1
|
Cheng YT, Sin CF, Ma ESK, Lam STS, Au Yeung SL, Cheung BMY, Tse HF, Yiu KH, Chan YH. Long-term risk of malignancies in persons with ischemic heart disease treated with trimetazidine dihydrochloride. COMMUNICATIONS MEDICINE 2025; 5:89. [PMID: 40133593 PMCID: PMC11937305 DOI: 10.1038/s43856-025-00805-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Metabolic reprogramming of energy processes is a cellular hallmark of various cancers. Whether trimetazidine, an anti-ischemic agent that preferentially potentiates cellular glucose oxidation, alters the long-term risk of malignancies is unknown. METHODS In this multi-center, retrospective cohort study, we studied the effect of trimetazidine on new-onset malignancies in 200,563 ischemic heart disease patients (mean age 70.8 years, 46.6% female) using the Hong Kong Clinical Data Analysis and Reporting System (CDARS), comparing trimetazidine users (n = 16,873) to nitrate users (n = 183,690, as control) over at least 30 days. The primary endpoint was defined as the estimated effect of trimetazidine on the overall new-onset occurrence of any malignancies a priori specified, diagnosed 90 days or more after the cohort entry. RESULTS Over a mean follow-up duration of 8.36 (6.42) years, the incidence rate of new-onset malignancies amongst trimetazidine users is significantly lower compared to the non-users (8.76 vs 12.3 per 1000-person years, trimetazidine to control incidence ratio, 0.71). Trimetazidine use is associated with improved event-free survival from new-onset malignancies (Mean survival: 231 [0.53] versus 225 [0.21] months, Chi-square = 161, P < 0.001). Multivariable Cox regression demonstrates an independently lower risk of new-onset malignancies associated with trimetazidine use, with (adjusted HRs, 0.71, 95% CI, 0.66-0.77, P < 0.001) and without (adjusted HRs, 0.71, 95% CI, 0.68-0.75, P < 0.001) propensity score matching. Subgroup analyses of new-onset malignancies including lung, colorectal, hepatobiliary & pancreatic, breast, stomach & oesophageal, renal & genito-urinary, prostate, and hematological malignancies, show similar risk reductions. CONCLUSIONS Modulation of metabolic reprogramming may represent a new therapeutic target for cancer prevention.
Collapse
Affiliation(s)
- Yuen-Ting Cheng
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chun-Fung Sin
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Edmond S K Ma
- Department of Molecular Pathology, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Stephen T S Lam
- Clinical Genetics Service, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Shiu-Lun Au Yeung
- School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Bernard M Y Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
- Genetics Research Program for Personalized Medicine in Cardiac Oncology, The University of Hong Kong, Hong Kong SAR, China
| | - Kai-Hang Yiu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, Shenzhen Hong Kong University Hospital, Hong Kong SAR, China
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yap-Hang Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Genetics Research Program for Personalized Medicine in Cardiac Oncology, The University of Hong Kong, Hong Kong SAR, China.
- Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Experimental Medicine and Immunotherapeutics, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK.
- Department of Cardiology, Royal Papworth Hospital, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
2
|
Gu Q, Wang Y, Yi P, Cheng C. Theoretical framework and emerging challenges of lipid metabolism in cancer. Semin Cancer Biol 2025; 108:48-70. [PMID: 39674303 DOI: 10.1016/j.semcancer.2024.12.002] [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: 10/11/2024] [Revised: 11/14/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Elevated lipid metabolism is one of hallmarks of malignant tumors. Lipids not only serve as essential structural components of biological membranes but also provide energy and substrates for the proliferation of cancer cells and tumor growth. Cancer cells meet their lipid needs by coordinating the processes of lipid absorption, synthesis, transport, storage, and catabolism. As research in this area continues to deepen, numerous new discoveries have emerged, making it crucial for scientists to stay informed about the developments of cancer lipid metabolism. In this review, we first discuss relevant concepts and theories or assumptions that help us understand the lipid metabolism and -based cancer therapies. We then systematically summarize the latest advancements in lipid metabolism including new mechanisms, novel targets, and up-to-date pre-clinical and clinical investigations of anti-cancer treatment with lipid metabolism targeted drugs. Finally, we emphasize emerging research directions and therapeutic strategies, and discuss future prospective and emerging challenges. This review aims to provide the latest insights and guidance for research in the field of cancer lipid metabolism.
