1
|
Chang X, Qiu X, Tong X, Gan S, Yi W, Xie S, Liu X, Zuo C, Tan W. Sortilin-Mediated Rapid, Precise and Sustained Degradation of Membrane Proteins via mRNA-Encoded Lysosome-Targeting Chimera. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501222. [PMID: 40305781 DOI: 10.1002/advs.202501222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Recent advances in lysosome-targeting degradation technologies have introduced strategies to regulate therapeutic membrane proteins (MPs), potentially transforming treatment paradigms. However, challenges persist, including limited degradation precision due to the broad distribution of lysosome-targeting receptors (LTRs), as well as the high cost and complexity of recombinant protein production or chemical synthesis. Herein, it identifies sortilin as a promising LTR, highly expressed in malignancies but minimally present in healthy tissues outside the nervous system. Using AlphaFold-Multimer, it screened for a specific non-endogenous protein binder to sortilin and developed a modular, mRNA-encoded lysosomal targeting chimera (MedTAC) strategy, enabling rapid design and precise degradation of oncogenic MPs. In a breast cancer-bearing mouse model, a single low dose of MedTACPTK7 (0.5 mg kg-1) reduced protein tyrosine kinase-7 (PTK7) levels by up to 80% within 24 h, with sustained degradation of 44% at 72 h, demonstrating excellent pharmacokinetics. MedTACPTK7 significantly extended survival to over 50 days without systemic toxicity, compared to 20-30 days in controls. This MedTAC strategy establishes sortilin as a selective and efficient shuttle for targeted protein degradation, offering a scalable, rapidly producible platform for biochemical research and precise therapeutic applications.
Collapse
Affiliation(s)
- Xin Chang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xinyu Qiu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian, 350001, China
| | - Xiaoning Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Shaoju Gan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Weicheng Yi
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Sitao Xie
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Chao Zuo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Weihong Tan
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| |
Collapse
|
2
|
Hagar FF, Abbas SH, Atef E, Abdelhamid D, Abdel-Aziz M. Benzimidazole scaffold as a potent anticancer agent with different mechanisms of action (2016-2023). Mol Divers 2025; 29:1821-1849. [PMID: 39031290 PMCID: PMC11909089 DOI: 10.1007/s11030-024-10907-8] [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: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
Benzimidazole scaffolds have potent anticancer activity due to their structure similarity to nucleoside. In addition, benzimidazoles could function as hydrogen donors or acceptors and bind to different drug targets that participate in cancer progression. The literature had many anticancer agents containing benzimidazole cores that gained much interest. Provoked by our endless interest in benzimidazoles as anticancer agents, we summarized the successful trials of the benzimidazole scaffolds in this concern. Moreover, we discuss the substantial opportunities in cancer treatment using benzimidazole-based drugs that may direct medicinal chemists for a compelling future design of more active chemotherapeutic agents with potential clinical applications. The uniqueness of this work lies in the highlighted benzimidazole scaffold hybridization with different molecules and benzimidazole-metal complexes, detailed mechanisms of action, and the IC50 of the developed compounds determined by different laboratories after 2015.
Collapse
Affiliation(s)
- Fatma Fouad Hagar
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman Atef
- College of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Dalia Abdelhamid
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
- Raabe College of Pharmacy, Ohio Northern University, Ohio, USA.
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
3
|
Gupta R, Roy D, Ghosh A, Begum Y, Ghosh D, Swarnakar S. Mebendazole Exerts Anticancer Activity in Ovarian Cancer Cell Lines via Novel Girdin-Mediated AKT/IKKα/β/NF-κB Signaling Axis. Cells 2025; 14:113. [PMID: 39851541 PMCID: PMC11763501 DOI: 10.3390/cells14020113] [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: 12/14/2024] [Revised: 01/02/2025] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Mebendazole (MBZ), a benzimidazole anthelmintic and cytoskeleton-disrupting compound, exhibits antitumor properties; however, its action on ovarian cancer (OC) is not clearly understood. This study evaluates the effect of MBZ on OC cell lines OVCAR3 and OAW42, focusing on cell proliferation, migration, invasion, and cancer stemness. The underlying mechanisms, including cytoskeletal disruption, epithelial-mesenchymal transition (EMT), and signaling pathways, were explored. MBZ inhibited OVCAR3 and OAW42 cell proliferation in a dose- and time-dependent manner. Additionally, MBZ significantly impedes migration, spheroid invasion, colony formation, and stemness. In addition, it reduced actin polymerization and down-regulated CSC markers (e.g., CD24, CD44, EpCAM). Moreover, MBZ suppressed MMP-9 activity and inhibited the EMT marker as judged by decreased N-Cadherin and Vimentin and increased E-Cadherin. Furthermore, MBZ induced G2/M cell cycle arrest by modulating Cyclin B1, CDC25C, and WEE1. Also, it triggered apoptosis by disrupting mitochondrial membrane potential. Mechanistic studies revealed a significant downregulation of Girdin, an Akt modulator, along with reduced p-Akt, p-IKKα/β, and p-NF-κB, indicating MBZ's novel mechanism of action through the Girdin-mediated Akt/IKKα/β/NF-κB signaling axis. Thus, by targeting Girdin, MBZ presents a promising repurposed therapeutic strategy to inhibit cancer cell proliferation and metastasis in ovarian cancer.
