1
|
Monchusi B, Dube P, Takundwa MM, Kenmogne VL, Malise T, Thimiri Govinda Raj DB. Combination Therapies in Drug Repurposing: Personalized Approaches to Combatting Leukaemia and Multiple Myeloma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40279000 DOI: 10.1007/5584_2025_863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Despite advances in cancer research, treating malignancies remains challenging due to issues like drug resistance, disease heterogeneity, and the limited efficacy of current therapies, particularly in relapsed or refractory cases. In recent years, several drugs originally approved for non-cancer indications have shown potential in cancer treatment, demonstrating anti-proliferative, anti-metastatic, and immunomodulatory effects. Drug repurposing has shown immense promise due to well-established safety profiles and mechanisms of action of the compounds. However, the implementation is fraught with clinical, logistical, regulatory, and ethical challenges, especially in diseases such as leukaemia and multiple myeloma. This chapter examines the treatment challenges in leukaemia and multiple myeloma, focusing on the role of drug repurposing in addressing therapeutic resistance and disease variability. It highlights the potential of personalized, tailored combination therapies, using repurposed drug components, to offer more effective, targeted, and cost-efficient treatment strategies, overcoming resistance and improving patient outcomes.
Collapse
Affiliation(s)
- B Monchusi
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - P Dube
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Haematology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - M M Takundwa
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - V L Kenmogne
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - T Malise
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa
| | - D B Thimiri Govinda Raj
- Synthetic Nanobiotechnology and Biomachines, Synthetic Biology and Precision Medicine Centre, Future production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa.
- Department of Surgery, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
2
|
Vital KD, Pires LO, Gallotti B, Silva JL, Lima de Jesus LC, Alvarez-Leite JI, Ferreira Ê, de Carvalho Azevedo VA, Santos Martins F, Nascimento Cardoso V, Antunes Fernandes SO. Atorvastatin attenuates intestinal mucositis induced by 5-fluorouracil in mice by modulating the epithelial barrier and inflammatory response. J Chemother 2025; 37:175-192. [PMID: 38711347 DOI: 10.1080/1120009x.2024.2345027] [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: 10/25/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024]
Abstract
Chemotherapy-induced intestinal mucositis is a major side effect of cancer treatment. Statins are 3-hydroxy-3-methyl glutaryl coenzyme reductase inhibitors used to treat hypercholesterolemia and atherosclerotic diseases. Recent studies have demonstrated that atorvastatin (ATV) has antioxidant, anti-inflammatory, and resulting from the regulation of different molecular pathways. In the present study, we investigated the effects of ATV on intestinal homeostasis in 5-fluorouracil (5-FU)-induced mucositis. Our results showed that ATV protected the intestinal mucosa from epithelial damage caused by 5-FU mainly due to inflammatory infiltrate and intestinal permeability reduction, downregulation of inflammatory markers, such as Tlr4, MyD88, NF-κB, Tnf-a, Il1β, and Il6 dose-dependent. ATV also improved epithelial barrier function by upregulating the mRNA transcript levels of mucin 2 (MUC2), and ZO-1 and occludin tight junction proteins. The results suggest that the ATV anti-inflammatory and protective effects on 5-FU-induced mice mucositis involve the inhibition of the TLR4/MYD88/NPRL3/NF-κB, iNos, and caspase 3.