Collapse
Affiliation(s)
- Qiuying Gu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Yuan Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China.
| | - Chunming Cheng
- Department of Oncology Science, OU Health Stephenson Cancer Center at University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
3
|
Yanagisawa A, Kim JD, Naito A, Kobayashi T, Misawa T, Sakao S, Jujo-Sanada T, Kawasaki T, Muroi SI, Sasaki SI, Suzuki T, Hayakawa Y, Nakagawa Y, Kasuya Y, Tatsumi K. Deciphering the inhibitory effects of trimetazidine on pulmonary hypertension development via decreasing fatty acid oxidation and promoting glucose oxidation. Sci Rep 2024; 14:27069. [PMID: 39511196 PMCID: PMC11544210 DOI: 10.1038/s41598-024-76100-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: 05/13/2024] [Accepted: 10/10/2024] [Indexed: 11/15/2024] Open
Abstract
Pulmonary hypertension (PH) is a devastating disease characterized by vascular remodeling, resulting in right ventricular failure and death. Dysregulation of energy metabolism is linked to PH pathogenesis. Trimetazidine (TMZ), a selective long-chain 3-ketoacyl coenzyme A thiolase inhibitor, is critical in maintaining energy metabolism. Despite the indicated TMZ's inhibitory effect on pulmonary vascular remodeling in PH development, the integrated evaluation of the changes in biomolecules, such as metabolites and transcripts, that TMZ induces in the lung and heart tissues is largely unknown in vivo. For an improved understanding of the molecular mechanism involving the effects of TMZ on PH development, we performed a comprehensive analysis of the changes in cardiac metabolites and pulmonary transcripts of SU5416-Hypoxia (Su/Hx) rats treated with TMZ. Metabolomic analysis of the Su/Hx-induced PH hearts demonstrated that TMZ reduced the long-chain fatty acid concentration. Additionally, TMZ alleviated PH degree and excessive strain on the right heart functions in rats with Su/Hx-induced PH. We identified the candidate target genes for TMZ treatment during PH development. Interestingly, the mRNA levels of the fatty acid transporters were substantially downregulated by TMZ administration in the lungs with Su/Hx-induced PH. Notably, TMZ suppressed excessive proliferation of human pulmonary artery smooth muscle cells under hypoxic conditions. Our study suggests that TMZ ameliorates PH development by involving energy metabolism in the lungs and heart.
Collapse
Affiliation(s)
- Asako Yanagisawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Jun-Dal Kim
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan.
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan.
| | - Akira Naito
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan.
| | - Takayuki Kobayashi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Tomoko Misawa
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Seiichiro Sakao
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Pulmonary Medicine, School of Medicine, International University of Health and Welfare, Chiba, Japan
| | - Takayuki Jujo-Sanada
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Microbial Research Center for Health and Medicine, National Institutes of Biomedical Innovation, National Institutes of Biomedical Innovation, Osaka, Japan
| | - Takeshi Kawasaki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shin-Ichi Muroi
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - So-Ichiro Sasaki
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Takuji Suzuki
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yoshimi Nakagawa
- Division of Complex Biosystem Research, Department of Research and Development, Institute of National Medicine, University of Toyama, Toyama, Japan
| | - Yoshitoshi Kasuya
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Department of Molecular and Systems Pharmacology, Faculty of Pharmacy, Juntendo University, Chiba, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
4
|
