1
|
Mondal T, Chattopadhyay D, Saha Mondal P, Das S, Mondal A, Das A, Samanta S, Saha T. Fusobacterium nucleatum modulates the Wnt/β-catenin pathway in colorectal cancer development. Int J Biol Macromol 2025; 299:140196. [PMID: 39848378 DOI: 10.1016/j.ijbiomac.2025.140196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The Wnt/β-catenin signalling pathway normally maintains cellular and tissue homeostasis by regulating cellular differentiation and survival in a controlled manner. An aberrantly regulated Wnt/β-catenin signalling pathway can transform into an oncogenic pathway, which is associated with Colorectal cancer (CRC) as well as other cancers. CRC is one of the most frequently occurring gastrointestinal cancers worldwide. In CRC tissues, deregulation of Wnt/β-catenin pathway is observed, which indicates that this oncogenic pathway directly promotes CRC malignancy, cell migration, angiogenesis, chemoresistance, as well as shorter lifespan of a patient. Growing evidence suggests that human commensal microbes have a strong association with carcinogenesis, particularly the prevalence and high enrichment of Fusobacterium nucleatum in CRC progression. The Wnt/β-catenin pathway is one of the targeted pathways by F. nucleatum in CRC, where Fusobacterium adhesin attaches to E-cadherin to initiate infection. Also, Wnt/β-catenin pathway can be a potential target for the treatment of both CRC and F. nucleatum-positive CRC. Here, we discuss the underlying mechanisms of F. nucleatum-positive CRC development through modulation of Wnt/β-catenin signalling and its possibility for the application in targeted therapy of F. nucleatum-positive CRC.
Collapse
Affiliation(s)
- Tanushree Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Deepanjan Chattopadhyay
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Paromita Saha Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Sanjib Das
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Amalesh Mondal
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India; Department of Physiology, Katwa Collage, Katwa, Purba Bardhaman, West Bengal 713130, India
| | - Abhishek Das
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Subhasree Samanta
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India
| | - Tanima Saha
- Department of Molecular Biology and Biotechnology, University of Kalyani, Kalyani 741235, Nadia, West Bengal, India.
| |
Collapse
|
2
|
Khamees N, Al-Ani A, Tamimi TA, Sarhan O, Matouq Y, Laswi D, Arabiyat D, Rayyan N, Ali MR, Al-Slaimieh AI, Rayyan YM. Epidemiology and clinical characteristics of colorectal cancer and advanced adenoma: a single center experience in Jordan. BMC Gastroenterol 2025; 25:120. [PMID: 40016635 PMCID: PMC11866683 DOI: 10.1186/s12876-024-03531-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 11/21/2024] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVES We evaluated the epidemiology and clinical characteristics of colorectal polyps to formulate an appropriate screening program. METHODS A retrospective chart review was conducted on all patients who underwent complete colonoscopy at Jordan University Hospital from January to September 2018. Demographics, comorbidities, lifestyle habits, medication history, family history of cancer, laboratory parameters, quality of bowel preparation, and polyp characteristics were evaluated. Binary logistic regression was utilized to find predictors of colorectal polyps. RESULTS A total of 965 patients were included in the study, with a mean age of 53.9 ± 17.1 years and a male predominance (52.7%). Polyps were detected in 28.1% of patients, with 18% having one polyp, 10.4% having two polyps, and 3.3% having more than two polyps. Multivariate analysis demonstrated that older age, high BMI, male gender, diabetes mellitus, dyslipidemia, ischemic heart disease, and family history of CRC were positive predictors of polyps. The right colon (cecum and ascending colon) was the most common location for polyps (51%), followed by the sigmoid colon (24.8%). The most common histologic subtype of polyps was tubular adenoma (48.2%). The prevalence of CRC was 18.65 per 1000 patients. CONCLUSION We highlight the fair prevalence of colorectal polyps and CRC in a Jordanian cohort. Awareness campaigns, screening strategies, and promotion of healthy lifestyles could help alleviate the burden of the disease, particularly among patients with classical risk factors for CRC.
Collapse
Affiliation(s)
- Nadia Khamees
- Department of Internal Medicine, Section of Gastroenterology, Jordan University Hospital, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Abdallah Al-Ani
- School of Medicine, The University of Jordan, Amman, Jordan
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
- King Hussein Cancer Center, Amman, Jordan
| | - Tarek A Tamimi
- Department of Internal Medicine, Section of Gastroenterology, Jordan University Hospital, Amman, Jordan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Omar Sarhan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Yazan Matouq
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Dana Laswi
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Dima Arabiyat
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Nadin Rayyan
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Mustafa Rami Ali
- School of Medicine, The University of Jordan, Amman, Jordan
- Office of Scientific Affairs and Research, King Hussein Cancer Center, Amman, Jordan
| | | | - Yaser M Rayyan
- Department of Internal Medicine, Section of Gastroenterology, Jordan University Hospital, Amman, Jordan.
- School of Medicine, The University of Jordan, Amman, Jordan.
| |
Collapse
|
3
|
Heriz MH, Razzak Mahmood AA, Tahtamouni LH, Al-Sakhen MF, Kanaan SI, Saleh KM, Yasin SR. New Carbothioamide and Carboxamide Derivatives of 3-Phenoxybenzoic Acid as Potent VEGFR-2 Inhibitors: Synthesis, Molecular Docking, and Cytotoxicity Assessment. Curr Cancer Drug Targets 2025; 25:412-430. [PMID: 38747227 DOI: 10.2174/0115680096307334240429050730] [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: 01/30/2024] [Revised: 03/16/2024] [Accepted: 03/31/2024] [Indexed: 04/12/2025]
Abstract
INTRODUCTION/BACKGROUND Because of the well-established link between angiogenesis and tumor development, the use of antiangiogenic therapeutics, such as those targeting VEGFR-2, presents a promising approach to cancer treatment. In the current study, a set of five hydrazine-1- carbothioamide (compounds 3a-e) and three hydrazine-1-carboxamide derivatives (compounds 4a-c) were successfully synthesized from 3-phenoxybenzoic acid. These compounds were specially created as antiproliferative agents with the goal of targeting cancer cells by inhibiting VEGFR- 2 tyrosine kinase. MATERIALS AND METHODS The new derivatives were synthesized by conventional organic methods, and their structure was versified by IR, 1HNMR, 13CNMR, and mass spectroscopy. In silico investigation was carried out to identify the compounds' target, molecular similarity, ADMET, and toxicity profile. The cytotoxic activity of the prepared compounds was evaluated in vitro against three human cancer cell lines (DLD1 colorectal adenocarcinoma, HeLa cervical cancer, and HepG2 hepatocellular carcinoma). The effects of the leading compound on cell cycle progression and apoptosis induction were investigated by flow cytometry, and the specific apoptotic pathway triggered by the treatment was evaluated by RT-PCR and immunoblotting. Finally, the inhibitory activities of the new compounds against VEGFR-2 was measured. RESULTS The designed derivatives exhibited comparable binding positions and interactions to the VEGFR-2 binding site to that of sorafenib (a standard VEGFR-2 tyrosine kinase inhibitor), as determined by molecular docking analysis. Compound 4b was the most cytotoxic compound, achieving the lowest IC50 against HeLa cells. Compound 4b, a strong representative of the synthesized series, induced cell cycle arrest at the G2/M phase, increased the proportion of necrotic and apoptotic HeLa cells, and activated caspase 3. The EC50 value of compound 4b against VEGFR-2 kinase activity was comparable to sorafenib's. CONCLUSION Overall, the findings suggest that compound 4b has a promising future as a starting point for the development of new anticancer drugs.
Collapse
Affiliation(s)
- Mohammad Hamza Heriz
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Al-Zahraa University for Women, Karbala-Baghdad Street, Karbala, Iraq
| | - Ammar A Razzak Mahmood
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Baghdad, Bab-Almoudam, 10001, Baghdad, Iraq
| | - Lubna H Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
- Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Mai F Al-Sakhen
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Sana I Kanaan
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Khaled M Saleh
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| | - Salem R Yasin
- Department of Biology and Biotechnology, Faculty of Science, The Hashemite University, Zarqa, Jordan
| |
Collapse
|
4
|
Tien P, Bih ZL, Chen WM, Shia BC, Wu SY, Chiang CW. Aspirin use reduces cancer risk in betel nut chewers: a nationwide population-based cohort study. Am J Cancer Res 2024; 14:5921-5934. [PMID: 39803639 PMCID: PMC11711529 DOI: 10.62347/jxmi9007] [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: 10/09/2024] [Accepted: 11/10/2024] [Indexed: 01/16/2025] Open
Abstract
Betel nut chewing, common in several Asian populations, is linked to increased cancer risk, including oral, esophageal, gastric, and hepatocellular carcinoma. Aspirin shows potential as a chemopreventive agent. This study investigates the association between aspirin use and cancer risk among betel nut chewers. Betel nut chewers aged 18 and older were included, with aspirin use defined as at least 28 cumulative defined daily doses (cDDDs). Propensity score matching and Cox proportional hazards models, adjusted for time-varying covariates, were used to assess cancer risk. The study included 46,302 betel nut chewers, equally divided between aspirin users and non-users. Aspirin use was associated with a 31% reduction in overall cancer risk (adjusted hazard ratio [aHR], 0.69; 95% confidence interval [CI], 0.66 to 0.73; P<0.0001). A dose-response relationship was observed, with higher cDDDs of aspirin corresponding to greater reductions in cancer risk. The highest quartile of aspirin use (Quartile 4) showed a 62% reduction in cancer risk (aHR, 0.38; 95% CI, 0.34 to 0.41; P<0.0001). Daily aspirin intensity was also associated with a significant reduction in cancer risk, with doses greater than 1 DDD showing an aHR of 0.54 (95% CI, 0.47 to 0.61; P<0.0001) compared to 1 DDD or less. Aspirin use significantly reduces cancer risk among betel nut chewers in a dose-dependent manner. These findings suggest aspirin as a potential chemopreventive agent in high-risk populations, warranting further investigation.
Collapse
Affiliation(s)
- Peng Tien
- Department of Otorhinolaryngology, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| | - Zen Lang Bih
- Department of Emergency Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| | - Wan-Ming Chen
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic UniversityTaipei 242, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic UniversityTaipei 242, Taiwan
| | - Ben-Chang Shia
- Graduate Institute of Business Administration, College of Management, Fu Jen Catholic UniversityTaipei 242, Taiwan
- Artificial Intelligence Development Center, Fu Jen Catholic UniversityTaipei 242, Taiwan
| | - Szu-Yuan Wu
- Department of Food Nutrition and Health Biotechnology, College of Medical and Health Science, Asia UniversityTaichung 413, Taiwan
- Division of Radiation Oncology, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
- Big Data Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
- Department of Healthcare Administration, College of Medical and Health Science, Asia UniversityTaichung 413, Taiwan
- Cancer Center, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
- Centers for Regional Anesthesia and Pain Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical UniversityTaipei 116, Taiwan
| | - Ching-Wen Chiang
- Department of Otorhinolaryngology, Lo-Hsu Medical Foundation, Lotung Poh-Ai HospitalYilan 265, Taiwan
| |
Collapse
|
5
|
Sikavi DR, Wang K, Ma W, Drew DA, Ogino S, Giovannucci EL, Cao Y, Song M, Nguyen LH, Chan AT. Aspirin Use and Incidence of Colorectal Cancer According to Lifestyle Risk. JAMA Oncol 2024; 10:1354-1361. [PMID: 39088221 PMCID: PMC11295063 DOI: 10.1001/jamaoncol.2024.2503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/19/2024] [Indexed: 08/02/2024]
Abstract
Importance Aspirin reduces the risk of colorectal cancer (CRC). Identifying individuals more likely to benefit from regular aspirin use for CRC prevention is a high priority. Objective To assess whether aspirin use is associated with the risk of CRC across different lifestyle risk factors. Design, Setting, and Participants A prospective cohort study among women in the Nurses' Health Study (1980-2018) and men in the Health Professionals Follow-Up Study (1986-2018) was conducted. Data analysis was performed from October 1, 2021, to May 22, 2023. Exposures A healthy lifestyle score was calculated based on body mass index, alcohol intake, physical activity, diet, and smoking with scores ranging from 0 to 5 (higher values corresponding to a healthier lifestyle). Regular aspirin use was defined as 2 or more standard tablets (325 mg) per week. Main Outcome and Measures Outcomes included multivariable-adjusted 10-year cumulative incidence of CRC, absolute risk reduction (ARR), and number needed to treat associated with regular aspirin use by lifestyle score and multivariable-adjusted hazard ratios for incident CRC across lifestyle scores. Results The mean (SD) baseline age of the 107 655 study participants (63 957 women from the Nurses' Health Study and 43 698 men from the Health Professionals Follow-Up Study) was 49.4 (9.0) years. During 3 038 215 person-years of follow-up, 2544 incident cases of CRC were documented. The 10-year cumulative CRC incidence was 1.98% (95% CI, 1.44%-2.51%) among participants who regularly used aspirin compared with 2.95% (95% CI, 2.31%-3.58%) among those who did not use aspirin, corresponding to an ARR of 0.97%. The ARR associated with aspirin use was greatest among those with the unhealthiest lifestyle scores and progressively decreased with healthier lifestyle scores (P < .001 for additive interaction). The 10-year ARR for lifestyle scores 0 to 1 (unhealthiest) was 1.28%. In contrast, the 10-year ARR for lifestyle scores 4 to 5 (healthiest) was 0.11%. The 10-year number needed to treat with aspirin was 78 for participants with lifestyle scores 0 to 1, 164 for score 2, 154 for score 3, and 909 for scores 4 to 5. Among the components of the healthy lifestyle score, the greatest differences in ARR associated with aspirin use were observed for body mass index and smoking. Conclusions and Relevance In this cohort study, aspirin use was associated with a greater absolute reduction in risk of CRC among individuals with less healthy lifestyles. The findings of the study suggest that lifestyle risk factors may be useful to identify individuals who may have a more favorable risk-benefit profile for cancer prevention with aspirin.