Collapse
Affiliation(s)
- Kátia Duarte Vital
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luiz Octavio Pires
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno Gallotti
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Janayne Luihan Silva
- Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luís Cláudio Lima de Jesus
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ênio Ferreira
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Vasco Ariston de Carvalho Azevedo
- Departamento de Genética, Ecologia e Evolução, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Flaviano Santos Martins
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Valbert Nascimento Cardoso
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Simone Odília Antunes Fernandes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
3
|
Hong JP, Choi RJ, Shim JK, Kim K, Kim RN, Cho H, Kim SJ, Kim S, Kim NH, Park HH, Moon JH, Kim EH, Teo WY, Chung S, Chang JH, Kang SG. Synergistic combination of perphenazine and temozolomide suppresses patient-derived glioblastoma tumorspheres. Neuro Oncol 2025; 27:654-667. [PMID: 39392921 PMCID: PMC11889716 DOI: 10.1093/neuonc/noae211] [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: 05/09/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM), a primary malignant brain tumor, has a poor prognosis, even with standard treatments such as radiotherapy and chemotherapy. In this study, we explored the anticancer effects of the synergistic combination of perphenazine (PER), a dopamine receptor D2/3 (DRD2/3) antagonist, and temozolomide (TMZ), a standard treatment for GBM, in patient-derived human GBM tumorspheres (TSs). METHODS The biological effects of the combination of PER and TMZ in GBM TSs were assessed by measuring cell viability, ATP, stemness, invasiveness, and apoptosis. Changes in protein and mRNA expression were analyzed using western blotting and RNA sequencing. Co-administration of PER and TMZ was evaluated in vivo using a mouse orthotopic xenograft model. RESULTS The Severance dataset showed that DRD2 and DRD3 expressions were higher in tumor tissues than in the tumor-free cortex of patients with GBM. DRD2/3 knockout by CRISPR/Cas9 in patient-derived human GBM TSs inhibited cell growth and ATP production. The combined treatment with PER and TMZ resulted in superior effects on cell viability and ATP assays compared to those in single treatment groups. Flow cytometry, western blotting, and RNA sequencing confirmed elevated apoptosis in GBM TSs following combination treatment. Additionally, the combination of PER and TMZ downregulated the expression of protein and mRNA associated with stemness and invasiveness. In vivo evaluation showed that combining PER and TMZ extended the survival period of the mouse orthotopic xenograft model. CONCLUSIONS The synergistic combination of PER and TMZ has potential as a novel combination treatment strategy for GBM.
Collapse
Affiliation(s)
- Jun Pyo Hong
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ran Joo Choi
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Kyoung Shim
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kibyeong Kim
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ryong Nam Kim
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - HyeJoung Cho
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seo Jin Kim
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sohyun Kim
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Nam Hwa Kim
- Department of Premedical, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hun Ho Park
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ju Hyung Moon
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Eui Hyun Kim
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Wan-Yee Teo
- Institute of Molecular and Cell Biology, A*STAR, Singapore, Singapore
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore
| | - Seungsoo Chung
- Department of Physiology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jong Hee Chang
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seok-Gu Kang
- Brain Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Tumor Translational Research Laboratory, Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Neurosurgery, Brain Tumor Center, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medical Science, Yonsei University Graduate School, Seoul, Republic of Korea
| |
Collapse
|
4
|
Czechowicz P, Więch-Walów A, Sławski J, Collawn JF, Bartoszewski R. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 2025; 30:27. [PMID: 40038587 PMCID: PMC11881393 DOI: 10.1186/s11658-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Anna Więch-Walów
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland.