Collapse
Affiliation(s)
- Rahul Gupta
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| | - Dipanjan Roy
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India;
| | - Arijit Ghosh
- Department of Molecular Biology, Netaji Subhash Chandra Bose Cancer Research Institute, Kolkata 700094, India;
| | - Yasmin Begum
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| | - Dipanjan Ghosh
- Department of Natural Products, National Institute of Pharmaceutical Education and Research, Kolkata 700054, India;
| | - Snehasikta Swarnakar
- Infectious Diseases & Immunology Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology, Jadavpur, Kolkata 700032, India or (R.G.); or (Y.B.)
| |
Collapse
|
4
|
Łukowska-Chojnacka E, Fedorov E, Kowalkowska A, Wielechowska M, Sobiepanek A, Koronkiewicz M, Wińska P. Synthesis and evaluation of anticancer activity of new 4,5,6,7-tetrabromo-1H-benzimidazole derivatives. Bioorg Chem 2024; 153:107880. [PMID: 39476601 DOI: 10.1016/j.bioorg.2024.107880] [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: 07/16/2024] [Revised: 09/16/2024] [Accepted: 10/05/2024] [Indexed: 12/10/2024]
Abstract
An efficient method for the synthesis of new 4,5,6,7-tetrabromo-1H-benzimidazole derivatives has been developed. New ketones were obtained by N-alkylation of TBBi or 2-Me-TBBi with various phenacyl halides and then reduced to the corresponding alcohols. All compounds were obtained with satisfactory yields in the range of 40-91 %. The synthesized compounds appeared a weak CK2 and PIM-1 inhibitors but exhibit an interesting cytotoxic activity against cancer cell lines, i.e. MCF-7, PC-3, CCRF-CEM, K-562. 1-Phenyl-2-(4,5,6,7-tetrabromo-1H-benzimidazol-1-yl)ethanone 3aA exhibits the highest cytotoxic activity with IC50 value of 5.30 µM for MCF-7 and 6.80 µM for CCRF-CEM. Moreover, this compound shows the highest selectivity against both MCF-7 and CCRF-CEM with SI selectivity coefficients (against MRC-5 and Vero cells) equal 5.45 and 4.30 for MCF-7 and 4.25 and 3.35 for CCRF-CEM, respectively. Furthermore, it was shown that compound 3aA exhibits very good pro-apoptotic properties, through induction of the mitochondrial apoptotic pathway in CCRF-CEM cells. These results correlate with data showing the effect of 3aA on intracellular level of CK2α protein and CK2-mediated phosphorylation of Ser529 in NF-κBp65. Study of the effect of compound 3aA on mRNA levels of CK2α and CK2α' showed no significant differences in gene expression levels in control CCRF-CEM and cells treated with 3aA, indicating 3aA action at the protein level.
Collapse
Affiliation(s)
- Edyta Łukowska-Chojnacka
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland.
| | - Egor Fedorov
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Anna Kowalkowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Monika Wielechowska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Anna Sobiepanek
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| | - Mirosława Koronkiewicz
- Department of Biomedical Research, National Medicines Institute, Chełmska St. 30/34, 00-725 Warsaw, Poland
| | - Patrycja Wińska
- Faculty of Chemistry, Warsaw University of Technology, Noakowskiego St. 3, 00-664 Warsaw, Poland
| |
Collapse
|
5
|
Lubau NSA, Chengebroyen N, Subramaniyan V. Investigation of Uncovering Molecular Mechanisms of Alcohol-Induced Female Infertility-A Rational Approach. Reprod Sci 2024; 31:3660-3672. [PMID: 39485609 PMCID: PMC11611948 DOI: 10.1007/s43032-024-01692-8] [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: 04/11/2024] [Accepted: 09/06/2024] [Indexed: 11/03/2024]
Abstract
This study aimed to investigate the molecular mechanisms by which chronic alcohol consumption impacts female infertility, highlighting significant societal implications. By conducting a comprehensive literature review, we examined existing evidence on the association between long-term alcohol use and female reproductive health. Relevant studies were identified through systematic searches of electronic databases and key journals. We synthesized information on the molecular pathways affected by alcohol consumption, with particular emphasis on oxidative stress, inflammation, and hormonal disruptions. Additionally, we reviewed efforts to address alcohol-related health issues, including public health interventions, regulatory measures, and educational initiatives. Our study found strong evidence linking chronic alcohol consumption to increased mortality rates and a range of preventable diseases globally. Alcohol's effects extend beyond physiological consequences to psychological, social, and economic burdens. Chronic alcohol consumption disrupts hormonal balance and reproductive function, contributing to female infertility. Future research should focus on quantifying mortality risks associated with alcohol consumption, understanding gender-specific patterns in alcohol-related health outcomes, and elucidating the molecular mechanisms underlying female infertility. Addressing these gaps will inform strategies to mitigate the burden of alcohol-induced health issues and promote overall well-being. Collaborative efforts among diverse stakeholders are essential for advancing research agendas and translating findings into effective interventions.
Collapse
Affiliation(s)
- Natasha Sura Anak Lubau
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Neevashini Chengebroyen
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Vetriselvan Subramaniyan
- Pharmacology Unit, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia.
