribution eLibrary, Weill Cornell Medicine in Qatar, P.O. Box 24144, Education City, Doha, Qatar
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology, a Cumming School of Medicine, University of Calgary, T2N 4N1, Canada
| | - Michael A Hill
- Dalton Cardiovascular Research Center, Department of Medical Pharmacology & Physiology, School of Medicine, University of Missouri, Columbia 65211, MO, USA
| |
Collapse
|
9
|
Alkhayyat M, Kumar P, Sanaka KO, Thota PN. Chemoprevention in Barrett's esophagus and esophageal adenocarcinoma. Therap Adv Gastroenterol 2021; 14:17562848211033730. [PMID: 34434254 PMCID: PMC8381453 DOI: 10.1177/17562848211033730] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 06/29/2021] [Indexed: 02/04/2023] Open
Abstract
There has been a dramatic increase in the incidence of Barrett's esophagus and esophageal adenocarcinoma over the past several decades with a continued rise expected in the future. Several strategies have been developed for screening and surveillance of patients with Barrett's esophagus and endoscopic treatment of Barrett's associated dysplasia and early esophageal cancer; however, they have not made a substantial impact on the incidence of cancer. Herein, chemoprevention becomes an attractive idea for reducing the incidence of cancer in Barrett's patients. Several agents appear promising in preclinical and observational studies but very few have been evaluated in randomized controlled trials. Strongest evidence to date is available for proton-pump inhibitors and Aspirin that have been evaluated in a large randomized controlled trial. Other agents such as statins, metformin, ursodeoxycholic acid, and dietary supplements have insufficient evidence for chemoprevention in Barrett's patients.
Collapse
Affiliation(s)
- Motasem Alkhayyat
- Department of Internal Medicine, Cleveland
Clinic, Cleveland, OH, USA
| | - Prabhat Kumar
- Department of Gastroenterology and Hepatology,
Cleveland Clinic, Cleveland, OH, USA
| | - Krishna O. Sanaka
- Department of Gastroenterology and Hepatology,
Cleveland Clinic, Cleveland, OH, USA
| | | |
Collapse
|
10
|
Chaeichi-Tehrani N, Ferns GA, Hassanian SM, Khazaei M, Avan A. The Therapeutic Potential of Targeting Autophagy in The Treatment of Cancer. Curr Cancer Drug Targets 2021; 21:725-736. [PMID: 34077348 DOI: 10.2174/1568009621666210601113144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 03/02/2021] [Accepted: 03/12/2021] [Indexed: 11/22/2022]
Abstract
Autophagy is a mechanism by which unwanted cellular components are degraded through a pathway that involves the lysosomes and contributes to several pathological conditions such as cancer. Gastrointestinal cancers affect the digestive organs from the esophagus to the anus and are among the most commonly diagnosed cancers globally. The modulation of autophagy using pharmacologic agents potentially offers a great potential for cancer therapy. In this review, some commonly used compounds, together with their molecular target and the mechanism through which they stimulate or block the autophagy pathway as well as their therapeutic benefit in treating patients with gastrointestinal cancers, are summarized.
Collapse
Affiliation(s)
- Negin Chaeichi-Tehrani
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex BN1 9PH, United Kingdom
| | - Seyed Mahdi Hassanian
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic syndrome Research centre, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Marabotto E, Pellegatta G, Sheijani AD, Ziola S, Zentilin P, De Marzo MG, Giannini EG, Ghisa M, Barberio B, Scarpa M, Angriman I, Fassan M, Savarino V, Savarino E. Prevention Strategies for Esophageal Cancer-An Expert Review. Cancers (Basel) 2021; 13:2183. [PMID: 34062788 PMCID: PMC8125297 DOI: 10.3390/cancers13092183] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
In the last 30 years, we have witnessed a rapid increase in the incidence and prevalence of esophageal cancer in many countries around the word. However, despite advancements in diagnostic technologies, the early detection of this cancer is rare, and its prognosis remains poor, with only about 20% of these patients surviving for 5 years. The two major forms are the esophageal squamous cell carcinoma (ESCC), which is particularly frequent in the so-called Asian belt, and the esophageal adenocarcinoma (EAC), which prevails in Western populations. This review provides a summary of the epidemiological features and risk factors associated with these tumors. Moreover, a major focus is posed on reporting and highlighting the various preventing strategies proposed by the most important international scientific societies, particularly in high-risk populations, with the final aim of detecting these lesions as early as possible and therefore favoring their definite cure. Indeed, we have conducted analysis with attention to the current primary, secondary and tertiary prevention guidelines in both ESCC and EAC, attempting to emphasize unresolved research and clinical problems related to these topics in order to improve our diagnostic strategies and management.
Collapse
Affiliation(s)
- Elisa Marabotto
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Gaia Pellegatta
- Digestive Endoscopy Unit, Department of Gastroenterology, IRCCS Humanitas Research Hospital, via Manzoni 56, 20089 Milan, Italy;
| | - Afscin Djahandideh Sheijani
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Sebastiano Ziola
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Patrizia Zentilin
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Maria Giulia De Marzo
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Edoardo Giovanni Giannini
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Matteo Ghisa
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| | - Brigida Barberio
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| | - Marco Scarpa
- Clinica Chirurgica 1, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.S.); (I.A.)
| | - Imerio Angriman
- Clinica Chirurgica 1, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.S.); (I.A.)
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padua, 35121 Padua, Italy;
| | - Vincenzo Savarino
- Gastroenterology Unit, Department of Internal Medicine, University of Genoa, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (A.D.S.); (S.Z.); (P.Z.); (M.G.D.M.); (E.G.G.); (V.S.)
| | - Edoardo Savarino
- Gastroenterology Unit, Department of Surgery, Oncology and Gastroenterology, University of Padova, 35128 Padova, Italy; (M.G.); (B.B.)
| |
Collapse
|
12
|
Abrams JA, Del Portillo A, Hills C, Compres G, Friedman RA, Cheng B, Poneros J, Lightdale CJ, De La Rue R, di Pietro M, Fitzgerald RC, Sepulveda A, Wang TC. Randomized Controlled Trial of the Gastrin/CCK 2 Receptor Antagonist Netazepide in Patients with Barrett's Esophagus. Cancer Prev Res (Phila) 2021; 14:675-682. [PMID: 33782049 DOI: 10.1158/1940-6207.capr-21-0050] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 11/16/2022]
Abstract
Hypergastrinemia has been associated with high-grade dysplasia and adenocarcinoma in patients with Barrett's esophagus, and experimental studies suggest proinflammatory and proneoplastic effects of gastrin on Barrett's esophagus. This is of potential concern, as patients with Barrett's esophagus are treated with medications that suppress gastric acid production, resulting in increased physiologic levels of gastrin. We aimed to determine whether treatment with the novel gastrin/CCK2 receptor antagonist netazepide reduces expression of markers associated with inflammation and neoplasia in Barrett's esophagus. This was a randomized, double-blind, placebo-controlled trial of netazepide in patients with Barrett's esophagus without dysplasia. Subjects were treated for 12 weeks, with endoscopic assessment at baseline and at end of treatment. The primary outcome was within-individual change in cellular proliferation as assessed by Ki67. Secondary analyses included changes in gene expression, assessed by RNA-sequencing, and safety and tolerability. A total of 20 subjects completed the study and were included in the analyses. There was no difference between arms in mean change in cellular proliferation (netazepide: +35.6 Ki67+ cells/mm2, SD 620.7; placebo: +307.8 Ki67+ cells/mm2, SD 640.3; P = 0.35). Netazepide treatment resulted in increased expression of genes related to gastric phenotype (TFF2, MUC5B) and certain cancer-associated markers (REG3A, PAX9, MUC1), and decreased expression of intestinal markers MUC2, FABP1, FABP2, and CDX1 No serious adverse events related to study drug occurred. The gastrin/CCK2 receptor antagonist netazepide did not reduce cellular proliferation in patients with nondysplastic Barrett's esophagus. Further research should focus on the biological effects of gastrin in Barrett's esophagus.Prevention Relevance: Treatment of patients with Barrett's esophagus with a gastrin/CCK2 receptor antagonist did not have obvious chemopreventive effects.