Collapse
Affiliation(s)
- Daniel R. Sikavi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Kai Wang
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Wenjie Ma
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - David A. Drew
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Shuji Ogino
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Cancer Immunology and Cancer Epidemiology Programs, Dana-Farber Harvard Cancer Center, Boston, Massachusetts
| | - Edward L. Giovannucci
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
- Alvin J. Siteman Cancer Center, Washington University School of Medicine, St Louis, Missouri
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St Louis, Missouri
| | - Mingyang Song
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Epidemiology, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
| | - Long H. Nguyen
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
- Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Immunology & Infectious Disease, Harvard T. H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| |
Collapse
|
6
|
Isingizwe ZR, Mortan LF, Benbrook DM. Platelet and epithelial cell interations can be modeled in cell culture, and are not affected by dihomo-gamma-linolenic acid. PLoS One 2024; 19:e0309125. [PMID: 39190751 PMCID: PMC11349180 DOI: 10.1371/journal.pone.0309125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Increasing evidence is implicating roles for platelets in the development and progression of ovarian cancer, a highly lethal disease that can arise from the fallopian tubes, and has no current method of early detection or prevention. Thrombosis is a major cause of mortality of ovarian cancer patients suggesting that the cancer alters platelet behavior. The objective of this study was to develop a cell culture model of the pathological interactions of human platelets and ovarian cancer cells, using normal FT epithelial cells as a healthy control, and to test effects of the anti-platelet dihomo-gamma-linolenic acid (DGLA) in the model. Both healthy and cancer cells caused platelet aggregation, however platelets only affected spheroid formation by cancer cells and had no effect on healthy cell spheroid formation. When naturally-formed spheroids of epithelial cells were exposed to platelets in transwell inserts that did not allow direct interactions of the two cell types, platelets caused increased size of the spheroids formed by cancer cells, but not healthy cells. When cancer cell spheroids formed using magnetic nanoshuttle technology were put in direct physical contact with platelets, the platelets caused spheroid condensation. In ovarian cancer cells, DGLA promoted epithelial-to-mesenchymal (EMT) transition at doses as low as 100 μM, and inhibited metabolic viability and induced apoptosis at doses ≥150 μM. DGLA doses ≤150 μM used to avoid direct DGLA effects on cancer cells, had no effect on the pathological interactions of platelets and ovarian cancer cells in our models. These results demonstrate that the pathological interactions of platelets with ovarian cancer cells can be modeled in cell culture, and that DGLA has no effect on these interactions, suggesting that targeting platelets is a rational approach for reducing cancer aggressiveness and thrombosis risk in ovarian cancer patients, however DGLA is not an appropriate candidate for this strategy.
Collapse
Affiliation(s)
- Zitha Redempta Isingizwe
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Laura F. Mortan
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Doris Mangiaracina Benbrook
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Pathology, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| |
Collapse
|
7
|
Larsson P, Olsson M, Sarathchandra S, Fäldt Beding A, Forssell-Aronsson E, Kovács A, Karlsson P, Helou K, Parris TZ. Multi-omics analysis identifies repurposing bortezomib in the treatment of kidney-, nervous system-, and hematological cancers. Sci Rep 2024; 14:18576. [PMID: 39127727 PMCID: PMC11316778 DOI: 10.1038/s41598-024-62339-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 05/15/2024] [Indexed: 08/12/2024] Open
Abstract
Repurposing of FDA-approved drugs is a quick and cost-effective alternative to de novo drug development. Here, we identify genes involved in bortezomib sensitivity, predict cancer types that may benefit from treatment with bortezomib, and evaluate the mechanism-of-action of bortezomib in breast cancer (BT-474 and ZR-75-30), melanoma (A-375), and glioblastoma (A-172) cells in vitro. Cancer cell lines derived from cancers of the blood, kidney, nervous system, and skin were found to be significantly more sensitive to bortezomib than other organ systems. The in vitro studies confirmed that although bortezomib effectively inhibited the β5 catalytic site in all four cell lines, cell cycle arrest was only induced in G2/M phase and apoptosis in A-375 and A-172 after 24h. The genomic and transcriptomic analyses identified 33 genes (e.g. ALDH18A1, ATAD2) associated with bortezomib resistance. Taken together, we identified biomarkers predictive of bortezomib sensitivity and cancer types that might benefit from treatment with bortezomib.
Collapse
Affiliation(s)
- Peter Larsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - Maxim Olsson
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | | | - Anna Fäldt Beding
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Southern Älvsborg Hospital, Borås, Sweden
| | - Eva Forssell-Aronsson
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Physics and Biomedical Engineering, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Anikó Kovács
- Department of Clinical Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Karlsson
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Khalil Helou
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Toshima Z Parris
- Department of Oncology, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Nafisi S, Støer NC, Veierød MB, Randel KR, Hoff G, Löfling L, Bosetti C, Botteri E. Low-Dose Aspirin and Prevention of Colorectal Cancer: Evidence From a Nationwide Registry-Based Cohort in Norway. Am J Gastroenterol 2024; 119:1402-1411. [PMID: 38300127 PMCID: PMC11208058 DOI: 10.14309/ajg.0000000000002695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/24/2024] [Indexed: 02/02/2024]
Abstract
INTRODUCTION To examine the association between low-dose aspirin use and risk of colorectal cancer (CRC). METHODS In this nationwide cohort study, we identified individuals aged 50 years or older residing for 6 months or more in Norway in 2004-2018 and obtained data from national registers on drug prescriptions, cancer occurrence, and sociodemographic factors. Multivariable Cox regression models were used to estimate the association between low-dose aspirin use and CRC risk. In addition, we calculated the number of CRC potentially averted by low-dose aspirin use. RESULTS We included 2,186,390 individuals. During the median follow-up of 10.9 years, 579,196 (26.5%) used low-dose aspirin, and 38,577 (1.8%) were diagnosed with CRC. Current use of aspirin vs never use was associated with lower CRC risk (hazard ratio [HR] 0.87, 95% confidence interval [CI] 0.84-0.90). The association was more pronounced for metastatic CRC (HR 0.79; 95% CI 0.74-0.84) than regionally advanced (HR 0.89; 95% CI 0.85-0.92) and localized CRC (HR 0.93; 95% CI 0.87-1.00; P heterogeneity = 0.001). A significant trend was found between duration of current use and CRC risk: HR 0.91 (95% CI 0.86-0.95) for <3 years, HR 0.85 (0.80-0.91) for ≥3 and <5 years, and HR 0.84 (0.80-0.88) for ≥5 years of use vs never use ( P trend < 0.001). For past use, HR were 0.89 (95% CI 0.84-0.94) for <3 years, 0.90 (0.83-0.99) for ≥3 and <5 years, and 0.98 (0.91-1.06) for ≥5 years since last use vs never use ( P -trend < 0.001). We estimated that aspirin use averted 1,073 cases of CRC (95% CI 818-1,338) in the study period. DISCUSSION In this nationwide cohort, use of low-dose aspirin was associated with a lower risk of CRC.
Collapse
Affiliation(s)
- Sara Nafisi
- Department of Research, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Nathalie C. Støer
- Department of Research, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
| | - Marit B. Veierød
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kristin R. Randel
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
| | - Geir Hoff
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
- Department of Research and Development, Telemark Hospital Trust, Skien, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lukas Löfling
- Department of Research, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
| | - Cristina Bosetti
- Department of Medical Epidemiology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Edoardo Botteri
- Department of Research, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, National Institute of Public Health, Oslo, Norway
| |
Collapse
|
9
|
Fryer E, Martin RM, Haycock P, Yarmolinsky J. Investigating the causal effect of previously reported therapeutic agents for colorectal cancer prevention: protocol for a Mendelian randomization analysis. Wellcome Open Res 2024; 9:30. [PMID: 38911899 PMCID: PMC11190651 DOI: 10.12688/wellcomeopenres.20861.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/10/2024] [Indexed: 06/25/2024] Open
Abstract
Background Colorectal cancer (CRC) is the third most common cancer worldwide, with 1.9 million new cases in 2020 and a predicted rise to 3.2 million in 2040. Screening programmes are already in place to aid early detection and secondary prevention of CRC, but the rising prevalence means additional approaches are required in both primary and secondary prevention settings. Preventive therapy, whereby natural or synthetic agents are used to prevent, reverse or delay disease development, could be an effective strategy to further reduce cancer risk and potential agents have already been identified in conventional observational studies. However, as such studies are vulnerable to confounding and reverse causation, we aim to evaluate these observed relationships using Mendelian randomization (MR), an alternative causal inference approach which should be less susceptible to these biases. Methods and analysis We will use two-sample MR, which uses two independent samples for the exposure and outcome data, to investigate previously reported observational associations of multiple potential preventive agents with CRC risk. We define preventive agents as any synthetic (e.g. approved medication) or natural (e.g. micronutrient, endogenous hormone) molecule used to reduce the risk of cancer. We will first extract potential preventive agents that have been previously linked to CRC risk in observational studies from reviews of the literature. We will then evaluate whether we can develop a genetic instrument for each preventive agent from previously published genome-wide association studies (GWASs) of direct measures of molecular traits (e.g. circulating levels of protein drug targets, blood-based biomarkers of dietary vitamins). The summary statistics from these GWASs, and a large GWAS of CRC, will be used in two-sample MR analyses to investigate the causal effect of putative preventive therapy agents on CRC risk. Sensitivity analyses will be conducted to evaluate the robustness of findings to potential violations of MR assumptions.
Collapse
Affiliation(s)
- Ella Fryer
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, England, BS8 2BN, UK
- Population Health Sciences, University of Bristol, Bristol, England, BS8 2BN, UK
| | - Richard M. Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, England, BS8 2BN, UK
- Population Health Sciences, University of Bristol, Bristol, England, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust, University of Bristol, Bristol, England, BS8 2BN, UK
| | - Philip Haycock
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, England, BS8 2BN, UK
- Population Health Sciences, University of Bristol, Bristol, England, BS8 2BN, UK
| | - James Yarmolinsky
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, England, W2 1PG, UK
| |
Collapse
|
10
|
Ghule P, Panic J, Malone DC. Risk of bleeding with concomitant use of oral anticoagulants and aspirin: A systematic review and meta-analysis. Am J Health Syst Pharm 2024; 81:494-508. [PMID: 38263263 DOI: 10.1093/ajhp/zxae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Indexed: 01/25/2024] Open
Abstract
PURPOSE Oral anticoagulants (OACs) and aspirin can trigger bleeding events when used alone or in combination. The purpose of this study was to compare the risk of any type of bleeding in individuals exposed to a combination of OAC and aspirin with the risk in those taking an OAC or aspirin alone. METHODS MEDLINE and Web of Science were queried in January 2021 for eligible articles. Studies were included if they were either randomized controlled trials (RCTs) or observational studies and evaluated the number of any bleeding events in two groups, one with exposure to both OAC and aspirin and one with exposure to OAC alone or aspirin alone. Pooled odds ratios were calculated using a random-effects model. RESULTS Forty-two studies were included. In an analysis of 15 RCTs and 19 observational studies evaluating OAC plus aspirin versus OAC alone, a significant difference in the risk of bleeding was observed in the combination groups, with an odds ratio [OR] of, 1.36 (95% CI, 1.15-1.59) for RCTs and an OR of 1.42 (95% CI-, 1.09-1.87) for observational studies. When OAC plus aspirin was compared to aspirin alone, a higher rate of bleeding was found in the combination group (OR, 2.36; 95%CI, 1.91-2.92) in the analysis of 15 RCTs, but no significant difference was found among 10 observational studies (OR, 1.93; 95% Cl, 0.99-3.75). CONCLUSION The risk of any type of bleeding was significantly increased among patients taking aspirin plus OAC compared to those taking OAC alone in both RCTs and observational studies. Evaluation of RCTs comparing OAC plus aspirin to aspirin alone suggests increased bleeding risk as well.
Collapse
Affiliation(s)
- Priyanka Ghule
- College of Pharmacy, University of Utah, Salt Lake City, UH, USA
| | - Jennifer Panic
- Froedtert and the Medical College of Wisconsin, Milwaukee, WI, USA
| | - Daniel C Malone
- College of Pharmacy, University of Utah, Salt Lake City, UH, USA
| |
Collapse
|
11
|
Lee KC, Chung KC, Chen HH, Cheng KC, Wu KL, Song LC. Potential beneficial effects of long-term aspirin use on the prevalence of colorectal cancer: a population-based study of the US Nationwide Inpatient Sample. Cancer Causes Control 2024; 35:477-486. [PMID: 37855925 DOI: 10.1007/s10552-023-01803-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/14/2023] [Indexed: 10/20/2023]
Abstract
PURPOSE Whether long-term aspirin usage is associated with colorectal cancer (CRC) risk needs more evidence. The study evaluated the association between long-term aspirin use and prevalence of CRC in a large, nationally representative database. METHODS Hospitalized patients aged ≥ 50 years during 2018 were identified in the United States (US) National Inpatient Sample (NIS). Patients without complete information of age, sex, race, income, and insurance status were excluded, as well as those with inflammatory bowel disease (IBD) or malignancies other than CRC. Propensity score matching (PSM) was applied to balance the characteristics between patients with and without long-term aspirin use. Logistic regressions were performed to determine the relationship between long-term aspirin use and the presence of CRC. CRC and aspirin use were identified through the administrative International Classification of Diseases (ICD) codes. RESULTS Data from 3,490,226 patients were included, in which 688,018 (19.7%) had a record of long-term aspirin use. After 1:1 PSM, there remained 1,376,006 patients, representing 6,880,029 individuals in the US after weighting. After adjusting for confounders, long-term aspirin use was significantly associated with lower CRC odds (adjusted odds ratio [aOR] = 0.64, 95% confidence interval [CI] 0.62, 0.67). This association was not changed when stratified by age, sex, race, body mass index (BMI), and smoking. CONCLUSIONS From a national inpatient dataset, US adults ≥ 50 years on long-term aspirin are less likely to have CRC, regardless of age, sex, race, BMI, and smoking status.