Chan YH, Yuen-Ting C, Sin CF, Ma ESK, Lam STS, Au Yeung SL, Cheung BMY, Ho CM, Yiu KH, Tse HF. Treatment with trimetazidine dihydrochloride and lung cancer survival: Implications on metabolic re-programming. Lung Cancer 2024; 197:107996. [PMID: 39490205 DOI: 10.1016/j.lungcan.2024.107996] [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: 08/30/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Metabolic re-wiring with preferential fatty acid oxidation has been observed in lung cancer cells. Whether the use of trimetazidine, an anti-anginal agent that inhibits fatty acid oxidation, alters clinical outcomes in ischemic heart disease (IHD) patients with lung cancers is unknown. METHODS We carried out this territory-wide, retrospective cohort study of 279,894 IHD patients prescribed with trimetazidine or long-acting oral nitrates in Hong Kong (population coverage of 7.5 millions, January 1999 - December 2020). A total of 6561 patients with pre-existing or de novo lung cancers were identified. Clinical outcomes of all-cause mortality were longitudinally compared between lung cancer patients who received trimetazidine (n = 547) versus non-users (control, n = 6014). RESULTS Over 902.9 ± 1410.6 days, lower incidence of deaths occurred in the trimetazidine group (79.0 %, n = 432/547) compared to controls (90.5 %, n = 5442/6014, P < 0.001). Kaplan-Meier analyses showed that trimetazidine use was associated with significantly higher survival from all-cause mortality in IHD patients (trimetazidine: mean survival = 1840.6 [95 %CI 1596.0-2085.3], versus control: 1056.7 [95 %CI 1011.3-1102.0] days, Log Rank = 69.4, P < 0.001). Cox regression showed that trimetazidine use was significantly associated with reduced risk of all-cause mortality in crude (HR = 0.60 [95 %CI: 0.53 to 0.68], P < 0.001) and multivariable models (HR = 0.65 [95 % CI: 0.57 to 0.74], P < 0.001). Pre-specified analyses amongst patients with pre-existing lung cancers yielded similar findings (HR = 0.49 [95 %CI: 0.35 to 0.67], P < 0.001). Survival benefits related to trimetazidine use was predominantly restricted to non-cardiovascular mortality (P < 0.001). CONCLUSIONS Trimetazidine use is associated with higher overall survival in IHD patients with lung cancers, particularly from non-cardiovascular death. These findings need to be confirmed by randomized controlled trials.
Collapse
Affiliation(s)
- Yap-Hang Chan
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Division of Experimental Medicine and Immunotherapeutics, Cambridge University Hospitals NHS Foundation Trust / University of Cambridge, UK; Department of Cardiology, Royal Papworth Hospital, Cambridge Biomedical Campus, UK; Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China; Genetics Research Program for Personalized Medicine in Cardiac Oncology, The University of Hong Kong, Hong Kong SAR, China.
| | - Cheng Yuen-Ting
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chun-Fung Sin
- Department of Pathology, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Edmond S K Ma
- Department of Molecular Pathology, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Stephen T S Lam
- Clinical Genetics Service, Hong Kong Sanatorium & Hospital, Hong Kong SAR, China
| | - Shiu-Lun Au Yeung
- School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Bernard M Y Cheung
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Chung-Man Ho
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kai-Hang Yiu
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, Shenzhen Hong Kong University Hospital, China; Institute of Cardiovascular Science and Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Hung-Fat Tse
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, Shenzhen Hong Kong University Hospital, China; Genetics Research Program for Personalized Medicine in Cardiac Oncology, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
5
|
Romo-Perez A, Domínguez-Gómez G, Chávez-Blanco AD, González-Fierro A, Correa-Basurto J, Dueñas-González A. PaSTe. Blockade of the Lipid Phenotype of Prostate Cancer as Metabolic Therapy: A Theoretical Proposal. Curr Med Chem 2024; 31:3265-3285. [PMID: 37287286 DOI: 10.2174/0929867330666230607104441] [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: 12/08/2022] [Revised: 04/10/2023] [Accepted: 05/09/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Prostate cancer is the most frequently diagnosed malignancy in 112 countries and is the leading cause of death in eighteen. In addition to continuing research on prevention and early diagnosis, improving treatments and making them more affordable is imperative. In this sense, the therapeutic repurposing of low-cost and widely available drugs could reduce global mortality from this disease. The malignant metabolic phenotype is becoming increasingly important due to its therapeutic implications. Cancer generally is characterized by hyperactivation of glycolysis, glutaminolysis, and fatty acid synthesis. However, prostate cancer is particularly lipidic; it exhibits increased activity in the pathways for synthesizing fatty acids, cholesterol, and fatty acid oxidation (FAO). OBJECTIVE Based on a