Collapse
Affiliation(s)
- Julian A Abrams
- Department of Medicine, Columbia University Irving Medical Center, New York, New York. .,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Armando Del Portillo
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Caitlin Hills
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Griselda Compres
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Richard A Friedman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York.,Department of Biomedical Informatics, Columbia University Irving Medical Center, New York, New York
| | - Bin Cheng
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, New York
| | - John Poneros
- Department of Medicine, Columbia University Irving Medical Center, New York, New York
| | - Charles J Lightdale
- Department of Medicine, Columbia University Irving Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| | - Rachel De La Rue
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Massimiliano di Pietro
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Rebecca C Fitzgerald
- MRC Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Antonia Sepulveda
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York.,Department of Pathology, George Washington University, Washington, D.C
| | - Timothy C Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, New York.,Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
13
|
Elliott JA, Reynolds JV. Visceral Obesity, Metabolic Syndrome, and Esophageal Adenocarcinoma. Front Oncol 2021; 11:627270. [PMID: 33777773 PMCID: PMC7994523 DOI: 10.3389/fonc.2021.627270] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/19/2021] [Indexed: 12/16/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) represents an exemplar of obesity-associated carcinogenesis, with a progressive increase in EAC risk with increased body mass index. In this context, there is increased focus on visceral adipose tissue and associated metabolic dysfunction, including hypertension, diabetes mellitus and hyperlipidemia, or combinations of these in the metabolic syndrome. Visceral obesity (VO) may promote EAC via both directly impacting on gastro-esophageal reflux disease and Barrett's esophagus, as well as via reflux-independent effects, involving adipokines, growth factors, insulin resistance, and the microbiome. In this review these pathways are explored, including the impact of VO on the tumor microenvironment, and on cancer outcomes. The current evidence-based literature regarding the role of dietary, lifestyle, pharmacologic and surgical interventions to modulate the risk of EAC is explored.
Collapse
Affiliation(s)
- Jessie A Elliott
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| | - John V Reynolds
- Trinity St. James's Cancer Institute, Trinity College Dublin and St. James's Hospital, Dublin, Ireland
| |
Collapse
|
14
|
Moayyedi P, El-Serag HB. Current Status of Chemoprevention in Barrett's Esophagus. Gastrointest Endosc Clin N Am 2021; 31:117-130. [PMID: 33213791 DOI: 10.1016/j.giec.2020.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Candidates for chemoprevention in Barrett's esophagus have long been suggested and there has been observational data to support many drugs, including statins, hormone replacement therapy, metformin, proton pump inhibitor therapy, and aspirin. Proton pump inhibitor therapy and aspirin are the most promising agents. Data suggest that aspirin and proton pump inhibitor therapy can decrease the risk of neoplastic progression in Barrett's esophagus. Further, the combination of aspirin and proton pump inhibitor therapy decrease all-cause mortality by approximately 33%. Future guideline groups need to evaluate the evidence rigorously, but the combination of proton pump inhibitor therapy and aspirin is promising.
Collapse
Affiliation(s)
- Paul Moayyedi
- McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada.
| | - Hashem B El-Serag
- Baylor College of Medicine Medical Center, McNair Campus (Clinic), 7200 Cambridge Street, 8th Floor, Suite 8B, Houston, TX 77030, USA
| |
Collapse
|
15
|
Snider EJ, Kaz AM, Inadomi JM, Grady WM. Chemoprevention of esophageal adenocarcinoma. Gastroenterol Rep (Oxf) 2020; 8:253-260. [PMID: 32843972 PMCID: PMC7434588 DOI: 10.1093/gastro/goaa040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022] Open
Abstract
Esophageal adenocarcinoma (EAC) is a major cause of cancer-related death, particularly in Western populations, and is rapidly rising in Asian populations at this time. Virtually all EACs develop from the precursor lesion Barrett's esophagus (BE), which is the most significant risk factor for EAC. However, the rates of progression from BE to EAC are low and patients with BE are asymptomatic. Thus, any strategy for EAC prevention must carry a low risk of harm in order to be clinically useful. Since current EAC-screening and BE-surveillance methods carry some procedural risk and are burdensome, there is an opportunity for chemoprevention, i.e. medications or dietary factors that may prevent BE from progressing to EAC. A variety of candidate chemoprevention therapies have been assessed to date. Proton-pump inhibitors (PPIs) are the best studied and have modest EAC-chemoprevention efficacy in BE patients, with a recent randomized trial showing that high-dose PPI may be more effective than low-dose PPI. Aspirin and other non-steroidal anti-inflammatory drugs have moderate quality observational and randomized-trial evidence for preventing progression of BE to EAC, but their risks for harm have precluded their routine clinical use. Other therapies (statins, metformin, female sex hormones) generally do not have strong evidence to support their use in EAC chemoprevention. Although progress has been made in this field, there is still a need for more effective and safe chemoprevention therapies for EAC.