Collapse
Affiliation(s)
- Ko-Chao Lee
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuan-Chih Chung
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| | - Hong-Hwa Chen
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kung-Chuan Cheng
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Kuen-Lin Wu
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ling-Chiao Song
- Division of Colon & Rectal Surgery, Department of Surgery, E-DA Hospital, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
12
|
Pluchart H, Chanoine S, Moro-Sibilot D, Chouaid C, Frey G, Villa J, Degano B, Giaj Levra M, Bedouch P, Toffart AC. Lung cancer, comorbidities, and medication: the infernal trio. Front Pharmacol 2024; 14:1016976. [PMID: 38450055 PMCID: PMC10916800 DOI: 10.3389/fphar.2023.1016976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/25/2023] [Indexed: 03/08/2024] Open
Abstract
Most patients with lung cancer are smokers and are of advanced age. They are therefore at high risk of having age- and lifestyle-related comorbidities. These comorbidities are subject to treatment or even polypharmacy. There is growing evidence of a link between lung cancer, comorbidities and medications. The relationships between these entities are complex. The presence of comorbidities and their treatments influence the time of cancer diagnosis, as well as the diagnostic and treatment strategy. On the other hand, cancer treatment may have an impact on the patient's comorbidities such as renal failure, pneumonitis or endocrinopathies. This review highlights how some comorbidities may have an impact on lung cancer presentation and may require treatment adjustments. Reciprocal influences between the treatment of comorbidities and anticancer therapy will also be discussed.
Collapse
Affiliation(s)
- Hélène Pluchart
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, Grenoble INP, TIMC UMR5525, Grenoble, France
| | - Sébastien Chanoine
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
| | - Denis Moro-Sibilot
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Christos Chouaid
- Service de Pneumologie, Centre Hospitalier Intercommunal de Créteil, Créteil, France
- Inserm U955, UPEC, IMRB, équipe CEpiA, CréteilFrance
| | - Gil Frey
- Service de Chirurgie Thoracique, Vasculaire et Endocrinienne, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Julie Villa
- Service de Radiothérapie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Bruno Degano
- Université Grenoble Alpes, Grenoble, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
- Laboratoire HP2, INSERM U1042, Université Grenoble Alpes, Grenoble, France
| | - Matteo Giaj Levra
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Pierrick Bedouch
- Pôle Pharmacie, Centre Hospitalier Universitaire Grenoble Alpes, La Tronche, France
- Université Grenoble Alpes, Grenoble, France
- Université Grenoble Alpes, CNRS, Grenoble INP, TIMC UMR5525, Grenoble, France
| | - Anne-Claire Toffart
- Université Grenoble Alpes, Grenoble, France
- Institut pour l’Avancée des Biosciences, UGA/INSERM U1209/CNRS 5309, Université Grenoble Alpes, La Tronche, France
- Service Hospitalier Universitaire de Pneumologie Physiologie, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| |
Collapse
|
13
|
Kumar P, Sheokand D, Grewal A, Saini V, Kumar A. Clinical side-effects based drug repositioning for anti-epileptic activity. J Biomol Struct Dyn 2024; 42:1443-1454. [PMID: 37042987 DOI: 10.1080/07391102.2023.2199874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 04/01/2023] [Indexed: 04/13/2023]
Abstract
Several generations of anti-epileptic drugs (AEDs) are available but have several associated side effects apart from a limited success rate. Drug repositioning strategies have gained importance in the last two decades owing to lower failure rates and economic burden. Drugs with similar side effect profiles may share a common mechanism of action and thus can be linked to other disease treatments. The present study was carried out to identify the newly approved drug candidate(s) as AEDs using clinical side-effects drug repositioning strategy. The clinical side effect similarity of drugs available in the SIDER v4.1 database was estimated against common side effects of 5 major marketed AEDs, using the 'dplyr' package library in the R. Further drugs were filtered based on Blood Brain Barrier permeability prediction and FDA-approval status. Molecular docking studies were performed for selected 26 hits (drugs) against previously identified epilepsy target receptors: Voltage-gated sodium channel α2 (Nav1.2), GABA receptor α1-β1 (GABAr α1-β1), and Voltage-gated calcium channel α-1 G (Cav3.1). Only 2 drugs (Ziprasidone and Paroxetine) showed better binding affinities against studied epilepsy receptors Nav1.2, GABAr α1-β1, and Cav3.1, than their corresponding standard AEDs, i.e. Carbamazepine, Clonazepam, and Pregabalin, respectively. Ziprasidone reportedly showed seizure-like symptoms in ∼3% of patients and was hence omitted from further study. The MDS study of docked complexes of Paroxetine with selected epilepsy target receptors showed stable RMSD values and better interaction energies. The study reveals Paroxetine as a potential candidate to be repurposed for 1st line epileptic seizure medication.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Pawan Kumar
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Deepak Sheokand
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Annu Grewal
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Vandana Saini
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| | - Ajit Kumar
- Toxicology and Computational Biology Group, Centre for Bioinformatics, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
14
|
Hamoya T, Tomono S, Miyamoto S, Fujii G, Wakabayashi K, Mutoh M. Theoretical basis validation and oxidative stress markers for cancer prevention clinical trials of aspirin. Sci Rep 2023; 13:21883. [PMID: 38072949 PMCID: PMC10711014 DOI: 10.1038/s41598-023-49254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Aspirin, a nonsteroidal anti-inflammatory drug, has been proven effective in a clinical trial of carcinogenesis blockade. However, various modes of action have been reported for these effects. Thus, in this study, we aimed to present reasonable mode of actions as a proof of concept for human trials, especially trials for patients with familial adenomatous polyposis (FAP). Aspirin treatment at 1000 ppm inhibited intestinal tumorigenesis in FAP model Min mice. As a mode of action, aspirin regulated β-catenin signaling, inflammation, and oxidative stress both in vivo and in vitro. Furthermore, we examined novel markers predictive of aspirin treatment based on liquid biopsy. Here, we demonstrated that aspirin reduced the levels of reactive carbonyl species in the serum of Min mice. These data are expected to be of use for proof of concept of aspirin human trials and implied for the prediction of aspirin efficacy.
Collapse
Affiliation(s)
- Takahiro Hamoya
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Susumu Tomono
- Department of Microbiology and Immunology, Aichi Medical University, Nagakute, Aichi, 480-1195, Japan
| | - Shingo Miyamoto
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Gen Fujii
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Keiji Wakabayashi
- Graduate Division of Nutritional and Environmental Sciences, University of Shizuoka, Shizuoka, 422-8526, Japan
| | - Michihiro Mutoh
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kyoto, Japan.
- Epidemiology and Prevention Division, Center for Public Health Sciences, National Cancer Center, Tokyo, 104-0045, Japan.
| |
Collapse
|
15
|
Gill I, Shams C, Hanna A, George J, Jamil LH, Patel A. Role of demographics in noninvasive testing for colorectal cancer screening: do targeted cut-off values improve detection? Minerva Gastroenterol (Torino) 2023; 69:459-469. [PMID: 35343664 DOI: 10.23736/s2724-5985.22.03124-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fecal immunochemical test (FIT) is a yearly alternative colorectal screening modality for average risk individuals unwilling or unable to undergo invasive colorectal cancer (CRC) screening due to cost and accessibility. This study aimed to determine whether FIT should be interpreted within the context of patient demographics and medical history. METHODS Patients >50 years old who had a FIT followed by colonoscopy within 1 year were analyzed based on age, race, BMI, social and medical comorbidities. False positive (FP) and false negative (FN) FIT results within each patient demographic and medical history variable were determined by comparing with the gold standard of colonoscopy using χ2 analysis. RESULTS One thousand twenty-five patients were reviewed. 21.8% of FIT results were positive. Factors which differed in positive FIT rates were age (P=0.003), smoking (P<0.001), alcohol (P=0.001), and hypertension (P<0.001). The difference in rates of FP and FN FIT outcomes among each variable underwent further subanalysis. The FP was 66.8% and the FN rate was 12.8%. Higher FN outcomes were noted in those above 70, males and smokers, though the result was only statistically significant for males (P=0.009). Females were observed to have higher FP rates (P=0.019). CONCLUSIONS Females had higher FP FIT rates compared to males, indicating that sex may influence FIT outcomes and should be accounted for when interpreting FIT results. This information can be utilized to identify populations at higher risk of FP or FN FIT results to target CRC screening. Additionally, recalculating the FP and FN rates for each variable may help determine new FIT targets.
Collapse
Affiliation(s)
- Inayat Gill
- Department of Internal Medicine, Beaumont Health, Royal Oak, MI, USA -
| | - Christienne Shams
- Department of Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Angy Hanna
- Section of Gastroenterology and Hepatology, Department of Internal Medicine, Detroit Medical Center, Detroit, MI, USA
| | - Julie George
- Section of Biostatistics, Department of Beaumont Research Institute, Royal Oak, MI, USA
| | - Laith H Jamil
- Section of Gastroenterology and Hepatology, Department of Internal Medicine, Beaumont Health, Royal Oak, MI, USA
| | - Atulkumar Patel
- Section of Gastroenterology and Hepatology, Department of Internal Medicine, Beaumont Health, Royal Oak, MI, USA
| |
Collapse
|
16
|
Goswami S, Zhang Q, Celik CE, Reich EM, Yilmaz ÖH. Dietary fat and lipid metabolism in the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2023; 1878:188984. [PMID: 37722512 PMCID: PMC10937091 DOI: 10.1016/j.bbcan.2023.188984] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/20/2023]
Abstract
Metabolic reprogramming has been considered a core hallmark of cancer, in which excessive accumulation of lipids promote cancer initiation, progression and metastasis. Lipid metabolism often includes the digestion and absorption of dietary fat, and the ways in which cancer cells utilize lipids are often influenced by the complex interactions within the tumor microenvironment. Among multiple cancer risk factors, obesity has a positive association with multiple cancer types, while diets like calorie restriction and fasting improve health and delay cancer. Impact of these diets on tumorigenesis or cancer prevention are generally studied on cancer cells, despite heterogeneity of the tumor microenvironment. Cancer cells regularly interact with these heterogeneous microenvironmental components, including immune and stromal cells, to promote cancer progression and metastasis, and there is an intricate metabolic crosstalk between these compartments. Here, we focus on discussing fat metabolism and response to dietary fat in the tumor microenvironment, focusing on both immune and stromal components and shedding light on therapeutic strategies surrounding lipid metabolic and signaling pathways.
Collapse
Affiliation(s)
- Swagata Goswami
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Qiming Zhang
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Cigdem Elif Celik
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Hacettepe Univ, Canc Inst, Department Basic Oncol, Ankara TR-06100, Turkiye
| | - Ethan M Reich
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ömer H Yilmaz
- Department of Biology, The David H. Koch Institute for Integrative Cancer Research at MIT, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pathology, Massachusetts General Hospital and Beth Israel Deaconness Medical Center and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Mohamed-Ezzat RA, Kariuki BM, Srour AM. Synthesis, crystal structure and in vitro anti-proliferative activity of 2-[(4-acetyl-phen-yl)carbamo-yl]phenyl acetate. Acta Crystallogr E Crystallogr Commun 2023; 79:999-1002. [PMID: 37936857 PMCID: PMC10626967 DOI: 10.1107/s2056989023008526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 09/27/2023] [Indexed: 11/09/2023]
Abstract
2-[(4-Acetyl-phen-yl)carbamo-yl]phenyl acetate, C17H15NO4, has been synthesized and structurally characterized. In the structure, N-H⋯O hydrogen-bonding inter-actions form chains of mol-ecules aligned along the [101] direction. The chains are linked by π-π and C-H⋯π inter-actions, forming a three dimensional network. The compound has been screened for in vitro anti-proliferative activity revealing considerable activity.
Collapse
Affiliation(s)
- Reham A. Mohamed-Ezzat
- Chemistry of Natural & Microbial Products Department, National Research Centre, Cairo, Egypt
| | - Benson M. Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff CF10, 3AT, United Kingdom
| | - Aladdin M. Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
18
|
Bakierzynska M, Cullinane MC, Redmond HP, Corrigan M. Prophylactic aspirin intake and breast cancer risk; A systematic review and meta-analysis of observational cohort studies. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2023; 49:106940. [PMID: 37321932 DOI: 10.1016/j.ejso.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 05/15/2023] [Accepted: 05/20/2023] [Indexed: 06/17/2023]
Abstract
Breast Cancer (BC) is the most common cancer amongst women. The chemo-preventative effects of aspirin on breast cancer have been demonstrated in several longitudinal studies however previous meta-analysis have shown inconsistent results. This study aimed to assess the relationship between aspirin use and BC risk, and to determine if there is a dose-response relationship between aspirin and BC risk. Studies incorporating BC risk with aspirin use published within the last twenty years were included. The study report is based on the guidelines of Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) and Meta-Analysis of Observational Studies in Epidemiology. Twenty-eight cohort studies that reported BC incidence during a follow up of 4.4-32 years were included. Compared to non-users, aspirin users had a reduced risk of BC (HR = 0.91, c.i 0.81-0.97, p = 0.002). There was no obvious association between BC risk reduction and aspirin dose (HR = 0.94, c.i 0.85-1.04) or duration (HR = 0.86, c.i 0.71-1.03). Frequency, however, was associated with a reduced risk of BC (HR = 0.90, c.i 0.82-0.98). A risk reduction was observed in oestrogen receptor (ER) positive tumours (HR = 0.90, c.i 0.86-0.96, p = 0.0004) while no relationship was observed with ER negative tumours (HR = 0.94, c.i 0.85-1.05). This meta-analysis found an association between aspirin intake and BC risk reduction. A more favourable outcome was noted with ingestion of greater than 6 tablets of aspirin per week. Aspirin had a significant risk reduction in patients with ER positive tumours compared to ER negative BC.