| |
Collapse
|
5
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
6
|
Hewitt E, Bouche G, Alencar AC, Bigelsen SJ, Radu R, Stoyanova-Beninska V, Carrato A, Valsecchi F, Soler Cantón A, van der Meer HG, García Bermejo ML, Budillon A, Cardone L, Rooman I, Platteeuw H, Baijet J, Fuchs C. Drug Repurposing in Pancreatic Cancer: A Multi-Stakeholder Perspective to Improve Treatment Options for Pancreatic Cancer Patients. Cancer Manag Res 2025; 17:429-440. [PMID: 40046652 PMCID: PMC11881603 DOI: 10.2147/cmar.s483151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/02/2025] [Indexed: 04/17/2025] Open
Abstract
Pancreatic cancer (PC) remains one of the most challenging malignancies to treat. Current therapeutic options are unsatisfactory, and there is an urgent need for more effective and less toxic drugs to improve the dismal prognosis of PC. In recent years, drug repurposing (DR) has emerged as an attractive strategy to identify novel treatments for PC by leveraging existing drugs approved for other indications. Through the use of electronic medical records, Artificial Intelligence, study of metabolic pathways, signalling pathways, and many other approaches, it has become much easier in recent years to identify potential novel uses for old drugs. Although policy, funding and research attention in this area are steadily growing, major challenges to efficient and effective patient-centric DR in PC need to be addressed. These include but are not limited to regulatory, financial and funding barriers and the lack of coordination and collaboration among several sectors and stakeholders. To explore the opportunities and challenges associated with DR in PC, a one-day multi-stakeholder meeting was held on 14th of November 2023 in Brussels, Belgium as part of the REMEDi4ALL project. This meeting provided a platform for researchers, clinicians, industry representatives, funders, regulatory experts, and patient advocates to discuss and propose actions to optimize and accelerate DR in PC. Insights from this meeting support the potential of DR to enhance PC treatment options while highlighting the importance of systemic and supportive changes in the regulatory, policy and funding landscapes, interdisciplinary collaboration, data sharing, and patient involvement in driving therapeutic innovation. This summary highlights key outcomes and recommendations from the meeting in informing future efforts to advance DR initiatives in the context of PC.
Collapse
Affiliation(s)
| | | | | | - Stephen J Bigelsen
- Patient Advocate, Department of Allergy, Asthma and Immunology, Rutgers New Jersey Medical School, Newark, NJ, USA
| | | | | | - Alfredo Carrato
- Department of Medical Oncology, Alcalá University, Instituto Ramón Y Cajal de Investigación Sanitaria (IRYCIS), Centro de Investigación Biomédica En Red de Cáncer (CIBERONC), Ramon Y Cajal University Hospital, Madrid, Spain
- Pancreatic Cancer Europe, Brussels, Belgium
| | | | - Alicia Soler Cantón
- EATRIS ERIC, European Infrastructure for Translational Medicine, Amsterdam, the Netherlands
| | | | - María Laura García Bermejo
- Biomarkers and Therapeutic Targets Group, Instituto Ramón Y Cajal de Investigación Sanitaria (IRYCIS), Ramon Y Cajal University Hospital, Madrid, Spain
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori – IRCCS - Fondazione G. Pascale, Naples, Italy
| | - Luca Cardone
- Institute of BiOChemistry and Cell Biology (IBBC), CNR c/o Campus Internazionale “a.buzzati-Traverso”, Monterotondo Scalo Roma (Rome), Italy
- Department of Tumour Immunology and Immunotherapy, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ilse Rooman
- The Anticancer Fund, Meise, Belgium
- Translational Oncology Research Center (TORC), Laboratory of Medical and Molecular Oncology, (LMMO) Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | |
Collapse
|
7
|
Abdelrady YA, Thabet HS, Sayed AM. The future of metronomic chemotherapy: experimental and computational approaches of drug repurposing. Pharmacol Rep 2025; 77:1-20. [PMID: 39432183 DOI: 10.1007/s43440-024-00662-w] [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: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024]
Abstract
Metronomic chemotherapy (MC), long-term continuous administration of anticancer drugs, is gaining attention as an alternative to the traditional maximum tolerated dose (MTD) chemotherapy. By combining MC with other treatments, the therapeutic efficacy is enhanced while minimizing toxicity. MC employs multiple mechanisms, making it a versatile approach against various cancers. However, drug resistance limits the long-term effectiveness of MC, necessitating ongoing development of anticancer drugs. Traditional drug discovery is lengthy and costly due to processes like target protein identification, virtual screening, lead optimization, and safety and efficacy evaluations. Drug repurposing (DR), which screens FDA-approved drugs for new uses, is emerging as a cost-effective alternative. Both experimental and computational methods, such as protein binding assays, in vitro cytotoxicity tests, structure-based screening, and several types of association analyses (Similarity-Based, Network-Based, and Target Gene), along with retrospective clinical analyses, are employed for virtual screening. This review covers the mechanisms of MC, its application in various cancers, DR strategies, examples of repurposed drugs, and the associated challenges and future directions.