Collapse
Affiliation(s)
- Erik J Snider
- Division of Gastroenterology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Andrew M Kaz
- Division of Gastroenterology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Gastroenterology Section, VA Puget Sound Health Care System, Seattle, WA, USA
| | - John M Inadomi
- Division of Gastroenterology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - William M Grady
- Division of Gastroenterology, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
16
|
Xie J, Ye J, Cai Z, Luo Y, Zhu X, Deng Y, Feng Y, Liang Y, Liu R, Han Z, Liang Y, Zheng Y, Mo R, Zhuo Y, Wu Y, Jiang F, Zhu J, Wu CL, Zhong W. GPD1 Enhances the Anticancer Effects of Metformin by Synergistically Increasing Total Cellular Glycerol-3-Phosphate. Cancer Res 2020; 80:2150-2162. [PMID: 32179514 DOI: 10.1158/0008-5472.can-19-2852] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/06/2020] [Accepted: 03/11/2020] [Indexed: 11/16/2022]
Abstract
Metformin is an oral drug widely used for the treatment of type 2 diabetes mellitus. Numerous studies have demonstrated the value of metformin in cancer treatment. However, for metformin to elicit effects on cancer often requires a high dosage, and any underlying mechanism for how to improve its inhibitory effects remains unknown. Here, we found that low mRNA expression of glycerol-3-phosphate dehydrogenase 1 (GPD1) may predict a poor response to metformin treatment in 15 cancer cell lines. In vitro and in vivo, metformin treatment alone significantly suppressed cancer cell proliferation, a phenotype enhanced by GPD1 overexpression. Total cellular glycerol-3-phosphate concentration was significantly increased by the combination of GPD1 overexpression and metformin treatment, which suppressed cancer growth via inhibition of mitochondrial function. Eventually, increased reactive oxygen species and mitochondrial structural damage was observed in GPD1-overexpressing cell lines treated with metformin, which may contribute to cell death. In summary, this study demonstrates that GPD1 overexpression enhances the anticancer activity of metformin and that patients with increased GPD1 expression in tumor cells may respond better to metformin therapy. SIGNIFICANCE: GPD1 overexpression enhances the anticancer effect of metformin through synergistic inhibition of mitochondrial function, thereby providing new insight into metformin-mediated cancer therapy.
Collapse
Affiliation(s)
- Jianjiang Xie
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianheng Ye
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Zhiduan Cai
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Luo
- Department of Urology, the Second People's Hospital of Foshan, Affiliated Foshan Hospital of Southern Medical University, Foshan, China
| | - Xuejin Zhu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yulin Deng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuanfa Feng
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yingke Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ren Liu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Zhaodong Han
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yuxiang Liang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yu Zheng
- Department of Urology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Rujun Mo
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yangjia Zhuo
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yongding Wu
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Funeng Jiang
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jianguo Zhu
- Department of Urology, Guizhou Provincial People's Hospital, the Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou Province, China
| | - Chin-Lee Wu
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
- School of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Weide Zhong
- Department of Urology, Guangdong Key Laboratory of Clinical Molecular Medicine and Diagnostics, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Urology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Guangdong Provincial Institute of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Urology Key Laboratory of Guangdong Province, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
17
|
Zell JA, McLaren CE, Morgan TR, Lawson MJ, Rezk S, Albers CG, Chen WP, Carmichael JC, Chung J, Richmond E, Rodriguez LM, Szabo E, Ford LG, Pollak MN, Meyskens FL. A Phase IIa Trial of Metformin for Colorectal Cancer Risk Reduction among Individuals with History of Colorectal Adenomas and Elevated Body Mass Index. Cancer Prev Res (Phila) 2019; 13:203-212. [PMID: 31818851 DOI: 10.1158/1940-6207.capr-18-0262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 07/09/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
Abstract
Obesity is associated with risk of colorectal adenoma (CRA) and colorectal cancer. The signaling pathway activated by metformin (LKB1/AMPK/mTOR) is implicated in tumor suppression in ApcMin/+ mice via metformin-induced reduction in polyp burden, increased ratio of pAMPK/AMPK, decreased pmTOR/mTOR ratio, and decreased pS6Ser235/S6Ser235 ratio in polyps. We hypothesized that metformin would affect colorectal tissue S6Ser235 among obese patients with recent history of CRA. A phase IIa clinical biomarker trial was conducted via the U.S. National Cancer Institute-Chemoprevention Consortium. Nondiabetic, obese subjects (BMI ≥30) ages 35 to 80 with recent history of CRA were included. Subjects received 12 weeks of oral metformin 1,000 mg twice every day. Rectal mucosa biopsies were obtained at baseline and end-of-treatment (EOT) endoscopy. Tissue S6Ser235 and Ki-67 immunostaining were analyzed in a blinded fashion using Histo score (Hscore) analysis. Among 32 eligible subjects, the mean baseline BMI was 34.9. Comparing EOT to baseline tissue S6Ser235 by IHC, no significant differences were observed. Mean (SD) Hscore at baseline was 1.1 (0.57) and 1.1 (0.51) at EOT; median Hscore change was 0.034 (P = 0.77). Similarly, Ki-67 levels were unaffected by the intervention. The adverse events were consistent with metformin's known side-effect profile. Among obese patients with CRA, 12 weeks of oral metformin does not reduce rectal mucosa pS6 or Ki-67 levels. Further research is needed to determine what effects metformin has on the target tissue of origin as metformin continues to be pursued as a colorectal cancer chemopreventive agent.
Collapse
Affiliation(s)
- Jason A Zell
- Department of Medicine, University of California, Irvine, California. .,Department of Epidemiology, University of California, Irvine, California.,Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | - Christine E McLaren
- Department of Epidemiology, University of California, Irvine, California.,Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | - Timothy R Morgan
- Medical Service, VA Long Beach Healthcare System, Long Beach, California
| | - Michael J Lawson
- Division of Gastroenterology, Kaiser Permanente Sacramento Medical Center, Sacramento, California
| | - Sherif Rezk
- Department of Pathology, University of California, Irvine, California
| | - C Gregory Albers
- Department of Medicine, University of California, Irvine, California
| | - Wen-Pin Chen
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | | | - Jinah Chung
- Chao Family Comprehensive Cancer Center, University of California, Irvine, California
| | - Ellen Richmond
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - L M Rodriguez
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland.,Department of Surgery, Walter Reed National Military Medical Center, Bethesda, Maryland
| | - Eva Szabo
- Department of Oncology, McGill University, Montreal, Canada
| | - Leslie G Ford
- Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | | | - Frank L Meyskens
- Department of Medicine, University of California, Irvine, California.,Chao Family Comprehensive Cancer Center, University of California, Irvine, California.,Department of Biological Chemistry, University of California, Irvine, California
| |
Collapse
|
18
|
Singh T, Sanghi V, Thota PN. Current management of Barrett esophagus and esophageal adenocarcinoma. Cleve Clin J Med 2019; 86:724-732. [PMID: 31710585 DOI: 10.3949/ccjm.86a.18106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Barrett esophagus is found in 5% to 15% of patients with gastroesophageal reflux disease and is a precursor of esophageal adenocarcinoma, yet the condition often goes undiagnosed. Patients with reflux disease who are male, over age 50, or white, and who smoke or have central obesity or a family history of Barrett esophagus or esophageal adenocarcinoma, should undergo initial screening endoscopy and, if no dysplasia is noted, surveillance endoscopy every 3 to 5 years. Dysplasia is treated with endoscopic eradication by ablation, resection, or both. Chemoprotective agents are being studied to prevent progression to dysplasia in Barrett esophagus. The authors discuss current recommendations for screening and management.