Collapse
Affiliation(s)
| | | | - Henry Paul Redmond
- Department of Surgery, Cork University Hospital, University College Cork, Cork, Ireland
| | - Mark Corrigan
- Department of Surgery, Cork University Hospital, University College Cork, Cork, Ireland
| |
Collapse
|
19
|
Nafisi S, Randel KR, Støer NC, Veierød MB, Hoff G, Holme Ø, Schult AL, Botteri E. Association between use of low-dose aspirin and detection of colorectal polyps and cancer in a screening setting. Dig Liver Dis 2023; 55:1126-1132. [PMID: 36754644 DOI: 10.1016/j.dld.2023.01.156] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/16/2023] [Accepted: 01/23/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND The possible protective effect of aspirin on risk of colorectal cancer (CRC) is still highly debated. METHODS We used data from Bowel Cancer Screening in Norway, a trial randomizing individuals from general population, aged 50-74 years, to flexible sigmoidoscopy or faecal immunochemical test (FIT), to study the association between aspirin use and detection of CRC and two CRC precursors: adenomas and advanced serrated lesions (ASL). Prescriptions of low-dose aspirin were obtained from Norwegian prescription database. Logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Among 64,889 screening participants (24,159 sigmoidoscopy, 40,730 FIT), 314 (0.5%) had CRC, 6,208 (9.6%) adenoma and 659 (1.0%) ASL. Overall and short-term use (<3 years) of low-dose aspirin, versus no use, were not associated with any colorectal lesion. Long-term use (≥3 years) was associated with lower detection of CRC (overall OR 0.66, 95%CI 0.46-0.93; sigmoidoscopy: 0.56, 0.33-0.97; FIT: 0.72, 0.45-1.15), adenomas in sigmoidoscopy arm (overall OR 0.95, 95%CI 0.87-1.03; sigmoidoscopy: 0.89, 0.80-0.99; FIT: 1.03, 0.89-1.18), but not ASLs. We did not observe significant differences in the effect of aspirin according to the location of colorectal lesions. CONCLUSION Our results suggest that long-term use of aspirin might have a protective effect against adenomas and colorectal cancer, but not ASLs.
Collapse
Affiliation(s)
- Sara Nafisi
- Department of Research, Cancer Registry of Norway, Oslo, Norway; Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Kristin R Randel
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway
| | - Nathalie C Støer
- Department of Research, Cancer Registry of Norway, Oslo, Norway; Norwegian Research Centre for Women's Health, Women's Clinic, Oslo University Hospital, Oslo, Norway
| | - Marit B Veierød
- Oslo Centre for Biostatistics and Epidemiology, Department of Biostatistics, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Geir Hoff
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway; Department of Research and Development, Telemark Hospital Trust, Skien, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Øyvind Holme
- Department of Medicine, Sørlandet Hospital, Kristiansand, Norway; Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Anna L Schult
- Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway; Department of Medicine, Vestre Viken Hospital Trust Bærum, Gjettum, Norway
| | - Edoardo Botteri
- Department of Research, Cancer Registry of Norway, Oslo, Norway; Section for Colorectal Cancer Screening, Cancer Registry of Norway, Oslo, Norway.
| |
Collapse
|
20
|
Lloyd KE, Hall LH, Ziegler L, Foy R, Borthwick GM, MacKenzie M, Taylor DG, Smith SG. GPs' willingness to prescribe aspirin for cancer preventive therapy in Lynch syndrome: a factorial randomised trial investigating factors influencing decisions. Br J Gen Pract 2023; 73:e302-e309. [PMID: 36997217 PMCID: PMC9997655 DOI: 10.3399/bjgp.2021.0610] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 09/27/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The National Institute for Health and Care Excellence (NICE) 2020 guidelines recommends aspirin for colorectal cancer prevention for people with Lynch syndrome. Strategies to change practice should be informed by understanding the factors influencing prescribing. AIM To investigate the optimal type and level of information to communicate with GPs to increase willingness to prescribe aspirin. DESIGN AND SETTING GPs in England and Wales (n = 672) were recruited to participate in an online survey with a 23 factorial design. GPs were randomised to one of eight vignettes describing a hypothetical patient with Lynch syndrome recommended to take aspirin by a clinical geneticist. METHOD Across the vignettes, the presence or absence of three types of information was manipulated: 1) existence of NICE guidance; 2) results from the CAPP2 trial; 3) information comparing risks/benefits of aspirin. The main effects and all interactions on the primary (willingness to prescribe) and secondary outcomes (comfort discussing aspirin) were estimated. RESULTS There were no statistically significant main effects or interactions of the three information components on willingness to prescribe aspirin or comfort discussing harms and benefits. In total, 80.4% (540/672) of GPs were willing to prescribe, with 19.7% (132/672) unwilling. GPs with prior awareness of aspirin for preventive therapy were more comfortable discussing the medication than those unaware (P = 0.031). CONCLUSION It is unlikely that providing information on clinical guidance, trial results, and information comparing benefits and harms will increase aspirin prescribing for Lynch syndrome in primary care. Alternative multilevel strategies to support informed prescribing may be warranted.
Collapse
Affiliation(s)
- Kelly E Lloyd
- Leeds Institute of Health Sciences, University of Leeds, Leeds
| | - Louise H Hall
- Leeds Institute of Health Sciences, University of Leeds, Leeds
| | - Lucy Ziegler
- Leeds Institute of Health Sciences, University of Leeds, Leeds
| | - Robbie Foy
- Leeds Institute of Health Sciences, University of Leeds, Leeds
| | - Gillian M Borthwick
- Translational and Clinical Research Institute, Newcastle University, Newcastle
| | | | | | - Samuel G Smith
- Leeds Institute of Health Sciences, University of Leeds, Leeds
| |
Collapse
|
21
|
Babu SSN, Singla S, Jena G. Role of Combination Treatment of Aspirin and Zinc in DMH-DSS-induced Colon Inflammation, Oxidative Stress and Tumour Progression in Male BALB/c Mice. Biol Trace Elem Res 2023; 201:1327-1343. [PMID: 35438409 DOI: 10.1007/s12011-022-03241-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/09/2022] [Indexed: 02/07/2023]
Abstract
Colitis-associated colorectal cancer serves as a prototype of inflammation-associated cancers which is linked with repeated cycles of inflammation and DNA repair deficits. Several preclinical and clinical data reported that aspirin has a chemo-preventive effect in colorectal cancer and is associated with dose-dependent side effects. Furthermore, it has been reported that zinc supplementation improves the quality of life in patients undergoing chemotherapy by alteration of colonic cancer cell gene expression. However, explication of the detailed molecular mechanisms involved in the combined administration of aspirin and zinc-mediated protection against colitis-associated colorectal cancer deserves further investigation. For the induction of colitis-associated colorectal cancer, male BALB/c mice were administered 1,2-dimethylhydrazine dihydrochloride (DMH) 20 mg/kg/bw thrice before the initiation of every DSS cycle (3%w/v in drinking water). One week after the initiation of DSS treatment, aspirin (40 mg/kg; p.o.) and zinc in the form of zinc sulphate (3 mg/kg; p.o.) were administered for 8 weeks. Combination of aspirin and zinc as intervention significantly ameliorated DAI score, myeloperoxidase activity, histological score, apoptotic cells and protein expression of various inflammatory markers including nuclear factor kappa light chain enhancer of activated B cells (NFκBp65), cycloxygenase-2 (COX-2) and interleukin-6 (IL-6); proliferation markers such as proliferating cell nuclear antigen (PCNA), signal transducer and activator of transcription 3 (STAT3) expression significantly decreased, and antioxidant enzymes nuclear factor erythroid 2-related factor 2 (Nrf-2), metallothionein, catalase and superoxide dismutase (SOD) significantly increased as evaluated by immunohistochemistry and western blot analysis.
Collapse
Affiliation(s)
- Singothu Siva Nagendra Babu
- Facility for Risk Assessment and Intervention Studies, Dept. of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India, 160062
| | - Shivani Singla
- Facility for Risk Assessment and Intervention Studies, Dept. of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India, 160062
| | - Gopabandhu Jena
- Facility for Risk Assessment and Intervention Studies, Dept. of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Punjab, India, 160062.
| |
Collapse
|
22
|
Khoziainova S, Rozenberg G, Levy M. Ketogenic Diet and Beta-Hydroxybutyrate in Colorectal Cancer. DNA Cell Biol 2022; 41:1007-1011. [PMID: 36454261 PMCID: PMC10162116 DOI: 10.1089/dna.2022.0486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 12/05/2022] Open
Abstract
Colorectal cancer (CRC) is one of the leading causes of cancer-related death in the United States. Although certain genetic predispositions may contribute to one's risk for developing CRC, dietary and lifestyle factors may play an important role as well. In a recent study in Nature, Dmitrieva-Posocco et al, reveal a potential protective role of the ketogenic diet in colorectal cancer growth and progression. Administration of a ketogenic diet to CRC-bearing mice demonstrated a tumor-suppressive effect. Specifically, the ketone body β-hydroxybutyrate (BHB) exhibited the ability to suppress epithelial cell proliferation and inhibit tumor growth. BHB acts on cancer cells through regulation of homeodomain-only protein Hopx, known regulator of CRC. Furthermore, BHB requires a surface receptor Hcar to induce Hopx expression and suppress proliferation of intestinal epithelial cells. Taken together, these results describe a new therapeutic approach of using dietary intervention for the prevention and treatment of colorectal cancer.
Collapse
Affiliation(s)
- Svetlana Khoziainova
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Galina Rozenberg
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maayan Levy
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
23
|
Chung KC, Juang SE, Chen HH, Cheng KC, Wu KL, Song LC, Lee KC. Association between metabolic syndrome and colorectal cancer incidence and all-cause mortality: a hospital-based observational study. BMC Gastroenterol 2022; 22:453. [DOI: 10.1186/s12876-022-02505-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 09/15/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Background
Metabolic syndrome (MetS) is a worldwide pandemic and complex disorder associated with colorectal cancer (CRC). This study aims to identify the influence of number of MetS components on CRC incidence and mortality, using a national, longitudinal dataset of hospital care in Taiwan.
Methods
Patient data from the Taiwan National Health Insurance Research Database (NHIRD) from 2001 to 2008 were extracted. Individuals with at least one inpatient diagnosis or 2 outpatient visits with any MetS component found within one year were identified and included. Subjects died within 12 months after the presence of MetS components or had any prior cancer were excluded. The study cohort were then divided into two groups: subjects who had more (i.e., 3 to 4) MetS components and those who had fewer (i.e., 1 to 2) MetS components. An 2:1 propensity score (PS) matching were performed to balance the baseline characteristics between the groups. Cox regression analyses were conducted to compare the CRC incidence and all-cause mortality at follow-up between subjects with more MetS components versus fewer components.
Results
After matching, a total of 119,843 subjects (78,274 with 1–2 and 41,569 with 3–4 MetS components) were analyzed. After adjusting for confounders, subjects with 3–4 MetS components had a significantly higher risk of CRC [adjusted hazard ratio (aHR), 1.28; 95% confidence interval (CI), 1.15–1.43, p < 0.001) and all-cause mortality (aHR, 1.13; 95% CI, 1.08–1.17, p < 0.001) than those with only 1–2 MetS components. In stratified analyses, the greatest increased risk of CRC incidence that 3–4 MetS components posed as compared to 1–2 MetS components was seen in subjects without CHD history (aHR, 1.41, 95% CI, 1.23–1.62, p < 0.001). In addition, 3–4 MetS components (vs. 1–2) led to greater all-cause mortality among the subjects < 65y, both genders, with or without CHD, subjects without CKD hisotry, both aspirin users and non-users, users of nonsteroidal anti-inflammatory drugs (NSAIDs), and users of statin.
Conclusion
Compared with 1–2 components, subjects with 3–4 MetS components are at greater risk of CRC and death at follow-up. This study also demonstrates the risks for CRC and all-cause mortality in certain subgroups of individuals with 3–4 MetS components compared to 1–2 components. These findings may help clinicians on the CRC risk stratification according to individuals’ characteristics, as well as to optimize the strategy of MetS surveillance and control in order to prevent CRC.
Collapse
|
24
|
Yu YC, Paragomi P, Jin A, Wang R, Schoen RE, Koh WP, Yuan JM, Luu HN. Dietary Nonstarch Polysaccharide Intake and Risk of Colorectal Cancer: Findings from the Singapore Chinese Health Study. CANCER RESEARCH COMMUNICATIONS 2022; 2:1304-1311. [PMID: 36425215 PMCID: PMC9683694 DOI: 10.1158/2767-9764.crc-22-0153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/13/2022] [Accepted: 07/06/2022] [Indexed: 11/16/2022]
Abstract
Dietary fiber or non-starch polysaccharides (NSP) may provide protection from CRC development. Epidemiologic studies on the association between dietary fiber and CRC is inconsistent are limited on NSP as a modifiable risk factor. Using the Singapore Chinese Health Study, a population-based prospective cohort of 61,321 cancer-free middle-aged or older Chinese Singaporeans, we examined the association between dietary fiber and NSP intakes and CRC risk. Fiber and NSP intakes at baseline were obtained using a validated semi-quantitative food frequency questionnaire coupled with the Singapore Food Composition Database. Cox proportional hazard regression model was used to estimate the hazard ratios (HRs) and respective 95% confidence intervals (CIs) for CRC associated with dietary fiber and NSP intakes after adjusting for potential confounders. After an average of 17.5 years of follow-up, 2,140 participants developed CRC. NSP was inversely associated with the risk of CRC in a dose-dependent manner whereas dietary fiber was not associated with risk of CRC overall or histologic subtypes. The multivariable-adjusted HRs (95% CIs) of CRC for quartiles 2, 3 and 4 of dietary NSP intake were 0.99 (0.88-1.11), 0.98 (0.87-1.11) and 0.84 (0.73-0.95), respectively, compared with the lowest quartile (P trend =0.006). This inverse association was more apparent for colon cancer (HRQ4 vs. Q1=0.79, 95% CI: 0.67-0.93, P trend =0.003) than rectal cancer (HR Q4 vs. Q1=0.92, 95% CI: 0.74-1.13, P trend =0.53). Our findings suggested that dietary NSP but not fiber is associated with a reduced risk of colon cancer in Chinese Singaporeans. Significance Non-starch polysaccharides may be beneficial for colorectal cancer primary prevention.