Collapse
Affiliation(s)
- Yousef A Abdelrady
- Institute of Pharmaceutical Sciences, University of Freiburg, 79104, Freiburg, Germany
| | - Hayam S Thabet
- Microbiology Department, Faculty of Veterinary Medicine, Assiut University, Asyut, 71526, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, Asyut, 71516, Egypt
- Bioscience Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Kingdom of Saudi Arabia
| |
Collapse
|
8
|
Elebyary TT, Sultan AA, Abu-Risha SE, El Maghraby GM, Amin M. Bilosomal Co-Encapsulated Tamoxifen and Propranolol for Potentiated Anti-Breast Cancer Efficacy: In Vitro and In Vivo Investigation. Pharmaceutics 2025; 17:123. [PMID: 39861770 PMCID: PMC11768151 DOI: 10.3390/pharmaceutics17010123] [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: 12/14/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Tamoxifen (TAM) is an anti-breast cancer drug suffering from acquired resistance development, prompting cancer relapse. Propranolol (PRO)'s repurposing for cancer therapy has gained interest. This work aimed to investigate combined TAM/PRO therapy for potentiating the anti-breast cancer activity of TAM. The work probed bilosomes versus standard noisome for simultaneous oral and intratumor delivery of TAM and PRO. Methods: Bilosomes comprising Span60, cholesterol, and increasing concentrations of bile salts were prepared together with bile salts containing free standard niosomes. The vesicular size and morphology were characterized. The entrapment and release efficiencies of TAM and PRO from the tailored vesicles were determined. The in vivo investigations of anti-tumor activity of TAM with or without PRO employed the solid Ehrlich carcinoma model. Results: The vesicles of all fabricated dispersions were spherical and negatively charged, with a size ranging from 104 to 182 nm. The entrapment efficiency depended on the nature of the drug, recording values ranging from 87.5% to 97.8% for TAM and from 31.0% to 46.8% for PRO. Incorporation of bile salts into vesicles increased TAM and PRO release compared to standard niosomes. Oral administration of combined TAM/PRO bilosomes showed a significant reduction in tumor growth volume compared to that recorded following naked drug administration. Histopathological investigations reflected a significant decline in tumor giant cells and mitotic figures, implying the in vivo capability of the TAM/PRO combination to interfere with cancer cell proliferation and persistence. Conclusions: The overall results demonstrated the impact of repurposed PRO to enhance the anti-breast cancer activity of TAM when both were co-encapsulated into bilosomes.
Collapse
Affiliation(s)
- Toka T. Elebyary
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Educational Hospital, Tanta University, Tanta 31527, Egypt
| | - Amal A. Sultan
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al-Batin, Hafr Al-Batin 39911, Saudi Arabia
| | - Sally E. Abu-Risha
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt;
| | - Gamal M. El Maghraby
- Department of Pharmaceutical Technology, Faculty of pharmacy, Tanta University, Tanta 31527, Egypt; (T.T.E.); (G.M.E.M.)