Collapse
Affiliation(s)
- Tavankit Singh
- Department of Gastroenterology and Hepatology, Cleveland Clinic
| | - Vedha Sanghi
- Department of Internal Medicine, Cleveland Clinic; Clinical Instructor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| | - Prashanthi N Thota
- Medical Director, Esophageal Center, Digestive Disease and Surgery Institute, Cleveland Clinic; Clinical Assistant Professor, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH
| |
Collapse
|
19
|
Vázquez-Borrego MC, Fuentes-Fayos AC, Herrera-Martínez AD, L-López F, Ibáñez-Costa A, Moreno-Moreno P, Alhambra-Expósito MR, Barrera-Martín A, Blanco-Acevedo C, Dios E, Venegas-Moreno E, Solivera J, Gahete MD, Soto-Moreno A, Gálvez-Moreno MA, Castaño JP, Luque RM. Biguanides Exert Antitumoral Actions in Pituitary Tumor Cells Through AMPK-Dependent and -Independent Mechanisms. J Clin Endocrinol Metab 2019; 104:3501-3513. [PMID: 30860580 DOI: 10.1210/jc.2019-00056] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 03/06/2019] [Indexed: 12/17/2022]
Abstract
CONTEXT Pituitary neuroendocrine tumors (PitNETs) are a commonly underestimated pathology in terms of incidence and associated morbimortality. Currently, an appreciable subset of patients are resistant or poorly responsive to the main current medical treatments [i.e., synthetic somatostatin analogs (SSAs) and dopamine agonists]. Thus, development and optimization of novel and available medical therapies is necessary. Biguanides (metformin, buformin, and phenformin) are antidiabetic drugs that exert antitumoral actions in several tumor types, but their pharmacological effects on PitNETs are poorly known. OBJECTIVE We aimed to explore the direct effects of biguanides on key functions (cell viability, hormone release, apoptosis, and signaling pathways) in primary cell cultures from human PitNETs and cell lines. Additionally, we evaluated the effect of combined metformin with SSAs on cell viability and hormone secretion. DESIGN A total of 13 corticotropinomas, 13 somatotropinomas, 13 nonfunctioning PitNETs, 3 prolactinomas, and 2 tumoral pituitary cell lines (AtT-20 and GH3) were used to evaluate the direct effects of biguanides on cell viability, hormone release, apoptosis, and signaling pathways. RESULTS Biguanides reduced cell viability in all PitNETs and cell lines (with phenformin being the most effective biguanide) and increased apoptosis in somatotropinomas. Moreover, buformin and phenformin, but not metformin, reduced hormone secretion in a cell type-specific manner. Combination metformin/SSA therapy did not increase SSA monotherapy effectiveness. Effects of biguanides on PitNETs could involve the modulation of AMP-activated protein kinase-dependent ([Ca2+]i, PI3K/Akt) and independent (MAPK) mechanisms. CONCLUSION Altogether, our data unveil clear antitumoral effects of biguanides on PitNET cells, opening avenues to explore their potential as drugs to treat these pathologies.
Collapse
Affiliation(s)
- Mari C Vázquez-Borrego
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Antonio C Fuentes-Fayos
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Aura D Herrera-Martínez
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Fernando L-López
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alejandro Ibáñez-Costa
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Paloma Moreno-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - María R Alhambra-Expósito
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Ana Barrera-Martín
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Cristóbal Blanco-Acevedo
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Elena Dios
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Eva Venegas-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Juan Solivera
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Neurosurgery, Reina Sofia University Hospital, Cordoba, Spain
| | - Manuel D Gahete
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Alfonso Soto-Moreno
- Metabolism and Nutrition Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - María A Gálvez-Moreno
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- Service of Endocrinology and Nutrition, IMIBIC, Reina Sofia University Hospital, Cordoba, Spain
| | - Justo P Castaño
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| | - Raúl M Luque
- Maimonides Institute of Biomedical Research of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Reina Sofia University Hospital, Cordoba, Spain
- CIBER Physiopathology of Obesity and Nutrition, Cordoba, Spain
| |
Collapse
|
20
|
Kurelac I, Iommarini L, Vatrinet R, Amato LB, De Luise M, Leone G, Girolimetti G, Umesh Ganesh N, Bridgeman VL, Ombrato L, Columbaro M, Ragazzi M, Gibellini L, Sollazzo M, Feichtinger RG, Vidali S, Baldassarre M, Foriel S, Vidone M, Cossarizza A, Grifoni D, Kofler B, Malanchi I, Porcelli AM, Gasparre G. Inducing cancer indolence by targeting mitochondrial Complex I is potentiated by blocking macrophage-mediated adaptive responses. Nat Commun 2019; 10:903. [PMID: 30796225 PMCID: PMC6385215 DOI: 10.1038/s41467-019-08839-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/30/2019] [Indexed: 02/08/2023] Open
Abstract
Converting carcinomas in benign oncocytomas has been suggested as a potential anti-cancer strategy. One of the oncocytoma hallmarks is the lack of respiratory complex I (CI). Here we use genetic ablation of this enzyme to induce indolence in two cancer types, and show this is reversed by allowing the stabilization of Hypoxia Inducible Factor-1 alpha (HIF-1α). We further show that on the long run CI-deficient tumors re-adapt to their inability to respond to hypoxia, concordantly with the persistence of human oncocytomas. We demonstrate that CI-deficient tumors survive and carry out angiogenesis, despite their inability to stabilize HIF-1α. Such adaptive response is mediated by tumor associated macrophages, whose blockage improves the effect of CI ablation. Additionally, the simultaneous pharmacological inhibition of CI function through metformin and macrophage infiltration through PLX-3397 impairs tumor growth in vivo in a synergistic manner, setting the basis for an efficient combinatorial adjuvant therapy in clinical trials.
Collapse
Affiliation(s)
- Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Luisa Iommarini
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Renaud Vatrinet
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Laura Benedetta Amato
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Monica De Luise
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Giulia Leone
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Giulia Girolimetti
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Nikkitha Umesh Ganesh
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | | | - Luigi Ombrato
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Marta Columbaro
- Laboratory of Musculoskeletal Cell Biology, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli 1, 40136, Bologna, Italy
| | - Moira Ragazzi
- Anatomia Patologica, Azienda Ospedaliera S. Maria Nuova di Reggio Emilia, Viale Risorgimento 80, 42123, Reggio Emilia, Italy
| | - Lara Gibellini
- Dipartimento di Scienze Mediche e Chirurgiche materno infantili e dell'adulto, Università degli Studi di Modena e Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Manuela Sollazzo
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Rene Gunther Feichtinger
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Silvia Vidali
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Maurizio Baldassarre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Sarah Foriel
- Khondrion BV, Philips van Leydenlaan 15, 6525 EX, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine (RCMM) at the Department of Pediatrics, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6500 HB, Nijmegen, The Netherlands
| | - Michele Vidone
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Andrea Cossarizza
- Dipartimento di Scienze Mediche e Chirurgiche materno infantili e dell'adulto, Università degli Studi di Modena e Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Daniela Grifoni
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Ilaria Malanchi
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK.
| | - Anna Maria Porcelli
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy.