Collapse
Affiliation(s)
- Yi-Chuan Yu
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Pedram Paragomi
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Aizhen Jin
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Renwei Wang
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Robert E. Schoen
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore
| | - Jian-Min Yuan
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Hung N. Luu
- University of Pittsburgh Medical Center (UPMC) Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
25
|
Minematsu H, Afify SM, Sugihara Y, Hassan G, Zahra MH, Seno A, Adachi M, Seno M. Cancer stem cells induced by chronic stimulation with prostaglandin E2 exhibited constitutively activated PI3K axis. Sci Rep 2022; 12:15628. [PMID: 36115905 PMCID: PMC9482612 DOI: 10.1038/s41598-022-19265-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 08/26/2022] [Indexed: 12/04/2022] Open
Abstract
Previously, our group has demonstrated establishment of Cancer Stem Cell (CSC) models from stem cells in the presence of conditioned medium of cancer cell lines. In this study, we tried to identify the factors responsible for the induction of CSCs. Since we found the lipid composition could be traced to arachidonic acid cascade in the CSC model, we assessed prostaglandin E2 (PGE2) as a candidate for the ability to induce CSCs from induced pluripotent stem cells (iPSCs). Mouse iPSCs acquired the characteristics of CSCs in the presence of 10 ng/mL of PGE2 after 4 weeks. Since constitutive Akt activation and pik3cg overexpression were found in the resultant CSCs, of which growth was found independent of PGE2, chronic stimulation of the receptors EP-2/4 by PGE2 was supposed to induce CSCs from iPSCs through epigenetic effect. The bioinformatics analysis of the next generation sequence data of the obtained CSCs proposed not only receptor tyrosine kinase activation by growth factors but also extracellular matrix and focal adhesion enhanced PI3K pathway. Collectively, chronic stimulation of stem cells with PGE2 was implied responsible for cancer initiation enhancing PI3K/Akt axis.
Collapse
|
26
|
Kanter MH, Schottinger JE, Joshua A, Slezak JM. Beyond Screening: An Interim Report and Analysis of a Multimodal Initiative to Decrease Colon Cancer Mortality. Jt Comm J Qual Patient Saf 2022; 48:388-394. [PMID: 35902142 DOI: 10.1016/j.jcjq.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Colon cancer, a potentially preventable and curable cancer, remains one of the leading causes of cancer-related death in the United States. In 2010 the researchers undertook a multifaceted initiative to reduce colon cancer mortality rates by 50% over 10 years. METHODS A baseline literature review of preventable causes of colon cancer mortality and review of 50 deaths from colon cancer in one institution identified a set of care process improvements that could be implemented to decrease colon cancer mortality. In 2017 a second mortality review identified a second set of care process improvements that were subsequently implemented. Compliance with these processes was monitored along with age and gender-adjusted mortality rates. RESULTS Identified care process improvements included improving the follow-up of patients with rectal bleeding and presumed iron deficiency anemia and improving the reliability of postsurgical surveillance for cancer recurrence, decreasing elapsed time from surgery to chemotherapy, increasing surgical referrals for patients with advanced colon cancer, increasing the upper age limit and overall rate of colon cancer screening, increasing vitamin D and aspirin use, and monitoring and increasing the adenoma detection rate. Compliance with these processes improved for most measures, including screening (73.7% to 79.9%), adenoma detection rates on screening colonoscopy (30% to 36% for women and 42% to 49% for men), and chemotherapy within 35 days of surgery for colon cancer (39.0% to 51.9%). Age- and gender-adjusted mortality decreased from 13.8 per 100,000 in 2009-2011 to 10.5 per 100,000 in 2016-2018. CONCLUSIONS This quality improvement program was feasible to implement, resulted in process improvements, and decreased colon cancer mortality over seven years.
Collapse
|
27
|
McDowell R, Perrott S, Murchie P, Cardwell C, Hughes C, Samuel L. Oral antibiotic use and early-onset colorectal cancer: findings from a case-control study using a national clinical database. Br J Cancer 2022; 126:957-967. [PMID: 34921228 PMCID: PMC8927122 DOI: 10.1038/s41416-021-01665-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/27/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Antibiotic-induced gut dysbiosis has been associated with colorectal cancer (CRC) in older adults. This study will investigate whether an association exists between antibiotic usage and early-onset colorectal cancer (CRC), and also evaluate this in later-onset CRC for comparison. METHODS A case-control study was conducted using primary care data from 1999-2011. Analysis were conducted separately in early-onset CRC cases (diagnosed < 50 years) and later-onset cases (diagnosed ≥ 50 years). Conditional logistic regression was used to calculate odds ratios and 95% confidence intervals (CI) for the associations between antibiotic exposure and CRC by tumour location, adjusting for comorbidities. RESULTS Seven thousands nine hundred and three CRC cases (445 aged <50 years) and 30,418 controls were identified. Antibiotic consumption was associated with colon cancer in both age-groups, particularly in the early-onset CRC cohort (<50 years: adjusted Odds Ratio (ORadj) 1.49 (95% CI 1.07, 2.07), p = 0·018; ≥50 years (ORadj (95% CI) 1.09 (1.01, 1.18), p = 0·029). Antibiotics were not associated with rectal cancer (<50 years: ORadj (95% CI) 1.17 (0.75, 1.84), p = 0.493; ≥50 years: ORadj (95% CI) 1.07 (0.96, 1.19), p = 0.238). CONCLUSION Our findings suggest antibiotics may have a role in colon tumour formation across all age-groups.
Collapse
Affiliation(s)
- Ronald McDowell
- Centre for Public Health, School of Medicine, Dentistry & Biomedical Science, Queen's University, Belfast, Northern Ireland
| | - Sarah Perrott
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, Scotland
| | - Peter Murchie
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, Scotland
| | - Christopher Cardwell
- Centre for Public Health, School of Medicine, Dentistry & Biomedical Science, Queen's University, Belfast, Northern Ireland
| | - Carmel Hughes
- School of Pharmacy, Queen's University, Belfast, Scotland
| | - Leslie Samuel
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, Scotland.
- Department of Clinical Oncology, Aberdeen Royal Infirmary, NHS Grampian, Aberdeen, Scotland.
| |
Collapse
|
28
|
Kaur R, McDonald C, Meiser B, Macrae F, Smith SK, Kang YJ, Caruana M, Mitchell G. The Risk-Reducing Effect of Aspirin in Lynch Syndrome Carriers: Development and Evaluation of an Educational Leaflet. ADVANCED GENETICS (HOBOKEN, N.J.) 2022; 3:2100046. [PMID: 36618023 PMCID: PMC9744515 DOI: 10.1002/ggn2.202100046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Indexed: 01/11/2023]
Abstract
Carriers of germline mutations in genes associated with Lynch syndrome are at increased risk for colorectal, endometrial, ovarian, and other cancers. There is evidence that daily consumption of aspirin may reduce cancer risk in these individuals. There is a need for educational resources to inform carriers of the risk-reducing effects of aspirin or to support decision-making. An educational leaflet describing the risks and benefits of using aspirin as risk-reducing medicine in carriers of Lynch-syndrome-related mutations is developed and pilot tested in 2017. Carriers are ascertained through a familial cancer clinic and surveyed using a mailed, self-administered questionnaire. The leaflet is highly rated for its content, clarity, length, relevance, and visual appeal by more than 70% of the participants. Most participants (91%) report "a lot" or "quite a bit" of improvement in perceived understanding in knowledge about who might benefit from taking aspirin, its benefits, how long to take it, the reduction in bowel cancer risk, and the optimal dosage. A few (14%) participants seek more information on the dosage of aspirin. This leaflet will be useful as an aid to facilitate discussion between patients and their health care professionals about the use of aspirin as a risk-reducing medication.
Collapse
Affiliation(s)
- Rajneesh Kaur
- Medical Education OfficeUNSW SydneyNew South WalesAustralia,Medical Education OfficeThe University of SydneyEdward Ford BuildingSydneyNew South Wales2006Australia
| | - Cassandra McDonald
- The Kinghorn Cancer CentreSt Vincent HospitalVictoria StreetDarlinghurstNew South Wales2010Australia
| | - Bettina Meiser
- Psychosocial Research GroupUNSW SydneyHigh StreetSydneyNew South Wales2052Australia
| | - Finlay Macrae
- Department of Colorectal Medicine and Geneticsand Department of MedicineThe Royal Melbourne HospitalUniversity of MelbourneParkvilleVictoria3010Australia
| | - Sian K Smith
- Psychosocial Research GroupUNSW SydneyHigh StreetSydneyNew South Wales2052Australia
| | - Yoon Jung Kang
- Daffodil CentreUniversity of SydneySydneyNew South Wales2006Australia
| | - Michael Caruana
- Daffodil CentreUniversity of SydneySydneyNew South Wales2006Australia
| | - Gillian Mitchell
- Familial Cancer CentrePeter MacCallum Cancer CentreParkvilleVictoria3010Australia,The Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoria3052Australia
| |
Collapse
|
29
|
Ionica E, Gaina G, Tica M, Chifiriuc MC, Gradisteanu-Pircalabioru G. Contribution of Epithelial and Gut Microbiome Inflammatory Biomarkers to the Improvement of Colorectal Cancer Patients' Stratification. Front Oncol 2022; 11:811486. [PMID: 35198435 PMCID: PMC8859258 DOI: 10.3389/fonc.2021.811486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
In order to ensure that primary endpoints of clinical studies are attained, the patients' stratification is an important aspect. Selection criteria include age, gender, and also specific biomarkers, such as inflammation scores. These criteria are not sufficient to achieve a straightforward selection, however, in case of multifactorial diseases, with unknown or partially identified mechanisms, occasionally including host factors, and the microbiome. In these cases, the efficacy of interventions is difficult to predict, and as a result, the selection of subjects is often random. Colorectal cancer (CRC) is a highly heterogeneous disease, with variable clinical features, outcomes, and response to therapy; the CRC onset and progress involves multiple sequential steps with accumulation of genetic alterations, namely, mutations, gene amplification, and epigenetic changes. The gut microbes, either eubiotic or dysbiotic, could influence the CRC evolution through a complex and versatile crosstalk with the intestinal and immune cells, permanently changing the tumor microenvironment. There have been significant advances in the development of personalized approaches for CRC screening, treatment, and potential prevention. Advances in molecular techniques bring new criteria for patients' stratification-mutational analysis at the time of diagnosis to guide treatment, for example. Gut microbiome has emerged as the main trigger of gut mucosal homeostasis. This may impact cancer susceptibility through maintenance of the epithelial/mucus barrier and production of protective metabolites, such as short-chain fatty acids (SCFAs) via interactions with the hosts' diet and metabolism. Microbiome dysbiosis leads to the enrichment of cancer-promoting bacterial populations, loss of protective populations or maintaining an inflammatory chronic state, all of which contribute to the development and progression of CRC. Meanwhile, variations in patient responses to anti-cancer immuno- and chemotherapies were also linked to inter-individual differences in intestine microbiomes. The authors aim to highlight the contribution of epithelial and gut microbiome inflammatory biomarkers in the improvement of CRC patients' stratification towards a personalized approach of early diagnosis and treatment.
Collapse
Affiliation(s)
- Elena Ionica
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Gisela Gaina
- Laboratory of Cell Biology, Neuroscience and Experimental Miology, Victor Babes National Institute of Pathology, Bucharest, Romania
| | - Mihaela Tica
- Bucharest Emergency University Hospital, Bucharest, Romania
| | - Mariana-Carmen Chifiriuc
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
- Biological Science Division, Romanian Academy of Sciences, Bucharest, Romania
| | | |
Collapse
|
30
|
Pavuluri K, Yang E, Ayyappan V, Sonkar K, Tan Z, Tressler CM, Bo S, Bibic A, Glunde K, McMahon MT. Unlabeled aspirin as an activatable theranostic MRI agent for breast cancer. Theranostics 2022; 12:1937-1951. [PMID: 35198081 PMCID: PMC8825591 DOI: 10.7150/thno.53147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 11/17/2021] [Indexed: 01/11/2023] Open
Abstract
Rationale: Chemical exchange saturation transfer (CEST) magnetic resonance imaging (MRI) is emerging as an alternative to gadolinium-based contrast MRI. We have evaluated the possibility of CEST MRI of orthotopic breast tumor xenografts with unlabeled aspirin's conversion to salicylic acid (SA) through various enzymatic activities, most notably inhibition of cyclooxygenase (COX)-1/-2 enzymes. Methods: We measured the COX-1/-2 expression in four breast cancer cell lines by Western Blot analysis and selected the highest and lowest expressing cell lines. We then performed CEST MRI following aspirin treatment to detect SA levels and ELISA to measure levels of downstream prostaglandin E2 (PGE2). We also injected aspirin into the tail vein of mice growing orthotopic tumor xenografts which expressed high and low COX-1/-2 and acquired SA CEST MR images of these tumor xenografts for up to 70 minutes. Tumors were then harvested to perform Western Blot and ELISA experiments to measure COX-1/-2 expression and PGE2 levels, respectively. Results: Western Blots determined that SUM159 cells contained significantly higher COX-1/-2 expression levels than MDA-MB-231 cells, in line with higher levels of downstream PGE2. SA CEST MRI yielded similar contrast at approximately 3% for both cell lines, independent of COX-1/-2 expression level. PGE2 levels decreased by about 50% following aspirin treatment. Results from our mouse study aligned with cultured cells, the overall SA CEST MRI contrast in both MDA-MB-231 and SUM159 tumor xenograft models was 5~8% at one hour post injection. PGE2 levels were ten times higher in SUM159 than MDA-MB-231 and decreased by 50%. The CEST contrast directly depended on the injected dose, with ~6%, ~3% and ~1.5% contrast observed following injection of 100 µL of 300 mM, 200 mM and 150 mM aspirin, respectively. Conclusions: Our data demonstrate the feasibility of using aspirin as a noninvasive activatable CEST MRI contrast agent for breast tumor detection.