literature review, we propose the PaSTe regimen (Pantoprazole, Simvastatin, Trimetazidine) as a metabolic therapy for prostate cancer. Pantoprazole and simvastatin inhibit the enzymes fatty acid synthase (FASN) and 3-hydroxy-3-methylglutaryl- coenzyme A reductase (HMGCR), therefore, blocking the synthesis of fatty acids and cholesterol, respectively. In contrast, trimetazidine inhibits the enzyme 3-β-Ketoacyl- CoA thiolase (3-KAT), an enzyme that catalyzes the oxidation of fatty acids (FAO). It is known that the pharmacological or genetic depletion of any of these enzymes has antitumor effects in prostatic cancer. RESULTS Based on this information, we hypothesize that the PaSTe regimen will have increased antitumor effects and may impede the metabolic reprogramming shift. Existing knowledge shows that enzyme inhibition occurs at molar concentrations achieved in plasma at standard doses of these drugs. CONCLUSION We conclude that this regimen deserves to be preclinically evaluated because of its clinical potential for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Adriana Romo-Perez
- Instituto de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | - Alma D Chávez-Blanco
- Subdirección de Investigación Básica, Instituto Nacional de Cancerologia, Mexico City, Mexico
| | - Aurora González-Fierro
- Subdirección de Investigación Básica, Instituto Nacional de Cancerologia, Mexico City, Mexico
| | - José Correa-Basurto
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alfonso Dueñas-González
- Subdirección de Investigación Básica, Instituto Nacional de Cancerologia, Mexico City, Mexico
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
6
|
Raphela-Choma P, Motadi L, Simelane M, Choene M. Anticancer Activity of Iso-Mukaadial Acetate on Pancreatic and Colon Cancer Cells. Rep Biochem Mol Biol 2024; 12:586-595. [PMID: 39086588 PMCID: PMC11288242 DOI: 10.61186/rbmb.12.4.586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 04/07/2024] [Indexed: 08/02/2024]
Abstract
Background Pancreatic cancer and colon cancer pose significant challenges in treatment, with poor prognoses. Natural products have long been explored for their potential as anticancer agents. Iso-mukaadial acetate has shown promise in inducing apoptosis in breast and ovarian cancer cells. The objective of this study was to investigate the effect of Iso-mukaadial acetate on pancreatic (MIA-PACA2) and colon (HT29) cancer cell lines. Methods Pancreatic (MIA-PACA2) cancer cells, colon (HT29) cancer cells, normal embryonic kidney cells (HEK 293), and normal lung cells (MRC5) were cultured and treated with Iso-mukaadial acetate (IMA) for 24 hours. The viability assays were conducted using Alamarblue reagent and a real-time cell viability monitoring system, xCELLigence. The IC50 values were determined, followed by assessments of ATP production, caspase 3/7 activation, mitochondrial function, morphological changes using a light microscope, and gene expression changes via RT-PCR. Results This study indicates that Iso-mukaadial acetate exhibited concentration-dependent cytotoxic effects, slowing cellular proliferation in both cancer cell lines. Activation of the mitochondrial apoptotic pathway and caspase 3/7 suggests induction of apoptosis. Reduced ATP production and altered gene expression further support its anticancer properties. Morphological changes after treatment with Iso-mukaadial acetate showed apoptotic characteristics which may suggest that apoptosis was induced. Conclusions According to the results obtained, Iso-mukaadial acetate shows potential as an anticancer agent, evidenced by its effects on cellular viability, mitochondrial function, ATP production, caspase activation, and gene expression in pancreatic and colon cancer cells. These findings highlight its promise for further investigation and potential in the development of therapeutic agents.
Collapse
Affiliation(s)
- Portia Raphela-Choma
- Department of Biochemistry, University of Johannesburg, Corner Kingsway and University Road, Auckland Park, Johannesburg, 2092, South Africa.
| | - Lesetja Motadi
- Department of Biochemistry, University of Johannesburg, Corner Kingsway and University Road, Auckland Park, Johannesburg, 2092, South Africa.
| | - Mthokosizi Simelane
- Department of Biochemistry, University of Johannesburg, Corner Kingsway and University Road, Auckland Park, Johannesburg, 2092, South Africa.
| | - Mpho Choene
- Department of Biochemistry, University of Johannesburg, Corner Kingsway and University Road, Auckland Park, Johannesburg, 2092, South Africa.