- Centro Interdipartimentale di Ricerca Industriale Scienze della Vita e Tecnologie per la Salute, Università di Bologna, Via Tolara di Sopra 41/E, 40064, Ozzano dell'Emilia, Italy.
| | - Giuseppe Gasparre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
- Centro di Ricerca Biomedica Applicata (CRBA), Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| |
Collapse
|
21
|
|
22
|
Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. Metformin as an Anticancer Agent. Trends Pharmacol Sci 2018; 39:867-878. [PMID: 30150001 DOI: 10.1016/j.tips.2018.07.006] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022]
Abstract
Metformin has been a frontline therapy for type 2 diabetes (T2D) for many years. Its effectiveness in T2D treatment is mostly attributed to its suppression of hepatic gluconeogenesis; however, the mechanistic aspects of metformin action remain elusive. In addition to its glucose-lowering effect, metformin possesses other pleiotropic health-promoting effects that include reduced cancer risk and tumorigenesis. Metformin inhibits the electron transport chain (ETC) and ATP synthesis; however, recent data reveal that metformin regulates AMP-activated protein kinase (AMPK) and the mechanistic target of rapamycin complex 1 (mTORC1) by multiple, mutually nonexclusive mechanisms that do not necessarily depend on the inhibition of ETC and the cellular ATP level. In this review, we discuss recent advances in elucidating the molecular mechanisms that are relevant for metformin use in cancer treatment.
Collapse
Affiliation(s)
- Ales Vancura
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA.
| | - Pengli Bu
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Madhura Bhagwat
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Joey Zeng
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| | - Ivana Vancurova
- Department of Biological Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
23
|
Le-Rademacher J, Hillman SL, Meyers J, Loprinzi CL, Limburg PJ, Mandrekar SJ. Statistical controversies in clinical research: Value of adverse events relatedness to study treatment: analyses of data from randomized double-blind placebo-controlled clinical trials. Ann Oncol 2018; 28:1183-1190. [PMID: 28184420 DOI: 10.1093/annonc/mdx043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Background Collection and reporting of adverse events (AEs) and their relatedness to study treatment, known commonly as attribution, in clinical trials is mandated by regulatory agencies (the National Cancer Institute and the Food and Drug Administration). Attribution is assigned by the treating physician using judgment based on various factors including patient's baseline status, disease history, and comorbidity as well as knowledge about the safety profile of the study treatments. We evaluate the patterns of AE attribution (unrelated, unlikely, possibly, probably, and definitely related to the treatment) in treatment, symptom intervention (cancer patients) and cancer prevention (participants at high risk for cancer) setting. Materials and methods Nine multicenter placebo-controlled trials (two treatment, two symptom intervention, and five cancer prevention) were analysed separately (2155 patients). Frequency and severity of AEs were summarized by arm. Attribution and percentage of repeated AEs whose attribution changed overtime were summarized for the placebo arms. Percentage of physician over- or under-reporting of AE relatedness was calculated for the treatment arms using the placebo arm as the reference. Results Across all trials and settings, a very high proportion of AEs reported as related to treatment were classified as possibly related, a significant proportion of AEs in the placebo arm were incorrectly reported as related to treatment, and clinician-reported attribution over-estimated the rate of AEs related to treatment. Fatigue, nausea, vomiting, diarrhea, constipation, and neurosensory were the common AEs that were over reported by clinician as related to treatment. Conclusions These analyses demonstrate that assigning causality to AE is a complex and difficult process that produces unreliable and subjective data. In randomized double-blind placebo-controlled trials where data are available to objectively assess relatedness of AE to treatment, attribution assignment should be eliminated.
Collapse
Affiliation(s)
| | | | - J Meyers
- Departments of Health Sciences Research
| | | | - P J Limburg
- Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| | | |
Collapse
|
24
|
Gong J, Kelekar G, Shen J, Shen J, Kaur S, Mita M. The expanding role of metformin in cancer: an update on antitumor mechanisms and clinical development. Target Oncol 2017; 11:447-67. [PMID: 26864078 DOI: 10.1007/s11523-016-0423-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metformin has been used for nearly a century to treat type 2 diabetes mellitus. Epidemiologic studies first identified the association between metformin and reduced risk of several cancers. The anticancer mechanisms of metformin involve both indirect or insulin-dependent pathways and direct or insulin-independent pathways. Preclinical studies have demonstrated metformin's broad anticancer activity across a spectrum of malignancies. Prospective clinical trials involving metformin in the chemoprevention and treatment of cancer now number in the hundreds. We provide an update on the anticancer mechanisms of metformin and review the results thus far available from prospective clinical trials investigating metformin's efficacy in cancer.
Collapse
Affiliation(s)
- Jun Gong
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Gauri Kelekar
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - James Shen
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - John Shen
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Sukhpreet Kaur
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Monica Mita
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA. .,Experimental Therapeutics Program, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, SCCT Mezzanine MS 35, Los Angeles, CA, 90048, USA.
| |
Collapse
|
25
|
Cummings LC, Thota PN, Willis JE, Chen Y, Cooper GS, Furey N, Bednarchik B, Alashkar BM, Dumot J, Faulx AL, Fink SP, Kresak AM, Abusneineh B, Barnholtz-Sloan J, Leahy P, Veigl ML, Chak A, Markowitz SD. A nonrandomized trial of vitamin D supplementation for Barrett's esophagus. PLoS One 2017; 12:e0184928. [PMID: 28922414 PMCID: PMC5602627 DOI: 10.1371/journal.pone.0184928] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023] Open
Abstract
Background Vitamin D deficiency may increase esophageal cancer risk. Vitamin D affects genes regulating proliferation, apoptosis, and differentiation and induces the tumor suppressor 15-hydroxyprostaglandin dehydrogenase (PGDH) in other cancers. This nonrandomized interventional study assessed effects of vitamin D supplementation in Barrett’s esophagus (BE). We hypothesized that vitamin D supplementation may have beneficial effects on gene expression including 15-PGDH in BE. Methods BE subjects with low grade or no dysplasia received vitamin D3 (cholecalciferol) 50,000 international units weekly plus a proton pump inhibitor for 12 weeks. Esophageal biopsies from normal plus metaplastic BE epithelium and blood samples were obtained before and after vitamin D supplementation. Serum 25-hydroxyvitamin D was measured to characterize vitamin D status. Esophageal gene expression was assessed using microarrays. Results 18 study subjects were evaluated. The baseline mean serum 25-hydroxyvitamin D level was 27 ng/mL (normal ≥30 ng/mL). After vitamin D supplementation, 25-hydroxyvitamin D levels rose significantly (median increase of 31.6 ng/mL, p<0.001). There were no significant changes in gene expression from esophageal squamous or Barrett’s epithelium including 15-PGDH after supplementation. Conclusion BE subjects were vitamin D insufficient. Despite improved vitamin D status with supplementation, no significant alterations in gene expression profiles were noted. If vitamin D supplementation benefits BE, a longer duration or higher dose of supplementation may be needed.