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan University, Bandar Sunway, 47500, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
6
|
Zhang Q, Qian F, Cai M, Liu R, Chen M, Li Z, Chen Y, Lu N. Research progress on factors affecting the sensitivity of breast cancer to radiotherapy: a narrative review. Transl Cancer Res 2024; 13:3869-3888. [PMID: 39145054 PMCID: PMC11319974 DOI: 10.21037/tcr-24-71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024]
Abstract
Background and Objective Radiation therapy (RT) is one of the important components of comprehensive treatment for breast cancer and has important value in improving the control rate of local areas, reducing the chance of recurrence and metastasis after breast cancer surgery, delaying disease progression, and improving the survival of breast cancer patients. The factors that affect the RT sensitivity of breast cancer are important. The above potential predictors of radiation efficacy can provide patients with a predictive method and therefore have significant value in clinical therapy. In this paper, we have summarised the predictive factors of radiotherapy sensitivity by reviewing recent research on breast cancer and focused on the following areas: tumor immune microenvironment (TIME), cancer stem cells, noncoding RNAs, signal transduction pathways, genes, etc. This review aims to provide theoretical basis and reference for improving the efficacy of radiotherapy and experimental individualized treatment of breast cancer. Methods We searched the Web of Science database to identify clinical studies published between 2010 and January 2024 that investigated radiotherapy sensitivity. The main findings of the validated studies were summarised. Key Content and Findings Improving the radiosensitivity of breast cancer is essential in the treatment of breast cancer. The radiosensitivity can be improved by modulating immune cells or immunomodulatory factors in the TIME, modulating signal transduction pathways, and other innovative combination therapy strategies. And we also summarized the predictive markers of breast cancer radiosensitivity. Conclusions In this paper, we reviewed the literature and summarized the newest research advances on the radiosensitivity of breast cancer patients. This review paper includes the following six aspects: the immune microenvironment, tumor stem cells, signaling pathways, regulation of gene/protein expression, small molecule drugs, and predictive markers for radiosensitivity.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Oncology, Affiliated Anhui Provincial Hospital, Bengbu Medical College, Bengbu, China
| | - Fusheng Qian
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Mengjie Cai
- Department of Oncology, Affiliated Anhui Provincial Hospital, Bengbu Medical College, Bengbu, China
| | - Ruijie Liu
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Manping Chen
- Department of Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Zhitong Li
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Ying Chen
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Nannan Lu
- Department of Oncology, Affiliated Anhui Provincial Hospital, Bengbu Medical College, Bengbu, China
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, China
| |
Collapse
|
7
|
Brownjohn PW, Zoufir A, O’Donovan DJ, Sudhahar S, Syme A, Huckvale R, Porter JR, Bange H, Brennan J, Thompson NT. Computational drug discovery approaches identify mebendazole as a candidate treatment for autosomal dominant polycystic kidney disease. Front Pharmacol 2024; 15:1397864. [PMID: 38846086 PMCID: PMC11154008 DOI: 10.3389/fphar.2024.1397864] [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: 03/08/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a rare genetic disorder characterised by numerous renal cysts, the progressive expansion of which can impact kidney function and lead eventually to renal failure. Tolvaptan is the only disease-modifying drug approved for the treatment of ADPKD, however its poor side effect and safety profile necessitates the need for the development of new therapeutics in this area. Using a combination of transcriptomic and machine learning computational drug discovery tools, we predicted that a number of existing drugs could have utility in the treatment of ADPKD, and subsequently validated several of these drug predictions in established models of disease. We determined that the anthelmintic mebendazole was a potent anti-cystic agent in human cellular and in vivo models of ADPKD, and is likely acting through the inhibition of microtubule polymerisation and protein kinase activity. These findings demonstrate the utility of combining computational approaches to identify and understand potential new treatments for traditionally underserved rare diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Hester Bange
- Crown Bioscience Netherlands B.V., Biopartner Center Leiden JH, Leiden, Netherlands
| | | | | |
Collapse
|
8
|
Rodrigues AJ, Chernikova SB, Wang Y, Trinh TTH, Solow-Cordero DE, Alexandrova L, Casey KM, Alli E, Aggarwal A, Quill T, Koegel AK, Feldman BJ, Ford JM, Hayden-Gephart M. Repurposing mebendazole against triple-negative breast cancer CNS metastasis. J Neurooncol 2024; 168:125-138. [PMID: 38563850 PMCID: PMC11093727 DOI: 10.1007/s11060-024-04654-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: 01/31/2024] [Accepted: 03/20/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) often metastasizes to the central nervous system (CNS) and has the highest propensity among breast cancer subtypes to develop leptomeningeal disease (LMD). LMD is a spread of cancer into leptomeningeal space that speeds up the disease progression and severely aggravates the prognosis. LMD has limited treatment options. We sought to test whether the common anti-helminthic drug mebendazole (MBZ) may be effective against murine TNBC LMD. METHODS A small-molecule screen involving TNBC cell lines identified benzimidazoles as potential therapeutic agents for further study. In vitro migration assays were used to evaluate cell migration capacity and the effect of MBZ. For in vivo testing, CNS metastasis was introduced into BALB/c athymic nude mice through internal carotid artery injections of brain-tropic MDA-MB-231-BR or MCF7-BR cells. Tumor growth and spread was monitored by bioluminescence imaging and immunohistochemistry. MBZ was given orally at 50 and 100 mg/kg doses. MBZ bioavailability was assayed by mass spectrometry. RESULTS Bioinformatic analysis and migration assays revealed higher migratory capacity of TNBC compared to other breast cancer subtypes. MBZ effectively slowed down migration of TNBC cell line MDA-MB-231 and its brain tropic derivative MDA-MB-231-BR. In animal studies, MBZ reduced leptomeningeal spread, and extended survival in brain metastasis model produced by MDA-MB-231-BR cells. MBZ did not have an effect in the non-migratory MCF7-BR model. CONCLUSIONS We demonstrated that MBZ is a safe and effective oral agent in an animal model of TNBC CNS metastasis. Our findings are concordant with previous efforts involving MBZ and CNS pathology and support the drug's potential utility to slow down leptomeningeal spread.
Collapse
Affiliation(s)
- Adrian J Rodrigues
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA, 94305, USA
- Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Sophia B Chernikova
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA, 94305, USA.