| |
Collapse
|
7
|
Hazem RM, Aboslema RF, Mehanna ET, Kishk SM, Elsayed M, El-Sayed NM. Antitumor effect of trimetazidine in a model of solid Ehrlich carcinoma is mediated by inhibition of glycolytic pathway and AKT signaling. Chem Biol Interact 2023; 383:110672. [PMID: 37591408 DOI: 10.1016/j.cbi.2023.110672] [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: 05/25/2023] [Revised: 07/18/2023] [Accepted: 08/12/2023] [Indexed: 08/19/2023]
Abstract
Disturbance in glucose metabolism was proposed to be a pathogenetic mechanism of breast cancer. Trimetazidine (TMZ) inhibits β-oxidation of fatty acids through blocking the activity of 3-ketoacylCoA thiolase enzyme, leading to enhancement of glucose oxidation and metabolic respiration. The present study aimed to examine the cytotoxic effect of TMZ in both in vivo and in vitro models of breast cancer, focusing on its impact on the expression of some glycolytic enzymes and AKT signaling. The cytotoxic effect of TMZ was screened against breast (MCF-7) cancer cell line at different concentrations [0.01-100 μM]. In vivo, graded doses (10, 20, 30 mg/kg) of TMZ were tested against solid Ehrlich carcinoma (SEC) in mice. Tumor tissues were isolated for assessment of the expression of glucose transporter-1 (GLUT-1) and glycolytic enzymes by quantitative PCR. The protein expression of AKT and cellular myelocytomatosis (c-Myc) was determined by western blotting, while p53 expression was evaluated by immunohistochemistry. Molecular docking study of TMZ effect on AKT and c-Myc was performed using Auto-Dock Vina docking program. TMZ showed a cytotoxic action against MCF-7 cells, having IC50 value of 2.95 μM. In vivo, TMZ reduced tumor weight, downregulated the expression of glycolytic enzymes, suppressed AKT signaling, but increased p53 expression. Molecular docking and in silico studies proposed that TMZ is an AKT and c-Myc selective inhibitor. In conclusion, TMZ demonstrated a viable approach to suppress tumor proliferation in biological models of breast cancer.
Collapse
Affiliation(s)
- Reem M Hazem
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Rasha F Aboslema
- The Egyptian Ministry of Health and Population, Port Said, Egypt
| | - Eman T Mehanna
- Department of Biochemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt.
| | - Safaa M Kishk
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Mohammed Elsayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
8
|
Atlı Şekeroğlu Z, Şekeroğlu V, Aydın B, Kontaş Yedier S. Cerium oxide nanoparticles exert antitumor effects and enhance paclitaxel toxicity and activity against breast cancer cells. J Biomed Mater Res B Appl Biomater 2023; 111:579-589. [PMID: 36221929 DOI: 10.1002/jbm.b.35175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/01/2022] [Accepted: 09/22/2022] [Indexed: 01/21/2023]
Abstract
Cerium oxide nanoparticles (CeONPs) displayed cytotoxic properties against some cancer cells. However, there is very limited data about the possible antitumoral potential of them in breast cancer cells when used alone and/or together with a chemotherapeutic drug. We investigated the effects of CeONPs alone or in combination with paclitaxel (PAC) on healthy or carcinoma breast cells. After human breast cancer cells (MCF-7) treated with CeONPs alone or together with PAC for 24, 48, and 72 h, the effects of CeONPs on cell viability, apoptosis, migration, and adhesion were investigated. All cell viability and IC50 values of CeONPs and PAC treatments in healthy breast cells (HTERT-HME1) were higher than MCF-7 cells. They showed higher cytotoxicity against MCF-7 cells. CeONPs (10, 20, and 30 mM) and/or abraxane (AB) (2 μM) significantly decreased cell viability values in MCF-7 cells. All CeONPs concentrations increased the number of apoptotic MCF-7 cells. CeONPs (20 and 30 mM) alone or in combination with AB for 72 h treatment also significantly increased the apoptosis in compared to AB alone. CeONPs and/or AB can significantly inhibit the migratory ability of breast cancer cells. The migration rates in co-treated groups with CeONPs and AB were lower than CeONPs treatments. Higher concentrations of CeONPs alone or together with AB inhibited cell adhesion. Our results showed CeONPs can increase cytotoxicity and apoptosis and decrease cell migration and cell adhesion when used alone or together with AB. Therefore, combination of chemotherapeutics with CeONPs may provide a good strategy against cancer.