Collapse
Affiliation(s)
- Linda C. Cummings
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- Medical Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- * E-mail:
| | - Prashanthi N. Thota
- Digestive Diseases Institute, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Joseph E. Willis
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- Department of Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yanwen Chen
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
| | - Gregory S. Cooper
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
| | - Nancy Furey
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Beth Bednarchik
- William T. Dahms Clinical Research Unit, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
| | - Bronia M. Alashkar
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - John Dumot
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Ashley L. Faulx
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Medical Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Stephen P. Fink
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
| | - Adam M. Kresak
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- Department of Pathology, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Basel Abusneineh
- Medical Service, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | | | - Patrick Leahy
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- Division of General Medical Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Martina L. Veigl
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
- Division of General Medical Sciences, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Amitabh Chak
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
| | - Sanford D. Markowitz
- Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, Ohio, United States of America
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, United States of America
- Case Comprehensive Cancer Center, Cleveland, Ohio, United States of America
| |
Collapse
|
26
|
Heckman-Stoddard BM, DeCensi A, Sahasrabuddhe VV, Ford LG. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia 2017; 60:1639-1647. [PMID: 28776080 PMCID: PMC5709147 DOI: 10.1007/s00125-017-4372-6] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 01/16/2023]
Abstract
Multiple epidemiological studies have documented an association between metformin, used for treatment of type 2 diabetes, and reduced cancer incidence and mortality. Cell line models may not accurately reflect the effects of metformin in the clinical setting. Moreover, findings from animal model studies have been inconsistent, whilst those from more recent epidemiological studies have tempered the overall effect size. The purpose of this review is to examine metformin's chemopreventive potential by outlining relevant mechanisms of action, the most recent epidemiologic evidence, and recently completed and ongoing clinical trials. Although repurposing drugs with excellent safety profiles is an appealing strategy for cancer prevention and treatment in the adjuvant setting, there is no substitute for well-executed, large randomised clinical trials to define efficacy and determine the populations that are most likely to benefit from an intervention. Thus, enthusiasm remains for understanding the role of metformin in cancer through ongoing clinical research.
Collapse
Affiliation(s)
- Brandy M Heckman-Stoddard
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA.
| | - Andrea DeCensi
- Division of Medical Oncology, Ente Ospedaliero Ospedali Galliera, Genoa, Italy
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK
| | - Vikrant V Sahasrabuddhe
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Leslie G Ford
- Division of Cancer Prevention, National Cancer Institute, 9609 Medical Center Drive, Bethesda, MD, 20892, USA
| |
Collapse
|
27
|
Hopkins BD, Goncalves MD, Cantley LC. Obesity and Cancer Mechanisms: Cancer Metabolism. J Clin Oncol 2016; 34:4277-4283. [PMID: 27903152 DOI: 10.1200/jco.2016.67.9712] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Obesity is a risk factor for cancer development and is associated with poor prognosis in multiple tumor types. The positive energy balance linked with obesity induces a variety of systemic changes including altered levels of insulin, insulin-like growth factor-1, leptin, adiponectin, steroid hormones, and cytokines. Each of these factors alters the nutritional milieu and has the potential to create an environment that favors tumor initiation and progression. Although the complete ramifications of obesity as it relates to cancer are still unclear, there is convincing evidence that reducing the magnitude of the systemic hormonal and inflammatory changes has significant clinical benefits. This review will examine the changes that occur in the obese state and review the biologic mechanisms that connect these changes to increased cancer risk. Understanding the metabolic changes that occur in obese individuals may also help to elucidate more effective treatment options for these patients when they develop cancer. Moving forward, targeted clinical trials examining the effects of behavioral modifications such as reduced carbohydrate intake, caloric restriction, structured exercise, and/or pharmacologic interventions such as the use of metformin, in obese populations may help to reduce their cancer risk.
Collapse
Affiliation(s)
- Benjamin D Hopkins
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Marcus D Goncalves
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| | - Lewis C Cantley
- All authors: Meyer Cancer Center, Weill Cornell Medical College, New York, NY
| |
Collapse
|
28
|
Banerjee B, Shaheen NJ, Martinez JA, Hsu CH, Trowers E, Gibson BA, Della'Zanna G, Richmond E, Chow HHS. Clinical Study of Ursodeoxycholic Acid in Barrett's Esophagus Patients. Cancer Prev Res (Phila) 2016; 9:528-33. [PMID: 26908564 PMCID: PMC4930711 DOI: 10.1158/1940-6207.capr-15-0276] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023]
Abstract
Prior research strongly implicates gastric acid and bile acids, two major components of the gastroesophageal refluxate, in the development of Barrett's esophagus and its pathogenesis. Ursodeoxycholic acid (UDCA), a hydrophilic bile acid, has been shown to protect esophageal cells against oxidative stress induced by cytotoxic bile acids. We conducted a pilot clinical study to evaluate the clinical activity of UDCA in patients with Barrett's esophagus. Twenty-nine patients with Barrett's esophagus received UDCA treatment at a daily dose of 13 to 15 mg/kg/day for 6 months. The clinical activity of UDCA was assessed by evaluating changes in gastric bile acid composition and markers of oxidative DNA damage (8-hydroxydeoxyguanosine), cell proliferation (Ki67), and apoptosis (cleaved caspase-3) in Barrett's esophagus epithelium. The bile acid concentrations in gastric fluid were measured by liquid chromatography/mass spectrometry. At baseline, UDCA (sum of unchanged and glycine/taurine conjugates) accounted for 18.2% of total gastric bile acids. After UDCA intervention, UDCA increased significantly to account for 93.4% of total gastric bile acids (P < 0.0001). The expression of markers of oxidative DNA damage, cell proliferation, and apoptosis was assessed in the Barrett's esophagus biopsies by IHC. The selected tissue biomarkers were unchanged after 6 months of UDCA intervention. We conclude that high-dose UDCA supplementation for 6 months resulted in favorable changes in gastric bile acid composition but did not modulate selected markers of oxidative DNA damage, cell proliferation, and apoptosis in the Barrett's esophagus epithelium. Cancer Prev Res; 9(7); 528-33. ©2016 AACRSee related article by Brian J. Reid, p. 512.
Collapse
Affiliation(s)
| | - Nicholas J Shaheen
- Division of Gastroenterology & Hepatology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Jessica A Martinez
- University of Arizona Cancer Center, Tucson, Arizona. Department of Nutritional Sciences, University of Arizona, Tucson, Arizona
| | | | - Eugene Trowers
- College of Medicine, University of Arizona, Tucson, Arizona
| | - Blake A Gibson
- College of Medicine, University of Arizona, Tucson, Arizona
| | - Gary Della'Zanna
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | - Ellen Richmond
- Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland
| | | |
Collapse
|
29
|
Krishnamoorthi R, Borah B, Heien H, Das A, Chak A, Iyer PG. Rates and predictors of progression to esophageal carcinoma in a large population-based Barrett's esophagus cohort. Gastrointest Endosc 2016; 84:40-46.e7. [PMID: 26772891 PMCID: PMC4912845 DOI: 10.1016/j.gie.2015.12.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 12/30/2015] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Rates of progression to esophageal adenocarcinoma in subjects with Barrett's esophagus (BE) are lower than previously estimated. Identification of predictors of progression will enable risk stratification of BE subjects, potentially making current surveillance programs more efficient. We aimed to assess the potential of demographic and lifestyle factors, obesity, and medications in predicting progression in BE. METHODS BE subjects were identified from the General Practice Research Database using validated diagnostic codes. BE subjects developing esophageal cancer (EC) 12 months after their index BE diagnosis were defined as progressors. Time-to-event analysis was used to assess the overall risk of progression to EC. Cox proportional hazards models and time-varying marginal structural models were used to assess predictors of progression. RESULTS Included in the analysis were 9660 BE patients. The mean age (SD) of the study subjects was 63 (13.5) years; 62.6% were men. One hundred three subjects (1.1%) progressed to EC. The mean (SD) follow-up since initial diagnosis was 4.8 (3.3) years. The incidence of EC was 2.23 per 1000 person-years of follow-up. Increasing age, male gender, and being overweight (body mass index, 25-29.9) were found to be independent predictors of progression. When time-varying models were used, proton pump inhibitor (PPI) and statin use were protective against progression. CONCLUSIONS In this large population-based cohort of patients with BE, increasing age, male gender, and being overweight predicted progression to EC, whereas PPI and statin use were protective against EC development. These factors may aid in developing a risk score to predict the risk of progression and chemopreventive strategies in patients with BE.