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Thy T H Trinh
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - David E Solow-Cordero
- High-Throughput Screening Knowledge Center, Sarafan ChEM-H, Stanford, CA, 94305, USA
| | - Ludmila Alexandrova
- Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University, Stanford, CA, 94305, USA
| | - Kerriann M Casey
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Elizabeth Alli
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC, 27109, USA
| | | | - Tyler Quill
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Ashley K Koegel
- Department of Pediatric Hematology-Oncology, University of California, San Francisco, CA, 94143, USA
| | - Brian J Feldman
- Department of Pediatrics, University of California, San Francisco, CA, 94143, USA
| | - James M Ford
- Department of Medicine (Oncology), Stanford School of Medicine, Stanford, CA, 94305, USA
| | | |
Collapse
|
9
|
Malla R, Viswanathan S, Makena S, Kapoor S, Verma D, Raju AA, Dunna M, Muniraj N. Revitalizing Cancer Treatment: Exploring the Role of Drug Repurposing. Cancers (Basel) 2024; 16:1463. [PMID: 38672545 PMCID: PMC11048531 DOI: 10.3390/cancers16081463] [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: 03/02/2024] [Revised: 04/04/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Cancer persists as a global challenge necessitating continual innovation in treatment strategies. Despite significant advancements in comprehending the disease, cancer remains a leading cause of mortality worldwide, exerting substantial economic burdens on healthcare systems and societies. The emergence of drug resistance further complicates therapeutic efficacy, underscoring the urgent need for alternative approaches. Drug repurposing, characterized by the utilization of existing drugs for novel clinical applications, emerges as a promising avenue for addressing these challenges. Repurposed drugs, comprising FDA-approved (in other disease indications), generic, off-patent, and failed medications, offer distinct advantages including established safety profiles, cost-effectiveness, and expedited development timelines compared to novel drug discovery processes. Various methodologies, such as knowledge-based analyses, drug-centric strategies, and computational approaches, play pivotal roles in identifying potential candidates for repurposing. However, despite the promise of repurposed drugs, drug repositioning confronts formidable obstacles. Patenting issues, financial constraints associated with conducting extensive clinical trials, and the necessity for combination therapies to overcome the limitations of monotherapy pose significant challenges. This review provides an in-depth exploration of drug repurposing, covering a diverse array of approaches including experimental, re-engineering protein, nanotechnology, and computational methods. Each of these avenues presents distinct opportunities and obstacles in the pursuit of identifying novel clinical uses for established drugs. By examining the multifaceted landscape of drug repurposing, this review aims to offer comprehensive insights into its potential to transform cancer therapeutics.
Collapse
Affiliation(s)
- RamaRao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam 530045, Andhra Pradesh, India
| | - Sathiyapriya Viswanathan
- Department of Biochemistry, ACS Medical College and Hospital, Chennai 600007, Tamil Nadu, India;
| | - Sree Makena
- Maharajah’s Institute of Medical Sciences and Hospital, Vizianagaram 535217, Andhra Pradesh, India
| | - Shruti Kapoor
- Department of Genetics, University of Alabama, Birmingham, AL 35233, USA
| | - Deepak Verma
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | - Manikantha Dunna
- Center for Biotechnology, Jawaharlal Nehru Technological University, Hyderabad 500085, Telangana, India
| | - Nethaji Muniraj
- Center for Cancer and Immunology Research, Children’s National Hospital, 111, Michigan Ave NW, Washington, DC 20010, USA
| |
Collapse
|
10
|
Rodrigues A, Chernikova SB, Wang Y, Trinh TTH, Solow-Cordero DE, Alexandrova L, Casey KM, Alli E, Aggarwal A, Quill T, Koegel A, Feldman BJ, Ford JM, Hayden-Gephart M. Repurposing mebendazole against triple-negative breast cancer leptomeningeal disease. RESEARCH SQUARE 2024:rs.3.rs-3915392. [PMID: 38405839 PMCID: PMC10889063 DOI: 10.21203/rs.3.rs-3915392/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Purpose Triple-negative breast cancer (TNBC) is an aggressive subtype that often metastasizes to the brain. Leptomeningeal disease (LMD), a devastating brain metastasis common in TNBC, has limited treatment options. We sought to test whether the common anti-helminthic drug mebendazole (MBZ) may be effective against murine TNBC LMD. Methods A small-molecule screen involving TNBC cell lines identified benzimidazoles as potential therapeutic agents for further study. In vitro migration assays were used to evaluate cell migration capacity and the effect of MBZ. For in vivo testing, LMD was introduced into BALB/c athymic nude mice through internal carotid artery injections of brain-tropic MDA-MB-231-BR or MCF7-BR cells. Tumor growth and spread was monitored by bioluminescence imaging. MBZ was given orally at 50 and 100 mg/kg doses. MBZ bioavailability was assayed by mass spectrometry. Results Bioinformatic analysis and migration assays revealed higher migratory capacity of TNBC compared to other breast cancer subtypes. MBZ effectively slowed down migration of TNBC cell line MDA-MB-231 and its brain tropic derivative MDA-MB-231-BR. In animal studies, MBZ reduced tumor growth and extended survival in the LMD model produced by MDA-MB-231-BR cells. MBZ did not have an effect in the non-migratory MCF7-BR model. Conclusions We demonstrated that MBZ is a safe and effective oral agent in an animal model of TNBC LMD. Our findings are concordant with previous efforts involving MBZ and central nervous system pathology and further support the drug's potential utility as an alternative therapeutic for TNBC LMD.
Collapse
Affiliation(s)
| | | | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Thy T H Trinh
- Department of Neurosurgery, Stanford School of Medicine, Stanford, CA 94305
| | | | - Ludmila Alexandrova
- Vincent Coates Foundation Mass Spectrometry Laboratory, Stanford University, Stanford, CA, 94305
| | - Kerriann M Casey
- Department of Comparative Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth Alli
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, NC 27109
| | - Abhishek Aggarwal
- High-Throughput Screening Knowledge Center, Sarafan ChEM-H, Stanford CA 94305
| | - Tyler Quill
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305
| | - Ashley Koegel
- Department of Pediatric Hematology-Oncology, University of California, San Francisco, CA 94143
| | - Brian J Feldman
- Department of Pediatrics, University of California, San Francisco, CA 94143
| | - James M Ford
- Department of Medicine (Oncology), Stanford School of Medicine, Stanford, CA 94305
| | | |
Collapse
|
11
|
Akar S, Cakir M, Ozkol H, Akkoc S, Ozdem B. A benzimidazolium salt induces apoptosis and arrests cells at sub-G1 phase in epithelial ovarian cancer cells. Mol Biol Rep 2024; 51:66. [PMID: 38170294 DOI: 10.1007/s11033-023-08981-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/24/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Ovarian cancer, also known as a silent killer, is the deadliest gynecological cancer in women worldwide. Epithelial ovarian cancers constitute the majority of ovarian cancers, and diagnosis can be made in advanced stages, which greatly reduces the likelihood of treatment and lowers the survival rate. For the treatment of epithelial ovarian cancers, the search for synthetic agents as well as agents of natural origin continues. The effects of 1-(2-cyanobenzyl)-3-(4-vinylbenzyl)-1H-benzo[d]imidazole-3-ium chloride (BD), a benzimidazole derivative, were investigated on epithelial ovarian cancer cells. METHODS AND RESULTS In our study, the effects of BD on proliferation, colony formation, cell death by apoptosis and the cell cycle in A2780 and A2780 Adriamycin (ADR) ovarian cancer cell lines were investigated. Proliferation was examined with cell viability analysis, colony formation and apoptosis with Annexin V staining and cell cycle analyses with PI staining, respectively. As a result of the analyses, BD inhibited cell proliferation and colony formation, induced apoptosis and cell death at 48 h in A2780 and A2780 ADR cells at 10.10 and 10.36 µM concentrations, respectively. In addition, A2780 and A2780ADR cells were arrested in the Sub-G1 phase of the cell cycle. CONCLUSIONS BD suppresses cancer cell progression by showing antiproliferative effects on ovarian cancer cells. Further analyses are required to determine the mechanism of action of this agent and to demonstrate its potential as a suitable candidate for the treatment of epithelial ovarian cancer.