Collapse
Affiliation(s)
- KowsalyaDevi Pavuluri
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ethan Yang
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Vinay Ayyappan
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kanchan Sonkar
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zheqiong Tan
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Caitlin M. Tressler
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shaowei Bo
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Adnan Bibic
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
| | - Kristine Glunde
- Division of Cancer Imaging Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
- The Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Michael T McMahon
- Division of MR Research, The Russell H. Morgan Department of Radiology and Radiological Science; The Johns Hopkins University School of Medicine, Baltimore, MD
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD
| |
Collapse
|
31
|
Colorectal Cancer Survivors' Receptivity toward Genomic Testing and Targeted Use of Non-Steroidal Anti-Inflammatory Drugs to Prevent Cancer Recurrence. J Community Genet 2022; 13:201-214. [PMID: 34997901 PMCID: PMC8941057 DOI: 10.1007/s12687-021-00574-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 12/22/2021] [Indexed: 10/19/2022] Open
Abstract
Genomic testing and targeted use of non-steroidal anti-inflammatory drugs (NSAIDs) may mitigate cancer recurrence risks. This study examines colorectal cancer (CRC) survivors' interest and receptivity to these strategies. Patients diagnosed with stage I-III CRC in 2004-2012 were recruited through the New Mexico Cancer Registry to complete a cancer survivorship experiences survey. We assessed interest in genomic testing, daily aspirin (ASA) and NSAID use, and receptivity to future daily ASA/NSAIDs. Descriptive statistics and multivariable logistic regression models estimated factors associated with genomic testing interest. Receptivity to future ASA/NSAIDs use was estimated for non-users of ASA/NSAIDs. Among CRC survivors (n = 273), 83% endorsed interest in genomic testing, 25% were ASA users and 47% ASA/NSAIDs users. In our final model, genomic testing interest was associated with being uncoupled [OR = 4.11; 95% CI = 1.49-11.35], low income [OR = 0.35, 95% CI: 0.14-0.88], smoking history [OR = 0.35, 95% CI: 0.14-0.90], low [OR: 0.33, 95% CI: 0.07-1.43] and moderate [OR: 0.26, 95% CI: 0.11-0.61] health literacy, and personal CRC risk worry [OR: 2.86, 95% CI: 1.63-5.02, p = 0.0002]. In our final model, ASA use was associated with age [OR: 1.05, 95% CI: 1.01-1.10] and cardiovascular disease history [OR: 2.42, 95% CI: 1.23-4.73, p = 0.010]. Among non-users ASA/NSAIDs, 83% reported receptivity to ASA/NSAIDs to reduce cancer risks, and no significant correlates were identified. The majority of survivors' expressed genomic testing interest and endorsed receptivity toward ASA/NSAIDs use for cancer risk management. Further research to optimize ASA/NSAIDs use guided by genomic testing is warranted.
Collapse
|
32
|
Abrahami D, McDonald EG, Schnitzer ME, Barkun AN, Suissa S, Azoulay L. Proton pump inhibitors and risk of colorectal cancer. Gut 2022; 71:111-118. [PMID: 34210775 DOI: 10.1136/gutjnl-2021-325096] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To determine whether proton pump inhibitors (PPIs) are associated with an increased risk of colorectal cancer, compared with histamine-2 receptor antagonists (H2RAs). DESIGN The United Kingdom Clinical Practice Research Datalink was used to identify initiators of PPIs and H2RA from 1990 to 2018, with follow-up until 2019. Cox proportional hazards models were fit to estimate marginal HRs and 95% CIs of colorectal cancer. The models were weighted using standardised mortality ratio weights using calendar time-specific propensity scores. Prespecified secondary analyses assessed associations with cumulative duration, cumulative dose and time since treatment initiation. The number needed to harm was calculated at five and 10 years of follow-up. RESULTS The cohort included 1 293 749 and 292 387 initiators of PPIs and H2RAs, respectively, followed for a median duration of 4.9 years. While the use of PPIs was not associated with an overall increased risk of colorectal cancer (HR: 1.02, 95% CI 0.92 to 1.14), HRs increased with cumulative duration of PPI use (<2 years, HR: 0.93, 95% CI 0.83 to 1.04; 2-4 years, HR: 1.45, 95% CI 1.28 to 1.60; ≥4 years, HR: 1.60, 95% CI 1.42 to 1.80). Similar patterns were observed with cumulative dose and time since treatment initiation. The number needed to harm was 5343 and 792 for five and 10 years of follow-up, respectively. CONCLUSION While any use of PPIs was not associated with an increased risk of colorectal cancer compared with H2RAs, prolonged use may be associated with a modest increased risk of this malignancy.
Collapse
Affiliation(s)
- Devin Abrahami
- Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada.,Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Montreal, Quebec, Canada
| | - Emily Gibson McDonald
- Division of General Internal Medicine, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Mireille E Schnitzer
- Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada.,Faculty of Pharmacy and the Department of Social and Preventive Medicine, Universite de Montreal, Montreal, Quebec, Canada
| | - Alan N Barkun
- Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada.,Gastroenterology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Samy Suissa
- Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada.,Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Laurent Azoulay
- Epidemiology, Biostatistics, and Occupational Health, McGill University, Montreal, Quebec, Canada .,Centre for Clinical Epidemiology, Lady Davis Institute for Medical Research, Montreal, Quebec, Canada.,Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
33
|
Ethanol Extract of Pomegranate ( Punica granatum) Peel in Increasing the Expression of Caspase-3 in DSS-Induced Mice. Int J Inflam 2021; 2021:4919410. [PMID: 34900217 PMCID: PMC8660243 DOI: 10.1155/2021/4919410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/04/2021] [Accepted: 11/10/2021] [Indexed: 12/24/2022] Open
Abstract
Background Colorectal cancer (CRC) is a malignancy derived from the glandular epithelial cells in the colon. Patients with inflammatory bowel disease (IBD) are more likely to develop CRC. Cancer proliferation is characterized by the loss of inhibition of apoptosis, which involves caspase-3 activation. This study examined the effects of the pomegranate peel extract on the expression of caspase-3 in mice crypt cells induced by dextran sodium sulfate (DSS) 2%. Methods The experimental study was done in six groups. All treatments were done in 42 days. The groups were all induced by DSS through water drinking, except for the normal group, which was only given water. The treatments given included the pomegranate extract in two doses (240 mg and 480 mg/kg bw/day), aspirin, and ellagic acid. The specimens were then fixated and stained for the immunohistochemistry scoring for the expression of caspase-3, which was then analyzed statistically. Results The H-scores of each treatment group were 213.23 ± 8.32 (DSS group), 243.81 ± 18.69 (normal group), 226.10 ± 12.38 (pomegranate peel extract of 240 mg/kg/d), 238.84 ± 15.81 (pomegranate peel extract of 480 mg/kg/d), 227.47 ± 12.15 (aspirin), and 224.01 ± 18.39 (ellagic acid). Statistical differences were found in one-way analysis of variance (ANOVA) and post hoc analysis among the DSS group, normal group, and dose 2 group (pomegranate peel extract of 480 mg/kg/day). Conclusions The ethanol extract of pomegranate was able to induce apoptosis, which was demonstrated by the increase of caspase-3 expression.
Collapse
|
34
|
Harlid S, Gunter MJ, Van Guelpen B. Risk-Predictive and Diagnostic Biomarkers for Colorectal Cancer; a Systematic Review of Studies Using Pre-Diagnostic Blood Samples Collected in Prospective Cohorts and Screening Settings. Cancers (Basel) 2021; 13:4406. [PMID: 34503217 PMCID: PMC8430893 DOI: 10.3390/cancers13174406] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022] Open
Abstract
This systematic review summarizes the evidence for blood-based colorectal cancer biomarkers from studies conducted in pre-diagnostic, asymptomatic settings. Of 1372 studies initially identified, the final selection included 30 studies from prospective cohorts and 23 studies from general screening settings. Overall, the investigations had high quality but considerable variability in data analysis and presentation of results, and few biomarkers demonstrated a clinically relevant discriminatory ability. One of the most promising biomarkers was the anti-p53 antibody, with consistent findings in one screening cohort and in the 3-4 years prior to diagnosis in two prospective cohort studies. Proteins were the most common type of biomarker assessed, particularly carcinoembryonic antigen (CEA) and C-reactive protein (CRP), with modest results. Other potentially promising biomarkers included proteins, such as AREG, MIC-1/GDF15, LRG1 and FGF-21, metabolites and/or metabolite profiles, non-coding RNAs and DNA methylation, as well as re-purposed routine lab tests, such as ferritin and the triglyceride-glucose index. Biomarker panels generally achieved higher discriminatory performance than single markers. In conclusion, this systematic review highlighted anti-p53 antibodies as a promising blood-based biomarker for use in colorectal cancer screening panels, together with other specific proteins. It also underscores the need for validation of promising biomarkers in independent pre-diagnostic settings.
Collapse
Affiliation(s)
- Sophia Harlid
- Department of Radiation Sciences, Oncology, Umeå University, 90187 Umeå, Sweden;
| | - Marc J. Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, 69372 Lyon, France;
| | - Bethany Van Guelpen
- Department of Radiation Sciences, Oncology, Umeå University, 90187 Umeå, Sweden;
- Wallenberg Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
35
|
Gao Y, Byrd DA, Prizment A, Lazovich D, Bostick RM. Associations of Novel Lifestyle- and Whole Foods-Based Inflammation Scores with Incident Colorectal Cancer Among Women. Nutr Cancer 2021; 74:1356-1369. [PMID: 34296959 PMCID: PMC9281615 DOI: 10.1080/01635581.2021.1952629] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/14/2021] [Accepted: 06/11/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND Chronic inflammation, associated with lifestyle and dietary factors, may contribute to colorectal carcinogenesis. To address this, we investigated associations of previously validated, inflammation biomarker panel-weighted, novel, 4-component lifestyle (LIS) and 19-component predominately whole foods-based dietary (DIS) inflammation scores with incident colorectal cancer (CRC) in the prospective Iowa Women's Health Study (IWHS; 1986-2012; n = 34,254, of whom 1,632 developed CRC). METHODS We applied the published scores' components' weights, summed the weighted components to constitute the scores (higher scores reflect a higher balance of pro-inflammatory exposures), and investigated LIS- and DIS-CRC associations using multivariable Cox proportional hazards regression. RESULTS The multivariable-adjusted hazards ratios (HR) and their 95% confidence intervals (CI) for CRC among participants in the highest relative to the lowest LIS and DIS quintiles were 1.47 (1.26, 1.72; Ptrend < 0.01) and 1.07 (0.91, 1.25; Ptrend = 0.22), respectively. The corresponding findings for distal colon cancers were HR 1.78 (1.29, 2.47) and HR 1.34 (0.98, 1.84), respectively. Among those in the highest relative to the lowest joint LIS/DIS quintile, the HR for CRC was 1.60 (95% CI 1.30, 1.98). CONCLUSIONS Our results suggest that a more pro-inflammatory lifestyle, alone and jointly with a more pro-inflammatory diet, may be associated with higher CRC risk.
Collapse
Affiliation(s)
- Yasheen Gao
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Doratha A. Byrd
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Anna Prizment
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - DeAnn Lazovich
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota
| | - Roberd M. Bostick
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
- Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
36
|
Jokinen EM, Gopinath K, Kurkinen ST, Pentikäinen OT. Detection of Binding Sites on SARS-CoV-2 Spike Protein Receptor-Binding Domain by Molecular Dynamics Simulations in Mixed Solvents. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1281-1289. [PMID: 33914685 PMCID: PMC8791430 DOI: 10.1109/tcbb.2021.3076259] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 04/13/2021] [Accepted: 04/20/2021] [Indexed: 06/12/2023]
Abstract
The novel SARS-CoV-2 uses ACE2 (Angiotensin-Converting Enzyme 2) receptor as an entry point. Insights on S protein receptor-binding domain (RBD) interaction with ACE2 receptor and drug repurposing has accelerated drug discovery for the novel SARS-CoV-2 infection. Finding small molecule binding sites in S protein and ACE2 interface is crucial in search of effective drugs to prevent viral entry. In this study, we employed molecular dynamics simulations in mixed solvents together with virtual screening to identify small molecules that could be potential inhibitors of S protein -ACE2 interaction. Observation of organic probe molecule localization during the simulations revealed multiple sites at the S protein surface related to small molecule, antibody, and ACE2 binding. In addition, a novel conformation of the S protein was discovered that could be stabilized by small molecules to inhibit attachment to ACE2. The most promising binding site on RBD-ACE2 interface was targeted with virtual screening and top-ranked compounds (DB08248, DB02651, DB03714, and DB14826) are suggested for experimental testing. The protocol described here offers an extremely fast method for characterizing key proteins of a novel pathogen and for the identification of compounds that could inhibit or accelerate spreading of the disease.
Collapse
|
37
|
Platelets and extracellular vesicles and their cross talk with cancer. Blood 2021; 137:3192-3200. [PMID: 33940593 DOI: 10.1182/blood.2019004119] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Platelets play significant and varied roles in cancer progression, as detailed throughout this review series, via direct interactions with cancer cells and by long-range indirect interactions mediated by platelet releasates. Microvesicles (MVs; also referred to as microparticles) released from activated platelets have emerged as major contributors to the platelet-cancer nexus. Interactions of platelet-derived MVs (PMVs) with cancer cells can promote disease progression through multiple mechanisms, but PMVs also harbor antitumor functions. This complex relationship derives from PMVs' binding to both cancer cells and nontransformed cells in the tumor microenvironment and transferring platelet-derived contents to the target cell, each of which can have stimulatory or modulatory effects. MVs are extracellular vesicles of heterogeneous size, ranging from 100 nm to 1 µm in diameter, shed by living cells during the outward budding of the plasma membrane, entrapping local cytosolic contents in an apparently stochastic manner. Hence, PMVs are encapsulated by a lipid bilayer harboring surface proteins and lipids mirroring the platelet exterior, with internal components including platelet-derived mature messenger RNAs, pre-mRNAs, microRNAs, and other noncoding RNAs, proteins, second messengers, and mitochondria. Each of these elements engages in established and putative PMV functions in cancer. In addition, PMVs contribute to cancer comorbidities because of their roles in coagulation and thrombosis and via interactions with inflammatory cells. However, separating the effects of PMVs from those of platelets in cancer contexts continues to be a major hurdle. This review summarizes our emerging understanding of the complex roles of PMVs in the development and progression of cancer and cancer comorbidities.