Collapse
Affiliation(s)
| | - Bijan Borah
- Division of Health Care Policy and Research, Mayo Clinic, Rochester, MN 55905
| | - Herbert Heien
- Division of Health Care Policy and Research, Mayo Clinic, Rochester, MN 55905
| | - Ananya Das
- Arizona Center for Digestive Health, Gilbert, AZ
| | - Amitabh Chak
- Division of Gastroenterology & Hepatology, Case Western Reserve University, Cleveland, OH
| | - Prasad G. Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
30
|
Jin Z, Yan W, Jin H, Ge C, Xu Y. Psoralidin inhibits proliferation and enhances apoptosis of human esophageal carcinoma cells via NF-κB and PI3K/Akt signaling pathways. Oncol Lett 2016; 12:971-976. [PMID: 27446379 DOI: 10.3892/ol.2016.4716] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/18/2015] [Indexed: 12/27/2022] Open
Abstract
Esophageal cancer is the most common gastrointestinal cancer. Psoralidin exhibits antioxidant, anti-apoptotic, anti-inflammatory and antitumor effects, which result in the inhibition of cancer formation. The present study aimed to investigate the effect of psoralidin on esophageal carcinoma proliferation and growth, and to elucidate its underlying mechanism of action. The effect of psoralidin on cell proliferation was investigated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay. Using an annexin V-fluorescein isothiocyanate/propidium iodide apoptosis detection kit and 4',6-diamidino-2-phenylindole staining assay, the present study demonstrated that psoralidin significantly enhanced apoptosis of human esophageal carcinoma Eca9706 cells. In addition, caspase-3 activity was analyzed with a caspase-3 colorimetric assay kit, while nuclear factor (NF)-κB activity and protein phosphatidylinositol 3-kinase (PI3K)/Akt expression were measured with an NF-κB enzyme-linked immunosorbent assay kit and western blot analysis, respectively. Eca9706 cells were treated with a PI3K agonist in order to investigate the mechanism of action of psoralidin. It was observed that psoralidin was able to decrease the proliferation and promote the cellular apoptosis of Eca9706 cells in a dose-dependent manner. Furthermore, psoralidin was also able to inhibit the caspase-3 activity of Eca9706 cells in a dose-dependent manner. In addition, psoralidin inhibited NF-κB activity and reduced PI3K and Akt protein expression in Eca9706 cells. Notably, the PI3K agonist was able to reverse the effect of psoralidin on Eca9706 cells. The results of the present study demonstrated that psoralidin was able to inhibit proliferation and enhance apoptosis of human esophageal carcinoma cells via the NF-κB and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Zhiliang Jin
- Department of Oncology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Wei Yan
- Department of Gastroenterology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Hui Jin
- Department of Neonatology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Changzheng Ge
- Department of Oncology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| | - Yanhua Xu
- Department of Oncology, Jingzhou Central Hospital, Jingzhou, Hubei 434020, P.R. China
| |
Collapse
|
31
|
Şenateş E. Chemoprevention of Barrett’s Esophagus and Adenocarcinoma. BARRETT'S ESOPHAGUS 2016:189-204. [DOI: 10.1016/b978-0-12-802511-6.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
32
|
Thrift AP. Esophageal Adenocarcinoma: The Influence of Medications Used to Treat Comorbidities on Cancer Prognosis. Clin Gastroenterol Hepatol 2015; 13:2225-32. [PMID: 25835331 DOI: 10.1016/j.cgh.2015.03.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 03/23/2015] [Accepted: 03/25/2015] [Indexed: 02/07/2023]
Abstract
Esophageal adenocarcinoma has undergone a continuous rise in incidence since the early 1970s and is the fastest rising cancer among white men in the United States. Epidemiologic studies have demonstrated that medications commonly used to treat multiple chronic conditions (for example, aspirin, non-aspirin nonsteroidal anti-inflammatory drugs, and statins) as well as powerful acid suppressants such as proton pump inhibitors are associated with a reduced risk of esophageal adenocarcinoma. The chemopreventive potential of these classes of medications appears to be especially applicable to persons with Barrett's esophagus, the only known premalignant condition for esophageal adenocarcinoma. However, it is not known whether these medications also influence cancer recurrence and cancer-specific mortality in persons diagnosed with esophageal adenocarcinoma. This is an important question because most patients with esophageal adenocarcinoma have 1 or more comorbid conditions at the time of their cancer diagnosis and are receiving medication to treat these conditions. This article summarizes the evidence on the associations between 4 commonly used classes of medications and (1) risk of developing esophageal adenocarcinoma and Barrett's esophagus and (2) risk of cancer recurrence and cancer-specific mortality in patients with esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Aaron P Thrift
- Department of Medicine and Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
33
|
Nguyen T, Duan Z, Naik AD, Kramer JR, El-Serag HB. Statin use reduces risk of esophageal adenocarcinoma in US veterans with Barrett's esophagus: a nested case-control study. Gastroenterology 2015. [PMID: 26208896 DOI: 10.1053/j.gastro.2015.07.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
BACKGROUND & AIMS Statins have been reported to protect against esophageal adenocarcinoma (EAC) in patients with Barrett's esophagus (BE). However, there are few data from adequately powered cohort studies of subjects with BE. METHODS We conducted a nested case-control study of a cohort of BE patients identified from national Veteran Affairs (VA) outpatient files, diagnosed with BE from 2004 through 2009. New cases of EAC recorded after BE diagnosis were identified during a follow-up period that ended in 2011 and verified using electronic medical records. We selected patients with BE without EAC (controls) using incidence density sampling; 3 controls were matched to each case based on birth year and date of BE diagnosis. Our analysis included only male patients with at least 1 VA visit per year of follow up. We identified prescriptions for statins and non-statin lipid lowering medications filled after BE diagnosis and up to 90 days before EAC diagnosis for cases and controls (during the corresponding time period); we examined the association between statin use and EAC in conditional logistic regression models. RESULTS We compared 311 EAC cases to 856 controls. Cases were less likely to use any statins than controls (40.2% vs 54.0%; P < .01). Significantly lower proportions of cases used statins for 6-18 months (10.0% cases vs 17.1% controls) and >18 months (19.3% vs 24.0%, respectively; P < .01). Simvastatin was the most commonly prescribed statin (accounting for 86.9% of statin use); the defined daily dose of simvastatin was lower in cases than in controls (21-40 mg/day, 9.3% vs 14.5%, respectively; and >40 mg/day, 8.4% vs 12.6%, respectively; P < .01). In multivariate analysis, statin use was inversely associated with development of EAC (adjusted odds ratio [OR], 0.65; 95% confidence interval [CI], 0.47-0.91). This protective association was strongest for patients with advanced-stage EAC: in a stratified analysis, comparison of 189 cases with stage 0-1 EAC to 520 controls produced an adjusted OR of 0.85 (95% CI, 0.54-1.33). Among patients with late-stage EAC (stages 2-4, n = 106) and 291 controls, the adjusted OR was 0.44 (95% CI, 0.25-0.79). We found no association between EAC and non-statin lipid-lowering medications. CONCLUSIONS In a case-control study of US veterans, statin use among those with BE appeared to decrease the risk of EAC. This protective effect was strongest against advanced-stage EAC, and increased with statin dose.