Collapse
Affiliation(s)
- Sakine Akar
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey.
| | - Mustafa Cakir
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey
| | - Halil Ozkol
- Department of Medical Biology, Faculty of Medicine, Van Yuzuncu Yil University, Van, 65090, Turkey
| | - Senem Akkoc
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
- Faculty of Engineering and Natural Sciences, Bahcesehir University, Istanbul, Turkey
| | - Berna Ozdem
- Department of Medical Biology and Genetics, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
12
|
Gutiérrez EL, Godoy AA, Brusau EV, Vega D, Narda GE, Suárez S, Di Salvo F. Mebendazolium mesylate anhydride salt: rational design based on supramolecular assembly, synthesis, and solid-state characterization. RSC Adv 2024; 14:181-192. [PMID: 38173618 PMCID: PMC10759312 DOI: 10.1039/d3ra07422f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
The design mebendazole (MBZ) multicomponent systems is important to obtain new materials that incorporate the API (active pharmaceutical ingredient) with better thermal stability, avoiding the interconversion of desmotropes. Interestingly, the presence of water molecules in the mebendazolium mesylate monohydrate prevents the formation of the R22(8) supramolecular synthon, found in all mebendazolium salts with polyatomic counterions. Here, we designed a new mebendazolium mesylate anhydrous salt based on statistical scrutiny of all mebendazole crystal structures identified in the literature and an exhaustive analysis of the conformational and geometrical requirements for the supramolecular assembly. The synthesis of this new salt and its solid-state characterization through single-crystal X-ray diffraction and complementary techniques are presented. As expected, mebendazole recrystallization in methanol with methanesulfonic acid - a Food and Drug Administration accepted coformer - in the absence of water yields a mesylate anhydrous salt with 1 : 1 stoichiometry. This new salt crystallizes in the P212121 (19) space group. The main intermolecular interactions found in the crystal structure are the hydrogen bonds that form a R22(8) supramolecular motif that assembles the ionic pairs. Additional non-classical H-bond, as well as π⋯π and carbonyl⋯cation interactions, contribute to the final stabilization of the crystal packing. This new salt is stable up to 205 °C when it undergoes the endothermic loss of the ester moiety to yield 2-amino-5-benzoylbenzimidazole. Moreover, preliminary dissolution experiments in aqueous 0.1 mol L-1 HCl suggest an apparent solubility of mebendazolium mesylate anhydride 2.67 times higher than that of the preferred for pharmaceutical formulations MBZ form C.
Collapse
Affiliation(s)
- Eduardo L Gutiérrez
- INQUISAL-CONICET, Área de Química Física, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Chacabuco y Pedernera CP 5700 San Luis Argentina
| | - Agustín A Godoy
- Instituto de Investigaciones en Tecnología Química (INTEQUI), Área de Química General e Inorgánica "Dr G. F. Puelles", Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis Almte. Brown 1500-1402, D5700APA, Chacabuco y Pedernera CP 5700 San Luis Argentina
| | - Elena V Brusau
- Instituto de Investigaciones en Tecnología Química (INTEQUI), Área de Química General e Inorgánica "Dr G. F. Puelles", Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis Almte. Brown 1500-1402, D5700APA, Chacabuco y Pedernera CP 5700 San Luis Argentina
| | - Daniel Vega
- Gerencia de Investigación y Aplicaciones, Centro Atómico Constituyentes, Comisión Nacional de Energía Atómica Av. Gral. Paz 1499, 1650 San Martín Buenos Aires Argentina
| | - Griselda E Narda
- Instituto de Investigaciones en Tecnología Química (INTEQUI), Área de Química General e Inorgánica "Dr G. F. Puelles", Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis Almte. Brown 1500-1402, D5700APA, Chacabuco y Pedernera CP 5700 San Luis Argentina
| | - Sebastián Suárez
- INQUIMAE-CONICET y Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CABA Argentina
| | - Florencia Di Salvo
- INQUIMAE-CONICET y Departamento de Química Inorgánica, Analítica y Química Física, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, CABA Argentina
| |
Collapse
|
13
|
Servidei T, Sgambato A, Lucchetti D, Navarra P, Ruggiero A. Drug Repurposing in Pediatric Brain Tumors: Posterior Fossa Ependymoma and Diffuse Midline Glioma under the Looking Glass. FRONT BIOSCI-LANDMRK 2023; 28:77. [PMID: 37114548 DOI: 10.31083/j.fbl2804077] [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: 12/23/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
Tumors of the Central Nervous System (CNS) represent the leading cause of cancer-related deaths in children. Current treatment options are not curative for most malignant histologies, and intense preclinical and clinical research is needed to develop more effective therapeutic interventions against these tumors, most of which meet the FDA definition for orphan diseases. Increased attention is being paid to the repositioning of already-approved drugs for new anticancer indications as a fast-tracking strategy for identifying new and more effective therapies. Two pediatric CNS tumors, posterior fossa ependymoma (EPN-PF) type A and diffuse midline glioma (DMG) H3K27-altered, share loss of H3K27 trimethylation as a common epigenetic hallmark and display early onset and poor prognosis. These features suggest a potentially common druggable vulnerability. Successful treatment of these CNS tumors raises several challenges due to the location of tumors, chemoresistance, drug blood-brain barrier penetration, and the likelihood of adverse side effects. Recently, increasing evidence demonstrates intense interactions between tumor cell subpopulations and supportive tumor microenvironments (TMEs) including nerve, metabolic, and inflammatory TMEs. These findings suggest the use of drugs, and/or multi-drug combinations, that attack both tumor cells and the TME simultaneously. In this work, we present an overview of the existing evidence concerning the most preclinically validated noncancer drugs with antineoplastic activity. These drugs belong to four pharmacotherapeutic classes: antiparasitic, neuroactive, metabolic, and anti-inflammatory. Preclinical evidence and undergoing clinical trials in patients with brain tumors, with special emphasis on pediatric EPN-PF and DMG, are summarized and critically discussed.