Collapse
|
38
|
Baumeister T, Ingermann J, Marcazzan S, Fang HY, Oellinger R, Rad R, Engleitner T, Kleigrewe K, Anand A, Strangmann J, Schmid RM, Wang TC, Quante M. Anti-inflammatory chemoprevention attenuates the phenotype in a mouse model of esophageal adenocarcinoma. Carcinogenesis 2021; 42:1068-1078. [PMID: 33878160 DOI: 10.1093/carcin/bgab032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 01/29/2021] [Accepted: 04/16/2021] [Indexed: 01/20/2023] Open
Abstract
Barrett´s Esophagus (BE) is the main known precursor condition of Esophageal Adenocarcinoma (EAC). BE is defined by the presence of metaplasia above the normal squamous columnar junction and has mainly been attributed to gastroesophageal reflux disease (GERD) and chronic reflux esophagitis. Thus, the rising incidence of EAC in the Western world is likely mediated by chronic esophageal inflammation, secondary to GERD in combination with environmental risk factors such as a Western diet and obesity. However, (at present) risk prediction tools and endoscopic surveillance have shown limited effectiveness. Chemoprevention as an adjunctive approach remains an attractive option to reduce the incidence of neoplastic disease. Here, we investigate the feasibility of chemopreventive approaches in BE and EAC via inhibition of inflammatory signaling in a transgenic mouse model of BE and EAC (L2-IL1B mice), with accelerated tumor formation on a high fat diet (HFD). L2-IL1B mice were treated with the IL-1 receptor antagonist Anakinra and the nonsteroidal anti-inflammatory drugs (NSAIDs) aspirin or Sulindac. Interleukin-1b antagonism reduced tumor progression in L2-IL1B mice with or without a HFD, while both NSAIDs were effective chemoprevention agents in the accelerated HFD fed L2-IL1B mouse model. Sulindac treatment also resulted in a marked change in the immune profile of L2-IL-1B mice. In summary, anti-inflammatory treatment of HFD-treated L2-IL1B mice acted protectively on disease progression. These results from a mouse model of BE support results from clinical trials that suggest that anti-inflammatory medication may be effective in the chemoprevention of EAC.
Collapse
Affiliation(s)
- Theresa Baumeister
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Jonas Ingermann
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Sabrina Marcazzan
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Hsin-Yu Fang
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Rupert Oellinger
- Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Roland Rad
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany.,Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Thomas Engleitner
- Institute of Molecular Oncology and Functional Genomics, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Karin Kleigrewe
- Leibniz-Institute for Food Systems Biology at the Technical University of Munich
| | - Akanksha Anand
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Julia Strangmann
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Roland M Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany
| | - Timothy C Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | - Michael Quante
- Department of Medicine II, Klinikum rechts der Isar, Technical University Munich (TUM), Ismaninger Str, München, Germany.,Innere Medizin II, Universitätskliniken Freiburg, Universität Freiburg, Germany
| |
Collapse
|
39
|
Joseph DF, Li E, Stanley III SL, Zhu YC, Li XN, Yang J, Ottaviano LF, Bucobo JC, Buscaglia JM, Miller JD, Veluvolu R, Follen M, Grossman EB. Impact of type 2 diabetes on adenoma detection in screening colonoscopies performed in disparate populations. World J Clin Cases 2021; 9:2433-2445. [PMID: 33889609 PMCID: PMC8040183 DOI: 10.12998/wjcc.v9.i11.2433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 12/23/2020] [Accepted: 02/25/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The Black/African Ancestry (AA) population has a higher prevalence of type 2 diabetes mellitus (T2DM) and a higher incidence and mortality rate for colorectal cancer (CRC) than all other races in the United States. T2DM has been shown to increase adenoma risk in predominantly white/European ancestry (EA) populations, but the effect of T2DM on adenoma risk in Black/AA individuals is less clear. We hypothesize that T2DM has a significant effect on adenoma risk in a predominantly Black/AA population.
AIM To investigate the effect of T2DM and race on the adenoma detection rate (ADR) in screening colonoscopies in two disparate populations.
METHODS A retrospective cohort study was conducted on ADR during index screening colonoscopies (age 45-75) performed at an urban public hospital serving a predominantly Black/AA population (92%) (2017-2018, n = 1606). Clinical metadata collected included basic demographics, insurance, body mass index (BMI), family history of CRC, smoking, diabetes diagnosis, and aspirin use. This dataset was combined with a recently reported parallel retrospective cohort data set collected at a suburban university hospital serving a predominantly White/EA population (87%) (2012-2015, n = 2882).
RESULTS The ADR was higher in T2DM patients than in patients without T2DM or prediabetes (35.2% vs 27.9%, P = 0.0166, n = 981) at the urban public hospital. Multivariable analysis of the combined datasets showed that T2DM [odds ratio (OR) = 1.29, 95% confidence interval (CI): 1.08-1.55, P = 0.0049], smoking (current vs never OR = 1.47, 95%CI: 1.18-1.82, current vs past OR = 1.32, 95%CI: 1.02-1.70, P = 0.0026), older age (OR = 1.05 per year, 95%CI: 1.04-1.06, P < 0.0001), higher BMI (OR = 1.02 per unit, 95%CI: 1.01-1.03, P = 0.0003), and male sex (OR = 1.87, 95%CI: 1.62-2.15, P < 0.0001) were associated with increased ADR in the combined datasets, but race, aspirin use and insurance were not.
CONCLUSION T2DM, but not race, is significantly associated with increased ADR on index screening colonoscopy while controlling for other factors.
Collapse
Affiliation(s)
- Dimitri F Joseph
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Ellen Li
- Department of Medicine, Division of Gastroenterology, Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Samuel L Stanley III
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Yi-Cong Zhu
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Xiao-Ning Li
- Department of Biostatistics and Bioinformatics Shared Resource, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794-3600, United States
| | - Jie Yang
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794-3600, United States
- Department of Family, Population, and Preventative Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8461, United States
| | - Lorenzo F Ottaviano
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Juan Carlos Bucobo
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Jonathan M Buscaglia
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Joshua D Miller
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY 11794-8173, United States
| | - Rajesh Veluvolu
- Department of Medicine, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| | - Michele Follen
- Department of Obstetrics and Gynecology, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
| | - Evan B Grossman
- Department of Medicine, NYC Health and Hospitals/Kings County, Brooklyn, NY 11203, United States
- Department of Medicine, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States
| |
Collapse
|
40
|
Belayneh YM, Amare GG, Meharie BG. Updates on the molecular mechanisms of aspirin in the prevention of colorectal cancer: Review. J Oncol Pharm Pract 2021; 27:954-961. [PMID: 33427041 DOI: 10.1177/1078155220984846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Colorectal cancer is one of the commonest malignancies worldwide. The estimated lifetime risk of the disease is about 5% with an incidence of one million new cases and 600,000 deaths worldwide every year. It is estimated that in 2019, approximately 134,490 new cases of colorectal cancer will be diagnosed with 49,190 mortalities. Though the disease is regarded as a disorder of the more developed world, the occurrence is steadily increasing in many developing countries. Since chronic inflammation is a known aggravating risk factor for colorectal cancer, anti-inflammatory agents such as aspirin have been used to prevent the development of colorectal cancer and related mortality. The potential mechanisms for the effect of aspirin in the prevention of colorectal cancer have been proposed and broadly classified as cyclooxygenase (COX) dependent and COX-independent. Some of the primary effectors of COX-dependent mechanisms in carcinogenesis are likely to be prostaglandins. In contrast to the reversible action of other nonsteroidal anti-inflammatory drugs, aspirin is known to irreversibly inactivate COX enzymes to suppress production of prostaglandins. COX-independent mechanisms of anticancer effects of aspirin include down-regulation of nuclear factor kappa B activity and Akt activation, modulation of Bcl-2 and Bax family proteins, suppression of vascular endothelial growth factor, induction of apoptosis, disruption of DNA repair mechanisms, and induction of spermidine/spermine N1-acetyltransferase that modulates polyamine catabolism.
Collapse
Affiliation(s)
- Yaschilal Muche Belayneh
- Department of Pharmacy, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Gedefew Getnet Amare
- Department of Pharmacy, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| | - Birhanu Geta Meharie
- Department of Pharmacy, College of Medicine and Health Sciences, Wollo University, Dessie, Ethiopia
| |
Collapse
|
41
|
Hurwitz LM, Michels KA, Cook MB, Pfeiffer RM, Trabert B. Associations between daily aspirin use and cancer risk across strata of major cancer risk factors in two large U.S. cohorts. Cancer Causes Control 2021; 32:57-65. [PMID: 33104910 PMCID: PMC7855934 DOI: 10.1007/s10552-020-01357-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Daily aspirin use has been shown to reduce risk of colorectal, and possibly other, cancers, but it is unknown if these benefits are consistent across subgroups of people with differing cancer risk factors. We investigated whether age, body mass index (BMI), smoking status, physical inactivity, and family history of cancer modify the effect of daily aspirin use on colorectal, ovarian, breast, endometrial and aggressive prostate cancer risk. METHODS We pooled 423,495 individuals from two prospective, U.S.-based studies: the NIH-AARP Diet and Health Study (1995-2011) and the Prostate, Lung, Colorectal, and Ovarian Cancer Screening Trial (1993-2009). Using Cox proportional hazards regression, we examined associations between daily aspirin use (≥ 5 days/week) and risk of colorectal, ovarian, breast, endometrial, and aggressive prostate cancer, overall and across strata of risk factors. RESULTS Daily aspirin use was associated with a 15% reduction in colorectal cancer risk (hazard ratio [HR]: 0.85, 95% confidence interval [CI] 0.80-0.89). Risk reductions were generally consistent across strata of risk factors but attenuated with increasing BMI (p-interaction = 0.16). For ovarian cancer, there was no significant association overall (HR: 0.93, 95% CI 0.80-1.08) but reduced risk among obese women (HR: 0.73, 95% CI 0.52-0.98, p-interaction = 0.12). Weak or null associations were observed for breast, endometrial, and aggressive prostate cancer, with no strong effect modification observed. CONCLUSIONS Daily aspirin use appears to reduce colorectal cancer risk regardless of other risk factors, though the potential modifying effect of BMI warrants further investigation and may need to be considered in risk-benefit calculations for aspirin use.
Collapse
Affiliation(s)
- Lauren M Hurwitz
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA.
| | - Kara A Michels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Michael B Cook
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| | - Britton Trabert
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Rockville, MD, 20850, USA
| |
Collapse
|
42
|
Sankaranarayanan R, Kumar DR, Altinoz MA, Bhat GJ. Mechanisms of Colorectal Cancer Prevention by Aspirin-A Literature Review and Perspective on the Role of COX-Dependent and -Independent Pathways. Int J Mol Sci 2020; 21:ijms21239018. [PMID: 33260951 PMCID: PMC7729916 DOI: 10.3390/ijms21239018] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Aspirin, synthesized and marketed in 1897 by Bayer, is one of the most widely used drugs in the world. It has a well-recognized role in decreasing inflammation, pain and fever, and in the prevention of thrombotic cardiovascular diseases. Its anti-inflammatory and cardio-protective actions have been well studied and occur through inhibition of cyclooxygenases (COX). Interestingly, a vast amount of epidemiological, preclinical and clinical studies have revealed aspirin as a promising chemopreventive agent, particularly against colorectal cancers (CRC); however, the primary mechanism by which it decreases the occurrences of CRC has still not been established. Numerous mechanisms have been proposed for aspirin’s chemopreventive properties among which the inhibition of COX enzymes has been widely discussed. Despite the wide attention COX-inhibition has received as the most probable mechanism of cancer prevention by aspirin, it is clear that aspirin targets many other proteins and pathways, suggesting that these extra-COX targets may also be equally important in preventing CRC. In this review, we discuss the COX-dependent and -independent pathways described in literature for aspirin’s anti-cancer effects and highlight the strengths and limitations of the proposed mechanisms. Additionally, we emphasize the potential role of the metabolites of aspirin and salicylic acid (generated in the gut through microbial biotransformation) in contributing to aspirin’s chemopreventive actions. We suggest that the preferential chemopreventive effect of aspirin against CRC may be related to direct exposure of aspirin/salicylic acid or its metabolites to the colorectal tissues. Future investigations should shed light on the role of aspirin, its metabolites and the role of the gut microbiota in cancer prevention against CRC.
Collapse
Affiliation(s)
- Ranjini Sankaranarayanan
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD 57007, USA;
| | - D. Ramesh Kumar
- Department of Entomology, University of Kentucky, Lexington, KY 40506, USA;
| | - Meric A. Altinoz
- Department of Biochemistry, Acibadem M.A.A. University, Istanbul, Turkey;
| | - G. Jayarama Bhat
- Department of Pharmaceutical Sciences and Translational Cancer Research Center, College of Pharmacy and Allied Health Professions, South Dakota State University, Brookings, SD 57007, USA;
- Correspondence: ; Tel.: +1-605-688-6894
| |
Collapse
|
43
|
Abdalla LF, Chaudhry Ehsanullah R, Karim F, Oyewande AA, Khan S. Role of Using Nonsteroidal Anti-Inflammatory Drugs in Chemoprevention of Colon Cancer in Patients With Inflammatory Bowel Disease. Cureus 2020; 12:e8240. [PMID: 32582499 PMCID: PMC7306635 DOI: 10.7759/cureus.8240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The process of inflammation occurs due to inflammatory mediators, including prostaglandins, cytokines, and tumor necrosis factor (TNF). All these mediators activate the process of tumorigenesis and dysplasia, leading to colitis-associated cancer. Several drugs used to decrease these mediators will help in the treatment of acute attacks and also help in prolonged remissions of the disease by using nonsteroidal anti-inflammatory drugs (NSAIDs), steroids, and biological factors. Reducing these inflammatory mediators also have a role in chemoprevention and prevent progression to colorectal carcinoma. The most researched drugs in this process of chemoprevention are NSAIDs as it has both cyclooxygenase-2 (COX-2) inhibitory and non-inhibitory effects. These drugs should be taken for a long time and in large doses to reach this effect, which puts the patient at risk for various side effects. Researchers will need to do more research in the future to find the lowest effective dose that can reach the chemopreventive effect. We used database Pubmed as the main source for data search and extracted articles exploring the relationship between NSAIDs and their role in chemoprevention of colorectal carcinoma in inflammatory bowel disease (IBD) patients. We chose 23 studies which included seven review articles. We found that inflammatory mediators have a key role in colitis-associated cancer.