- Faculty of Pharmacy, Alsalam University, Tanta 31527, Egypt
| | - Manna Amin
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| |
Collapse
|
9
|
Kassab AE, Gedawy EM. Repurposing of Indomethacin and Naproxen as anticancer agents: progress from 2017 to present. RSC Adv 2024; 14:40031-40057. [PMID: 39717807 PMCID: PMC11664213 DOI: 10.1039/d4ra07581a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
Inflammation is strongly linked to cancer and is essential for the growth and development of tumors. Targeting inflammation and the mediators involved in the inflammatory process could therefore provide a suitable method for cancer prevention and therapy. Numerous studies have shown that inflammation can predispose tumors. Non-steroidal anti-inflammatory drugs (NSAIDs) can affect the tumor microenvironment through increasing apoptosis and chemo-sensitivity while decreasing cell migration. Since the development of novel drugs requires a significant amount of money and time and poses a significant challenge for drug discovery, there has been a recent increase in interest in drug repositioning or repurposing. The growing body of research suggests that drug repurposing is essential for the quicker and less expensive development of anticancer therapies. In order to set the course for potential future repositioning of NSAIDs for clinical deployment in the treatment of cancer, the antiproliferative activity of derivatives of Indomethacin and Naproxen as well as their mechanism of action and structural activity relationships (SARs) published in the time frame from 2017 to 2024 are summarized in this review.
Collapse
Affiliation(s)
- Asmaa E Kassab
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt +2023635140 +2023639307
| | - Ehab M Gedawy
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street, P. O. Box 11562 Cairo Egypt +2023635140 +2023639307
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Pharmaceutical Industries, Badr University in Cairo (BUC) Badr City, P. O. Box 11829 Cairo Egypt
| |
Collapse
|
10
|
Nayak R, Mallick B. BMS345541 is predicted as a repurposed drug for the treatment of TMZ-resistant Glioblastoma using target gene expression and virtual drug screening. Cancer Genet 2024; 288-289:20-31. [PMID: 39213700 DOI: 10.1016/j.cancergen.2024.08.082] [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: 08/05/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) is one of the most aggressive and fatal cancers, for which Temozolomide (TMZ) chemo drug is commonly used for its treatment. However, patients gradually develop resistance to this drug, leading to tumor relapse. In our previous study, we have identified lncRNAs that regulate chemoresistance through the competing endogenous RNA (ceRNA) mechanism. In this study, we tried to find FDA-approved drugs against the target proteins of these ceRNA networks through drug repurposing using differential gene expression profiles, which could be used to nullify the effect of lncRNAs and promote the sensitivity of TMZ in GBM. We performed molecular docking and simulation studies of predicted repurposed drugs and their targets. Among the predicted repurposed drugs, we found BMS345541 has a higher binding affinity towards its target protein - FOXG1, making it a more stable complex with FOXG1-DNA. The ADMET analysis of this drug BMS345541 shows a higher half-life and lower cytotoxicity level than other predicted repurposed drugs. Hence, we conjecture that this could be a better drug for increasing the sensitivity of TMZ for treating GBM patients.
Collapse
Affiliation(s)
- Rojalin Nayak
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha 769008, India
| | - Bibekanand Mallick
- RNAi and Functional Genomics Lab., Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha 769008, India.
| |
Collapse
|
11
|
Makgoo L, Mosebi S, Mbita Z. Susceptibility of HPV-18 Cancer Cells to HIV Protease Inhibitors. Viruses 2024; 16:1622. [PMID: 39459955 PMCID: PMC11512214 DOI: 10.3390/v16101622] [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: 09/20/2024] [Revised: 10/12/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Cervical cancer cases continue to rise despite all the advanced screening and preventative measures put in place, which include human papillomavirus (HPV) vaccination. These soaring numbers can be attributed to the lack of effective anticancer drugs against cervical cancer; thus, repurposing the human immunodeficiency virus protease inhibitors is an attractive innovation. Therefore, this work was aimed at evaluating the potential anticancer activities of HIV-PIs against cervical cancer cells. The MTT viability assay was used to evaluate the effect of HIV protease inhibitors on the viability of cervical cancer cells (HeLa) and non-cancerous cells (HEK-293). Further confirmation of the MTT assay was performed by confirming the IC50s of these HIV protease inhibitors on cervical cancer cells and non-cancerous cells using the Muse™ Count and Viability assay. To confirm the mode of death induced by HIV protease inhibitors in the HPV-associated cervical cancer cell line, apoptosis was performed using Annexin V assay. In addition, the Muse™ Cell Cycle assay was used to check whether the HIV protease inhibitors promote or halt cell cycle progression in cervical cancer cells. HIV protease inhibitors did not affect the viability of non-cancerous cells (HEK-293), but they decreased the viability of HeLa cervical cancer cells in a dose-dependent manner. HIV protease inhibitors induced apoptosis in HPV-related cervical cancer cells. Furthermore, they also induced cell cycle arrest, thus halting cell cycle progression. Therefore, the use of HIV drugs, particularly HIV-1 protease inhibitors, as potential cancer therapeutics represents a promising strategy. This is supported by our study demonstrating their anticancer properties, notably in HPV-associated cervical cancer cell line.