Collapse
Affiliation(s)
- Tiziana Servidei
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Donatella Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore -- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
14
|
Lee YT, Tan YJ, Oon CE. Benzimidazole and its derivatives as cancer therapeutics: The potential role from traditional to precision medicine. Acta Pharm Sin B 2023; 13:478-497. [PMID: 36873180 PMCID: PMC9978992 DOI: 10.1016/j.apsb.2022.09.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/11/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Cancer is the second leading cause of mortality globally which remains a continuing threat to human health today. Drug insensitivity and resistance are critical hurdles in cancer treatment; therefore, the development of new entities targeting malignant cells is considered a high priority. Targeted therapy is the cornerstone of precision medicine. The synthesis of benzimidazole has garnered the attention of medicinal chemists and biologists due to its remarkable medicinal and pharmacological properties. Benzimidazole has a heterocyclic pharmacophore, which is an essential scaffold in drug and pharmaceutical development. Multiple studies have demonstrated the bioactivities of benzimidazole and its derivatives as potential anticancer therapeutics, either through targeting specific molecules or non-gene-specific strategies. This review provides an update on the mechanism of actions of various benzimidazole derivatives and the structure‒activity relationship from conventional anticancer to precision healthcare and from bench to clinics.
Collapse
Affiliation(s)
- Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Minden, Penang, Malaysia
| |
Collapse
|
15
|
Meco D, Attinà G, Mastrangelo S, Navarra P, Ruggiero A. Emerging Perspectives on the Antiparasitic Mebendazole as a Repurposed Drug for the Treatment of Brain Cancers. Int J Mol Sci 2023; 24:1334. [PMID: 36674870 PMCID: PMC9862092 DOI: 10.3390/ijms24021334] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/02/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
Repurposing approved non-antitumor drugs is a promising and affordable strategy in drug discovery to identify new therapeutic uses different from the original medical indication that may help increase the number of possible, effective anticancer drugs. The use of drugs in ways other than their original FDA-approved indications could offer novel avenues such as bypassing the chemoresistance and recurrence seen with conventional therapy and treatment; moreover, it can offer a safe and economic strategy for combination therapy. Recent works have demonstrated the anticancer properties of the FDA-approved drug Mebendazole. This synthetic benzimidazole proved effective against a broad spectrum of intestinal Helminthiasis. Mebendazole can penetrate the blood-brain barrier and has been shown to inhibit the malignant progression of glioma by targeting signaling pathways related to cell proliferation, apoptosis, or invasion/migration, or by increasing the sensitivity of glioma cells to conventional chemotherapy or radiotherapy. Moreover, several preclinical models and ongoing clinical trials explore the efficacy of Mebendazole in multiple cancers, including acute myeloid leukemia, brain cancer, oropharyngeal squamous cell carcinoma, breast cancer, gastrointestinal cancer, lung carcinoma, adrenocortical carcinoma, prostate cancer, and head and neck cancer. The present review summarizes central literature regarding the anticancer effects of MBZ in cancer cell lines, animal tumor models, and clinical trials to suggest possible strategies for safe and economical combinations of anticancer therapies in brain cancer. Mebendazole might be an excellent candidate for the treatment of brain tumors because of its efficacy both when used as monotherapy and in combination as an enhancement to standard chemotherapeutics and radiotherapy, due to its effectiveness on tumor angiogenesis inhibition, cell cycle arrest, apoptosis induction, and targeting of critical pathways involved in cancer such as Hedgehog signaling. Therefore, attention to MBZ repurposing has recently increased because of its potential therapeutic versatility and significant clinical implications, such as reducing medical care costs and optimizing existing therapies. Using new treatments is essential, particularly when current therapeutics for patients with brain cancer fail.
Collapse
Affiliation(s)
- Daniela Meco
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Giorgio Attinà
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Stefano Mastrangelo
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
16
|
Choi HS, Ko YS, Jin H, Kang KM, Ha IB, Jeong H, Lee JH, Jeong BK, Kim HJ. Mebendazole Increases Anticancer Activity of Radiotherapy in Radiotherapy-Resistant Triple-Negative Breast Cancer Cells by Enhancing Natural Killer Cell-Mediated Cytotoxicity. Int J Mol Sci 2022; 23:15493. [PMID: 36555137 PMCID: PMC9779603 DOI: 10.3390/ijms232415493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is the most commonly diagnosed cancer worldwide and ranks first in terms of both prevalence and cancer-related mortality in women. In this study, we aimed to evaluate the anticancer effect of mebendazole (MBZ) and radiotherapy (RT) concomitant use in triple-negative breast cancer (TNBC) cells and elucidate the underlying mechanisms of action. Breast cancer mouse models and several types of breast cancer cells, including TNBC-derived RT-resistant (RT-R) MDA-MB-231 cells, were treated with MBZ and/or RT. In mice, changes in body weight, renal and liver toxicity, tumor volume, and number of lung metastases were determined. In cells, cell viability, colony formation, scratch wound healing, Matrigel invasion, and protein expression using western blotting were determined. Our findings showed that MBZ and RT combined treatment increased the anticancer effect of RT without additional toxicity. In addition, we noted that cyclin B1, PH2AX, and natural killer (NK) cell-mediated cytotoxicity increased following MBZ + RT treatment compared to unaided RT. Our results suggest that MBZ + RT have an enhanced anticancer effect in TNBC which acquires radiation resistance through blocking cell cycle progression, initiating DNA double-strand breaks, and promoting NK cell-mediated cytotoxicity.