Collapse
Affiliation(s)
- Lamis F Abdalla
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | | | - Fazida Karim
- Psychology, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA.,Business & Management, University Sultan Zainal Abidin, Terengganu, MYS
| | - Azeezat A Oyewande
- Family Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA.,Family Medicine, Lagos State Health Service Commission/Alimosho General Hospital, Lagos, NGA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
44
|
The Role of Nicotinamide in Cancer Chemoprevention and Therapy. Biomolecules 2020; 10:biom10030477. [PMID: 32245130 PMCID: PMC7175378 DOI: 10.3390/biom10030477] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/09/2020] [Accepted: 03/17/2020] [Indexed: 12/24/2022] Open
Abstract
Nicotinamide (NAM) is a water-soluble form of Vitamin B3 (niacin) and a precursor of nicotinamide-adenine dinucleotide (NAD+) which regulates cellular energy metabolism. Except for its role in the production of adenosine triphosphate (ATP), NAD+ acts as a substrate for several enzymes including sirtuin 1 (SIRT1) and poly ADP-ribose polymerase 1 (PARP1). Notably, NAM is an inhibitor of both SIRT1 and PARP1. Accumulating evidence suggests that NAM plays a role in cancer prevention and therapy. Phase III clinical trials have confirmed its clinical efficacy for non-melanoma skin cancer chemoprevention or as an adjunct to radiotherapy against head and neck, laryngeal, and urinary bladder cancers. Evidence for other cancers has mostly been collected through preclinical research and, in its majority, is not yet evidence-based. NAM has potential as a safe, well-tolerated, and cost-effective agent to be used in cancer chemoprevention and therapy. However, more preclinical studies and clinical trials are needed to fully unravel its value.
Collapse
|
45
|
Byrd DA, Judd SE, Flanders WD, Hartman TJ, Fedirko V, Agurs-Collins T, Bostick RM. Associations of Novel Dietary and Lifestyle Inflammation Scores With Incident Colorectal Cancer in the NIH-AARP Diet and Health Study. JNCI Cancer Spectr 2020; 4:pkaa009. [PMID: 32455332 PMCID: PMC7236782 DOI: 10.1093/jncics/pkaa009] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/27/2020] [Accepted: 02/28/2020] [Indexed: 12/13/2022] Open
Abstract
Background Chronically higher inflammation, likely contributed to by dietary and lifestyle exposures, may increase risk for colorectal cancer (CRC). To address this, we investigated associations of novel dietary (DIS) and lifestyle (LIS) inflammation scores with incident CRC in the prospective National Institutes of Health–American Association of Retired Persons Diet and Health Study (N = 453 465). Methods The components of our previously developed and externally validated 19-component DIS and 4-component LIS were weighted based on their strengths of associations with a panel of circulating inflammation biomarker concentrations in a diverse subset (N = 639) of participants in the REasons for Geographic and Racial Differences in Stroke Study cohort. We calculated the components and applied their weights in the National Institutes of Health-American Association of Retired Persons cohort at baseline, summed the weighted components (higher scores reflect a higher balance of proinflammatory exposures), and investigated associations of the scores with incident CRC using Cox proportional hazards regression. All statistical tests were two-sided. Results Over a mean 13.5 years of follow-up, 10 336 participants were diagnosed with CRC. Among those in the highest relative to the lowest DIS and LIS quintiles, the multivariable-adjusted hazards ratios (HRs) and their 95% confidence intervals (CIs) were HR = 1.27 (95% CI = 1.19 to 1.35; Ptrend < .001) and 1.38 (95% CI = 1.30 to 1.48; Ptrend < .001), respectively. The associations were stronger among men and for colon cancers. The hazards ratio for those in the highest relative to the lowest joint DIS and LIS quintile was HR = 1.83 (95% CI = 1.68 to 1.99; Pinteraction < .001). Conclusions Aggregates of proinflammatory dietary and lifestyle exposures may be associated with higher risk for CRC.
Collapse
Affiliation(s)
- Doratha A Byrd
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Suzanne E Judd
- Department of Biostatistics, University of Alabama, Birmingham School of Public Health, Birmingham, AL, USA
| | - W Dana Flanders
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Terryl J Hartman
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Veronika Fedirko
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | | | - Roberd M Bostick
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA.,Winship Cancer Institute, Emory University, Atlanta, GA, USA
| |
Collapse
|
46
|
Kim SW, Goossens A, Libert C, Van Immerseel F, Staal J, Beyaert R. Phytohormones: Multifunctional nutraceuticals against metabolic syndrome and comorbid diseases. Biochem Pharmacol 2020; 175:113866. [PMID: 32088261 DOI: 10.1016/j.bcp.2020.113866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is characterized by the co-occurrence of diverse symptoms initiating the development of type 2 diabetes, cardiovascular diseases, and a variety of comorbid diseases. The complex constellation of numerous comorbidities makes it difficult to develop common therapeutic approaches that ameliorate these pathological features simultaneously. The plant hormones abscisic acid, salicylic acid, auxin, and cytokinins, have shown promising anti-inflammatory and pro-metabolic effects that could mitigate several disorders relevant to metabolic syndrome. Intriguingly, besides plants, human cells and gut microbes also endogenously produce these molecules, indicating a role in the complex interplay between inflammatory responses associated with metabolic syndrome, the gut microbiome, and nutrition. Here, we introduce how bioactive phytohormones can be generated endogenously and through the gut microbiome. These molecules subsequently influence immune responses and metabolism. We also elaborate on how phytohormones can beneficially modulate metabolic syndrome comorbidities, and propose them as nutraceuticals.
Collapse
Affiliation(s)
- Seo Woo Kim
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Alain Goossens
- VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Claude Libert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jens Staal
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
47
|
Galon J, Bruni D. Tumor Immunology and Tumor Evolution: Intertwined Histories. Immunity 2020; 52:55-81. [PMID: 31940273 DOI: 10.1016/j.immuni.2019.12.018] [Citation(s) in RCA: 372] [Impact Index Per Article: 74.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 07/01/2019] [Accepted: 12/20/2019] [Indexed: 12/11/2022]
Abstract
Cancer is a complex disease whose outcome depends largely on the cross-talk between the tumor and its microenvironment. Here, we review the evolution of the field of tumor immunology and the advances, in lockstep, of our understanding of cancer as a disease. We discuss the involvement of different immune cells at distinct stages of tumor progression and how immune contexture determinants shaping tumor development are being exploited therapeutically. Current clinical stratification schemes focus on the tumor histopathology and the molecular characteristics of the tumor cell. We argue for the importance of revising these stratification systems to include immune parameters so as to address the immediate need for improved prognostic and/or predictive information to guide clinical decisions.
Collapse
Affiliation(s)
- Jérôme Galon
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Université de Paris; Centre de Recherche des Cordeliers, F-75006 Paris, France.
| | - Daniela Bruni
- INSERM, Laboratory of Integrative Cancer Immunology, Equipe Labellisée Ligue Contre le Cancer, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université Paris Diderot, Université de Paris; Centre de Recherche des Cordeliers, F-75006 Paris, France
| |
Collapse
|
48
|
Camara Planek MI, Silver AJ, Volgman AS, Okwuosa TM. Exploratory Review of the Role of Statins, Colchicine, and Aspirin for the Prevention of Radiation-Associated Cardiovascular Disease and Mortality. J Am Heart Assoc 2020; 9:e014668. [PMID: 31960749 PMCID: PMC7033839 DOI: 10.1161/jaha.119.014668] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
| | - Adam J. Silver
- Rush Heart Center for WomenRush University Medical CenterChicagoIL
| | | | | |
Collapse
|
49
|
Sun X, Ng TTH, Sham KWY, Zhang L, Chan MTV, Wu WKK, Cheng CHK. Bufalin, a Traditional Chinese Medicine Compound, Prevents Tumor Formation in Two Murine Models of Colorectal Cancer. Cancer Prev Res (Phila) 2019; 12:653-666. [PMID: 31431500 DOI: 10.1158/1940-6207.capr-19-0134] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 06/24/2019] [Accepted: 08/16/2019] [Indexed: 12/24/2022]
Abstract
Chemoprevention is cost-effective for colorectal cancer when targeted at intermediate- or high-risk populations. Bufalin is a cardiac glycoside extracted from the traditional Chinese medicine (TCM) "Chan Su," which has been used as an anticancer agent. On the basis of the relative safety of bufalin, we investigated whether bufalin could act as a chemoprophylactic agent to prevent colon tumorigenesis in two murine models, namely colitis-associated colorectal cancer and Apc germline mutation-developed colorectal cancer. Our results revealed that long-term (12-16 weeks) administration of low-dose bufalin (0.5 mg/kg) effectively suppressed tumorigenesis in both colorectal cancer models, accompanied by attenuated epithelial cell proliferation (reduced bromodeoxyuridine incorporation, lower levels of cyclin A, cyclin D1, cyclin E, and cyclin-dependent kinases-2/4, and higher levels of p21 and p27) and promoted apoptosis (increased TUNEL positivity and caspase-3/9 cleavages, reduced levels of Bcl-2, Bcl-xL and survivin, and increased levels of Bax and Bak). Bufalin also suppressed the expression of proinflammatory mediators [reduced levels of cyclooxygenase-2, tumor TNFα, IL1β, IL6, C-X-C motif chemokine ligand (CXCL)-1, CXCL-2, and CXCL-5] in the colitis-associated colorectal cancer model. These effects were associated with the inhibition of oncogenic NF-κB and PI3K/Akt pathways. Our findings unveil a novel chemoprophylactic action of bufalin in colorectal cancer in vivo and provided efficacy data and mechanistic evidence for further clinical evaluation of this TCM compound for colorectal cancer chemoprevention in individuals at risk of colorectal cancer.
Collapse
Affiliation(s)
- Xiao Sun
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Tony T H Ng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Kathy W Y Sham
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Lin Zhang
- State Key Laboratory of Digestive Diseases, Institute of Digestive Diseases and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong.,Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong
| | - William K K Wu
- State Key Laboratory of Digestive Diseases, Institute of Digestive Diseases and LKS Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong. .,Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong.,CUHK Shenzhen Research Institute, Shenzhen, China
| | - Christopher H K Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong. .,CUHK Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
50
|
Yap ML, McFadyen JD, Wang X, Ziegler M, Chen YC, Willcox A, Nowell CJ, Scott AM, Sloan EK, Hogarth PM, Pietersz GA, Peter K. Activated platelets in the tumor microenvironment for targeting of antibody-drug conjugates to tumors and metastases. Theranostics 2019; 9:1154-1169. [PMID: 30867822 PMCID: PMC6401411 DOI: 10.7150/thno.29146] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 01/12/2019] [Indexed: 12/16/2022] Open
Abstract
Rationale: Platelets are increasingly recognized as mediators of tumor growth and metastasis. Hypothesizing that activated platelets in the tumor microenvironment provide a targeting epitope for tumor-directed chemotherapy, we developed an antibody-drug conjugate (ADC), comprised of a single-chain antibody (scFv) against the platelet integrin GPIIb/IIIa (scFvGPIIb/IIIa) linked to the potent chemotherapeutic microtubule inhibitor, monomethyl auristatin E (MMAE). Methods: We developed an ADC comprised of three components: 1) A scFv which specifically binds to the high affinity, activated integrin GPIIb/IIIa on activated platelets. 2) A highly potent microtubule inhibitor, monomethyl auristatin E. 3) A drug activation/release mechanism using a linker cleavable by cathepsin B, which we demonstrate to be abundant in the tumor microenvironment. The scFvGPIIb/IIIa-MMAE was first conjugated with Cyanine7 for in vivo imaging. The therapeutic efficacy of the scFvGPIIb/IIIa-MMAE was then tested in a mouse metastasis model of triple negative breast cancer. Results: In vitro studies confirmed that this ADC specifically binds to activated GPIIb/IIIa, and cathepsin B-mediated drug release/activation resulted in tumor cytotoxicity. In vivo fluorescence imaging demonstrated that the newly generated ADC localized to primary tumors and metastases in a mouse xenograft model of triple negative breast cancer, a difficult to treat tumor for which a selective tumor-targeting therapy remains to be clinically established. Importantly, we demonstrated that the scFvGPIIb/IIIa-MMAE displays marked efficacy as an anti-cancer agent, reducing tumor growth and preventing metastatic disease, without any discernible toxic effects. Conclusion: Here, we demonstrate the utility of a novel ADC that targets a potent cytotoxic drug to activated platelets and specifically releases the cytotoxic agent within the confines of the tumor. This unique targeting mechanism, specific to the tumor microenvironment, holds promise as a novel therapeutic approach for the treatment of a broad range of primary tumors and metastatic disease, particularly for tumors that lack specific molecular epitopes for drug targeting.
Collapse
Affiliation(s)
- May Lin Yap
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, Australia
| | - James D McFadyen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Medicine, Monash University, Melbourne, 3800, Australia
- Department of Hematology, The Alfred Hospital, Melbourne, 3004, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Medicine, Monash University, Melbourne, 3800, Australia
| | - Melanie Ziegler
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Yung-Chih Chen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Abbey Willcox
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Medicine, Monash University, Melbourne, 3800, Australia
- Department of Hematology, The Alfred Hospital, Melbourne, 3004, Australia
| | - Cameron J Nowell
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia
- Department of Molecular Imaging and Therapy, Austin Health, and University of Melbourne, Melbourne, Victoria, Australia
| | - Erica K Sloan
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - P Mark Hogarth
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, Australia
- Burnet Institute, Melbourne, 3004, Australia
- Department of Immunology, Monash University, Melbourne, 3800, Australia
| | - Geoffrey A Pietersz
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Clinical Pathology, The University of Melbourne, Melbourne, 3010, Australia
- Burnet Institute, Melbourne, 3004, Australia
- Department of Immunology, Monash University, Melbourne, 3800, Australia
- College of Health and Biomedicine, Victoria University, Melbourne, 3021, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, 3004, Australia
- Department of Medicine, Monash University, Melbourne, 3800, Australia
- Department of Immunology, Monash University, Melbourne, 3800, Australia
| |
Collapse
|