Collapse
Affiliation(s)
- Lilian Makgoo
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X 1106, Sovenga, Polokwane 0727, South Africa;
| | - Salerwe Mosebi
- Department of Life and Consumer Sciences, University of South Africa, Private Bag X06, Florida 1710, South Africa;
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag X 1106, Sovenga, Polokwane 0727, South Africa;
| |
Collapse
|
12
|
Crittenden D, Gallagher R, Milans del Bosch F, Fox DM, Kleiman LB. Pathways for non-manufacturers to drive generic drug repurposing for cancer in the U.S. Front Pharmacol 2024; 15:1419772. [PMID: 39444616 PMCID: PMC11496753 DOI: 10.3389/fphar.2024.1419772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024] Open
Abstract
Repurposing generic drugs as new treatments for life-threatening diseases such as cancer is an exciting yet largely overlooked opportunity due to a lack of market-driven incentives. Nonprofit organizations and other non-manufacturers have been ramping up efforts to repurpose widely available generic drugs and rapidly expand affordable treatment options for patients. However, these non-manufacturers find it difficult to obtain regulatory approval in the U.S. Without a straightforward path for approval and updating drug labeling, non-manufacturers have relied on off-label use of repurposed drugs. This limits the broad clinical adoption of these drugs and patient access. In this paper, we explore the regulatory landscape for repurposing of small molecule generic drugs within the U.S. We describe case studies of repurposed drugs that have been successfully incorporated into clinical treatment guidelines for cancer without regulatory approval. To encourage greater adoption of generic drugs in clinical practice-that is, to encourage the repurposing of these drugs-we examine existing Food and Drug Administration (FDA) pathways for approval of new uses or indications for generic drugs. We show how non-manufacturers, who are generally more active in generic drug repurposing than manufacturers, could utilize existing regulatory authorities and pathways, and we describe the challenges they face. We propose an extension of the existing 505(b)(2) new drug application (NDA) approval pathway, called a "labeling-only" 505(b)(2) NDA, that would enable non-manufacturers to seek approval of new indications for well-established small molecule drugs when multiple generic products are already available. It would not require new chemistry, manufacturing, and controls (CMC) data or introducing new drug products into the marketplace. This pathway would unlock innovation broadly and enable patients to benefit from the enormous potential of low-cost generic drugs.