Collapse
Affiliation(s)
- Hoon Sik Choi
- Department of Radiation Oncology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - Young Shin Ko
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pharmacology, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Hana Jin
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pharmacology, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Ki Mun Kang
- Department of Radiation Oncology, Gyeongsang National University Changwon Hospital, Gyeongsang National University College of Medicine, Changwon 51472, Republic of Korea
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
| | - In Bong Ha
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Hojin Jeong
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Jeong-hee Lee
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pathology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Bae Kwon Jeong
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju 52727, Republic of Korea
- Department of Radiation Oncology, Gyeongsang National University Hospital, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| | - Hye Jung Kim
- Institute of Health Science, Gyeongsang National University, Jinju 52727, Republic of Korea
- Department of Pharmacology, Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
- Department of Convergence Medical Science (BK21 Plus), Gyeongsang National University College of Medicine, Jinju 52727, Republic of Korea
| |
Collapse
|
17
|
Gales L, Forsea L, Mitrea D, Stefanica I, Stanculescu I, Mitrica R, Georgescu M, Trifanescu O, Anghel R, Serbanescu L. Antidiabetics, Anthelmintics, Statins, and Beta-Blockers as Co-Adjuvant Drugs in Cancer Therapy. Medicina (B Aires) 2022; 58:medicina58091239. [PMID: 36143915 PMCID: PMC9503803 DOI: 10.3390/medicina58091239] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/29/2022] [Accepted: 09/02/2022] [Indexed: 11/25/2022] Open
Abstract
Over the last years, repurposed agents have provided growing evidence of fast implementation in oncology treatment such as certain antimalarial, anthelmintic, antibiotics, anti-inflammatory, antihypertensive, antihyperlipidemic, antidiabetic agents. In this study, the four agents of choice were present in our patients’ daily treatment for nonmalignant-associated pathology and have known, light toxicity profiles. It is quite common for a given patient’s daily administration schedule to include two or three of these drugs for the duration of their treatment. We chose to review the latest literature concerning metformin, employed as a first-line treatment for type 2 diabetes; mebendazole, as an anthelmintic; atorvastatin, as a cholesterol-lowering drug; propranolol, used in cardiovascular diseases as a nonspecific inhibitor of beta-1 and beta-2 adrenergic receptors. At the same time, certain key action mechanisms make them feasible antitumor agents such as for mitochondrial ETC inhibition, activation of the enzyme adenosine monophosphate-activated protein kinase, amelioration of endogenous hyperinsulinemia, inhibition of selective tyrosine kinases (i.e., VEGFR2, TNIK, and BRAF), and mevalonate pathway inhibition. Despite the abundance of results from in vitro and in vivo studies, the only solid data from randomized clinical trials confirm metformin-related oncological benefits for only a small subset of nondiabetic patients with HER2-positive breast cancer and early-stage colorectal cancer. At the same time, clinical studies confirm metformin-related detrimental/lack of an effect for lung, breast, prostate cancer, and glioblastoma. For atorvastatin we see a clinical oncological benefit in patients and head and neck cancer, with a trend towards radioprotection of critical structures, thus supporting the role of atorvastatin as a promising agent for concomitant association with radiotherapy. Propranolol-related increased outcomes were seen in clinical studies in patients with melanoma, breast cancer, and sarcoma.
Collapse
Affiliation(s)
- Laurentia Gales
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Oncology, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Leyla Forsea
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Diana Mitrea
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Irina Stefanica
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Irina Stanculescu
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Radu Mitrica
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
- Correspondence: ; Tel.: +40-741-964-311
| | - Mihai Georgescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Oana Trifanescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Rodica Anghel
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| | - Luiza Serbanescu
- Department of Oncology, “Carol Davila” University of Medicine & Pharmacy, 022328 Bucharest, Romania
- Department of Radiotherapy, “Prof. Dr. Alexandru Trestioreanu” Institute of Oncology, 022328 Bucharest, Romania
| |
Collapse
|
18
|
Basha NJ. Therapeutic Efficacy of Benzimidazole and Its Analogs: An Update. Polycycl Aromat Compd 2022. [DOI: 10.1080/10406638.2022.2118334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- N. Jeelan Basha
- Department of Chemistry, Indian Academy Degree College-Autonomous Bengaluru, India
| |
Collapse
|
19
|
Lee YT, Tan YJ, Mok PY, Kaur G, Sreenivasan S, Falasca M, Oon CE. Sex-divergent expression of cytochrome P450 and SIRTUIN 1-7 proteins in toxicity evaluation of a benzimidazole-derived epigenetic modulator in mice. Toxicol Appl Pharmacol 2022; 445:116039. [PMID: 35489524 DOI: 10.1016/j.taap.2022.116039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 10/18/2022]
Abstract
Efforts in precision medicine to combat aberrant epigenome have led to the development of epigenetic targeting drugs. We have previously reported the capability of the BZD9L1 epigenetic modulator to impede colorectal tumour growth in vitro and in vivo through sirtuin (SIRT) inhibition. Although most benzimidazole derivatives are commonly less toxic, their effects on SIRTs and cytochrome P450 (CYP) regulations have not been explored alongside toxicity assessments. SIRTs are histone deacetylases that are crucial in maintaining metabolic homeostasis, whereas CYP is essential in drug metabolism. This study aims to determine the toxicology profile of BZD9L1 through oral acute and repeated dose toxicity evaluations, along with molecular analyses of SIRT, CYP and relevant toxicity markers through western blot and quantitative polymerase chain reaction (qPCR). BZD9L1 demonstrated no sign of acute toxicity at the limit dose (2000 mg/kg). The 28-day toxicity study highlighted the tolerability of repeated dose administration without adverse effects. BZD9L1 showed a sex-divergent regulation of hepatic SIRT1-7, CYP2A5 and CYP2D proteins. Furthermore, BZD9L1 did not induce the expression of organ injury proteins or alter the gene expression of cellular function indicators in mouse liver and kidneys, hence demonstrating, at least in part, the safety of BZD9L1 in short-term evaluations. The present study cautions for personalised strategies when employing benzimidazole-derived epigenetic therapeutics.