Collapse
Affiliation(s)
- Theresa Nguyen
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas; Section of Health Services Research, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas
| | - Zhigang Duan
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas
| | - Aanand D Naik
- Section of Health Services Research, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas
| | - Jennifer R Kramer
- Section of Health Services Research, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas
| | - Hashem B El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas; Section of Health Services Research, Department of Medicine, Michael E. DeBakey Veterans' Affairs Medical Center, Baylor College Medicine, Houston, Texas.
| |
Collapse
|
34
|
Heckman-Stoddard BM, Gandini S, Puntoni M, Dunn BK, DeCensi A, Szabo E. Repurposing old drugs to chemoprevention: the case of metformin. Semin Oncol 2015; 43:123-133. [PMID: 26970131 DOI: 10.1053/j.seminoncol.2015.09.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Multiple epidemiologic studies have documented an association between the anti-diabetic agent metformin and reduced cancer incidence and mortality. However, this effect has not been consistently demonstrated in animal models or more recent epidemiological studies. The purpose of this paper is to examine metformin's chemopreventive potential by reviewing relevant mechanisms of action, preclinical evidence of efficacy, updated epidemiologic evidence after correction for potential biases and confounders, and recently completed and ongoing clinical trials. Although repurposing drugs with well described mechanisms of action and safety profiles is an appealing strategy for cancer prevention, there is no substitute for well executed late phase clinical trials to define efficacy and populations that are most likely to benefit from an intervention.
Collapse
Affiliation(s)
| | - Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Matteo Puntoni
- Office of the Scientific Director, E.O. Ospedali Galliera, Genoa, Italy
| | - Barbara K Dunn
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Andrea DeCensi
- Division of Medical Oncology, E.O. Ospedali Galliera, Genoa, Italy; Division of Cancer Prevention and Genetics, European Institute of Oncology, Milan, Italy
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
35
|
Halland M, Katzka D, Iyer PG. Recent developments in pathogenesis, diagnosis and therapy of Barrett's esophagus. World J Gastroenterol 2015; 21:6479-6490. [PMID: 26074687 PMCID: PMC4458759 DOI: 10.3748/wjg.v21.i21.6479] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Revised: 01/31/2015] [Accepted: 04/28/2015] [Indexed: 02/06/2023] Open
Abstract
The burden of illness from esophageal adenocarcinoma continues to rise in the Western world, and overall prognosis is poor. Given that Barrett’s esophagus (BE), a metaplastic change in the esophageal lining is a known cancer precursor, an opportunity to decrease disease development by screening and surveillance might exist. This review examines recent updates in the pathogenesis of BE and comprehensively discusses known risk factors. Diagnostic definitions and challenges are outlined, coupled with an in-depth review of management. Current challenges and potential solutions related to screening and surveillance are discussed. The effectiveness of currently available endoscopic treatment techniques, particularly with regards to recurrence following successful endotherapy and potential chemopreventative agents are also highlighted. The field of BE is rapidly evolving and improved understanding of pathophysiology, combined with emerging methods for screening and surveillance offer hope for future disease burden reduction.
Collapse
|
36
|
Zeb MH, Baruah A, Kossak SK, Buttar NS. Chemoprevention in Barrett's Esophagus: Current Status. Gastroenterol Clin North Am 2015; 44:391-413. [PMID: 26021201 DOI: 10.1016/j.gtc.2015.02.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chemoprevention in Barrett's esophagus is currently applied only in research settings. Identifying pathways that can be targeted by safe, pharmaceutical or natural compounds is key to expanding the scope of chemoprevention. Defining meaningful surrogate markers of cancer progression is critical to test the efficacy of chemopreventive approaches. Combinatorial chemoprevention that targets multiple components of the same pathway or parallel pathways could reduce the risk and improve the efficacy of chemoprevention. Here we discuss the role of chemoprevention as an independent or an adjuvant management option in BE-associated esophageal adenocarcinoma.
Collapse
Affiliation(s)
- Muhammad H Zeb
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Anushka Baruah
- Department of Internal Medicine, John H. Stroger, Jr. Hospital of Cook County, 1901 W. Harrison Street, Chicago, IL 60612, USA
| | - Sarah K Kossak
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Navtej S Buttar
- Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
37
|
Abstract
Central obesity is involved in the pathogenesis and progression of Barrett's esophagus to esophageal adenocarcinoma. Involved are likely both mechanical and nonmechanical effects. Mechanical effects of increased abdominal fat cause disruption of the gastroesophageal reflux barrier leading to increased reflux events. Nonmechanical effects may be mediated by inflammation, via classically activated macrophages, pro-inflammatory cytokines, and adipokines such as Leptin, all of which likely potentiate reflux-mediated inflammation. Insulin resistance, associated with central obesity, is also associated with both Barrett's pathogenesis and progression to adenocarcinoma. Molecular pathways activated in obesity, inflammation and insulin resistance overlap with those involved in Barrett's pathogenesis and progression.
Collapse
|
38
|
Baruah A, Buttar NS. Chemoprevention in Barrett's oesophagus. Best Pract Res Clin Gastroenterol 2015; 29:151-65. [PMID: 25743463 DOI: 10.1016/j.bpg.2014.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 12/11/2014] [Indexed: 01/31/2023]
Abstract
Increasing incidence of oesophageal adenocarcinoma along with poor survival entails novel preventive strategies. Agents that target pro-oncogenic pathways in Barrett's mucosa could halt this neoplastic transformation. In this review, we will use epidemiological associations and molecular mechanisms to identify novel chemoprevention targets in Barrett's oesophagus. We will also discuss recent chemoprevention trials.
Collapse
Affiliation(s)
- Anushka Baruah
- Mayo Clinic College of Medicine, Department of Gastroenterology and Hepatology, Rochester, MN, USA
| | - Navtej S Buttar
- Mayo Clinic College of Medicine, Department of Gastroenterology and Hepatology, Rochester, MN, USA.
| |
Collapse
|