Collapse
Affiliation(s)
- Zülal Atlı Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Vedat Şekeroğlu
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| | - Birsen Aydın
- Department of Biology, Faculty of Medicine, Faculty of Science and Letters, Amasya University, Amasya, Turkey
| | - Seval Kontaş Yedier
- Department of Molecular Biology and Genetics, Faculty of Science and Letters, Ordu University, Ordu, Turkey
| |
Collapse
|
9
|
Abstract
Glioblastoma (GBM) is a primary tumor of the brain defined by its uniform lethality and resistance to conventional therapies. There have been considerable efforts to untangle the metabolic underpinnings of this disease to find novel therapeutic avenues for treatment. An emerging focus in this field is fatty acid (FA) metabolism, which is critical for numerous diverse biological processes involved in GBM pathogenesis. These processes can be classified into four broad fates: anabolism, catabolism, regulation of ferroptosis, and the generation of signaling molecules. Each fate provides a unique perspective by which we can inspect GBM biology and gives us a road map to understanding this complicated field. This Review discusses the basic, translational, and clinical insights into each of these fates to provide a contemporary understanding of FA biology in GBM. It is clear, based on the literature, that there are far more questions than answers in the field of FA metabolism in GBM, and substantial efforts should be made to untangle these complex processes in this intractable disease.
Collapse
Affiliation(s)
| | - Navdeep S. Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
10
|
Effects of trimetazidine on anticancer activity and toxicity of abraxane in MCF-7 breast cancer cells. RENDICONTI LINCEI. SCIENZE FISICHE E NATURALI 2022. [DOI: 10.1007/s12210-022-01100-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
11
|
Engin S, Barut EN, Yaşar YK, Soysal AÇ, Arıcı T, Kerimoğlu G, Kadıoğlu M, Sezen SF. Trimetazidine attenuates cyclophosphamide-induced cystitis by inhibiting TLR4-mediated NFκB signaling in mice. Life Sci 2022; 301:120590. [PMID: 35504331 DOI: 10.1016/j.lfs.2022.120590] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/26/2022]
Abstract
AIM Cyclophosphamide (CP)-induced cystitis is a challenging clinical problem involving inflammation and dysfunction of bladder. Trimetazidine (TMZ) is an anti-anginal drug with anti-oxidant and anti-inflammatory properties. We aimed to investigate the protective effects of TMZ in CP-induced cystitis via inhibiting TLR4/NFκB signaling. MAIN METHODS Balb/c mice were administrated TMZ (10 or 20 mg/kg/day) intraperitoneally (i.p.) for 5 consecutive days before CP. On day 6, cystitis was induced by a single dose of CP (300 mg/kg, i.p.). Mesna (2-mercaptoethane sulfonate sodium; 30 mg/kg, i.p.) was administered 20 min before and at 4 and 8 h after the CP injection. After 24 h of cystitis induction, the bladders were removed for histopathological evaluation, contractility studies, biochemical analysis and western blotting. MTT assay was performed in a cancer cell line (MDA-MB-231) to evaluate the effect of TMZ on the cytotoxicity of CP. KEY FINDINGS CP-induced severe cystitis was confirmed by histological disturbances and the decrease in carbachol-evoked contractions of detrusor strips, which was partially improved by TMZ (20 mg/kg/day). SOD activity and GSH content were decreased whereas TNF-α and IL-1β levels were increased in the bladders of CP-treated mice, which were restored by TMZ or mesna. TMZ reduced the CP-induced increase in the protein expressions of caspase-3, TLR4 and phosphorylated-NFκB in bladder tissues. TMZ alone decreased the cell viability and TMZ also enhanced the cytotoxicity of CP. SIGNIFICANCE Our study provides the first preclinical evidence that TMZ attenuates CP-induced urotoxicity by enhancing anti-oxidant capacity and suppressing inflammation possibly via downregulating TLR4-mediated NFκB signaling while augmenting the cytotoxicity of CP.
Collapse
Affiliation(s)
- Seçkin Engin
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye.
| | - Elif Nur Barut
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye
| | - Yeşim Kaya Yaşar
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye; Drug and Pharmaceutical Technology Application and Research Center, Karadeniz Technical University, Trabzon, Turkiye
| | - Aysun Çelik Soysal
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Bülent Ecevit University, Zonguldak, Turkiye
| | - Tuğba Arıcı
- Başaksehir Cam and Sakura City Hospital, İstanbul, Turkiye
| | - Gökçen Kerimoğlu
- Department of Histology and Embryology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Mine Kadıoğlu
- Department of Medical Pharmacology, Faculty of Medicine, Karadeniz Technical University, Trabzon, Turkiye
| | - Sena F Sezen
- Department of Pharmacology, Faculty of Pharmacy, Karadeniz Technical University, Trabzon, Turkiye; Drug and Pharmaceutical Technology Application and Research Center, Karadeniz Technical University, Trabzon, Turkiye
| |
Collapse
|