Collapse
Affiliation(s)
- Yeuan Ting Lee
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Yi Jer Tan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Pei Yi Mok
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Sasidharan Sreenivasan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Marco Falasca
- Curtin Medical School, Curtin Health Innovation Research Institute (CHIRI), Curtin University, GPO Box U1987, Perth, WA 6845, Australia
| | - Chern Ein Oon
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, 11800 Penang, Malaysia.
| |
Collapse
|
20
|
Joe NS, Godet I, Milki N, Ain NUI, Oza HH, Riggins GJ, Gilkes DM. Mebendazole prevents distant organ metastases in part by decreasing ITGβ4 expression and cancer stemness. Breast Cancer Res 2022; 24:98. [PMID: 36578038 PMCID: PMC9798635 DOI: 10.1186/s13058-022-01591-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/09/2022] [Indexed: 12/29/2022] Open
Abstract
Breast cancer is the most diagnosed cancer among women. Approximately 15-20% of all breast cancers are highly invasive triple-negative breast cancer (TNBC) and lack estrogen, progesterone, and ERBB2 receptors. TNBC is challenging to treat due to its aggressive nature with far fewer targeted therapies than other breast cancer subtypes. Current treatments for patients with TNBC consist of cytotoxic chemotherapies, surgery, radiation, and in some instances PARP inhibitors and immunotherapy. To advance current therapeutics, we repurposed mebendazole (MBZ), an orally available FDA-approved anthelmintic that has shown preclinical efficacy for cancers. MBZ has low toxicity in humans and efficacy in multiple cancer models including breast cancer, glioblastoma multiforme, medulloblastoma, colon cancer, pancreatic and thyroid cancer. MBZ was well-tolerated in a phase I clinical trial of adults recently diagnosed with glioma. We determined that the half-maximal inhibitory concentration (IC50) of MBZ in four breast cancer cell lines is well within the range reported for other types of cancer. MBZ reduced TNBC cell proliferation, induced apoptosis, and caused G2/M cell cycle arrest. MBZ reduced the size of primary tumors and prevented lung and liver metastases. In addition, we uncovered a novel mechanism of action for MBZ. We found that MBZ reduces integrin β4 (ITGβ4) expression and cancer stem cell properties. ITGβ4 has previously been implicated in promoting "cancer stemness," which may contribute to the efficacy of MBZ. Collectively, our results contribute to a growing body of evidence suggesting that MBZ should be considered as a therapeutic to slow tumor progression and prevent metastasis.
Collapse
Affiliation(s)
- Natalie S. Joe
- grid.21107.350000 0001 2171 9311Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA
| | - Inês Godet
- grid.21107.350000 0001 2171 9311Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218 USA ,grid.21107.350000 0001 2171 9311Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218 USA
| | - Nubaira Milki
- grid.21107.350000 0001 2171 9311Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218 USA
| | - Noor U. I. Ain
- grid.21107.350000 0001 2171 9311NIH NIDDK Short-Term Research Experience Program to Unlock Potential (STEP-UP), The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA
| | - Harsh H. Oza
- grid.21107.350000 0001 2171 9311Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA
| | - Gregory J. Riggins
- grid.21107.350000 0001 2171 9311Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA
| | - Daniele M. Gilkes
- grid.21107.350000 0001 2171 9311Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Cellular and Molecular Medicine Program, The Johns Hopkins University School of Medicine, Baltimore, MD 21231 USA ,grid.21107.350000 0001 2171 9311Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD 21218 USA ,grid.21107.350000 0001 2171 9311Johns Hopkins Institute for NanoBioTechnology, The Johns Hopkins University, Baltimore, MD 21218 USA
| |
Collapse
|
21
|
Tyagi YK, Jali G, Singh R. Synthesis and Anti-Cancer Applications of Benzimidazole Derivatives - Recent Studies. Anticancer Agents Med Chem 2022; 22:3280-3290. [PMID: 36221180 DOI: 10.2174/1871520622666220429134818] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/13/2022] [Accepted: 02/28/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer is a life-threatening disease. Anti-cancer drugs are the focus of research. The heterocyclic molecules like benzimidazole occupy a central position in searching for novel and effective anti-cancer drugs. The medicinal chemists designed and synthesized several benzimidazole derivatives and conjugates to evaluate them as potential anti-cancer agents. OBJECTIVE The purpose of this compilation of literature is to cover the progress of benzimidazole-based anti-cancer agents, their synthesis, and their evaluation for cancer disease treatment. METHODS The recent literatures have been collected from various search engines and peer-reviewed journals. RESULTS The compounds like benzimidazole derivatives of dehydroabietic acid, piperidyl benzimidazole carboxamide, benzimidazole-quinazolinone hybrids, benzimidazole-thiazole conjugate, and benzimidazole pendant cyanopyrimidine derivatives have been discussed in detail. CONCLUSION This review article will help the medicinal chemists to design and synthesize benzimidazole-based molecules and evaluate them as anti-cancer agents.
Collapse
Affiliation(s)
- Yogesh K Tyagi
- University School of Basic and Applied Sciences, Guru Gobind Singh Indraprastha University, Dwarka, New Delhi, India
| | - Geetan Jali
- Department of Chemistry, Kirori Mal College, University of Delhi, New Delhi, India
| | - Ram Singh
- Department of Applied Chemistry, Delhi Technological University, New Delhi, India
| |
Collapse
|