Collapse
|
13
|
Bano N, Parveen S, Saeed M, Siddiqui S, Abohassan M, Mir SS. Drug Repurposing of Selected Antibiotics: An Emerging Approach in Cancer Drug Discovery. ACS OMEGA 2024; 9:26762-26779. [PMID: 38947816 PMCID: PMC11209889 DOI: 10.1021/acsomega.4c00617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/19/2024] [Accepted: 05/29/2024] [Indexed: 07/02/2024]
Abstract
Drug repurposing is a method of investigating new therapeutic applications for previously approved medications. This repurposing approach to "old" medications is now highly efficient, simple to arrange, and cost-effective and poses little risk of failure in treating a variety of disorders, including cancer. Drug repurposing for cancer therapy is currently a key topic of study. It is a way of exploring recent therapeutic applications for already-existing drugs. Theoretically, the repurposing strategy has various advantages over the recognized challenges of creating new molecular entities, including being faster, safer, easier, and less expensive. In the real world, several medications have been repurposed, including aspirin, metformin, and chloroquine. However, doctors and scientists address numerous challenges when repurposing drugs, such as the fact that most drugs are not cost-effective and are resistant to bacteria. So the goal of this review is to gather information regarding repurposing pharmaceuticals to make them more cost-effective and harder for bacteria to resist. Cancer patients are more susceptible to bacterial infections. Due to their weak immune systems, antibiotics help protect them from a variety of infectious diseases. Although antibiotics are not immune boosters, they do benefit the defense system by killing bacteria and slowing the growth of cancer cells. Their use also increases the therapeutic efficacy and helps avoid recurrence. Of late, antibiotics have been repurposed as potent anticancer agents because of the evolutionary relationship between the prokaryotic genome and mitochondrial DNA of eukaryotes. Anticancer antibiotics that prevent cancer cells from growing by interfering with their DNA and blocking growth of promoters, which include anthracyclines, daunorubicin, epirubicin, mitoxantrone, doxorubicin, and idarubicin, are another type of FDA-approved antibiotics used to treat cancer. According to the endosymbiotic hypothesis, prokaryotes and eukaryotes are thought to have an evolutionary relationship. Hence, in this study, we are trying to explore antibiotics that are necessary for treating diseases, including cancer, helping people reduce deaths associated with various infections, and substantially extending people's life expectancy and quality of life.
Collapse
Affiliation(s)
- Nilofer Bano
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Bioengineering, Faculty of Engineering, Integral University, Kursi Road, Lucknow 226026, India
| | - Sana Parveen
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| | - Mohd Saeed
- Department
of Biology, College of Sciences, University
of Hail, P.O. Box 2240, Hail 55476, Saudi Arabia
| | - Samra Siddiqui
- Department
of Health Services Management, College of Public Health and Health
Informatics, University of Hail, Hail 55476, Saudi Arabia
| | - Mohammad Abohassan
- Department
of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Snober S. Mir
- Molecular
Cell Biology Laboratory, Integral Centre of Excellence for Interdisciplinary
Research (ICEIR-4), Integral University, Kursi Road, Lucknow 226026, India
- Department
of Biosciences, Faculty of Science, Integral
University, Kursi Road, Lucknow 226026, India
| |
Collapse
|
14
|
Tripathi S, Gupta E, Galande S. Statins as anti-tumor agents: A paradigm for repurposed drugs. Cancer Rep (Hoboken) 2024; 7:e2078. [PMID: 38711272 PMCID: PMC11074523 DOI: 10.1002/cnr2.2078] [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: 11/14/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Statins, frequently prescribed medications, work by inhibiting the rate-limiting enzyme HMG-CoA reductase (HMGCR) in the mevalonate pathway to reduce cholesterol levels. Due to their multifaceted benefits, statins are being adapted for use as cost-efficient, safe and effective anti-cancer treatments. Several studies have shown that specific types of cancer are responsive to statin medications since they rely on the mevalonate pathway for their growth and survival. RECENT FINDINGS Statin are a class of drugs known for their potent inhibition of cholesterol production and are typically prescribed to treat high cholesterol levels. Nevertheless, there is growing interest in repurposing statins for the treatment of malignant neoplastic diseases, often in conjunction with chemotherapy and radiotherapy. The mechanism behind statin treatment includes targeting apoptosis through the BCL2 signaling pathway, regulating the cell cycle via the p53-YAP axis, and imparting epigenetic modulations by altering methylation patterns on CpG islands and histone acetylation by downregulating DNMTs and HDACs respectively. Notably, some studies have suggested a potential chemo-preventive effect, as decreased occurrence of tumor relapse and enhanced survival rate were reported in patients undergoing long-term statin therapy. However, the definitive endorsement of statin usage in cancer therapy hinges on population based clinical studies with larger patient cohorts and extended follow-up periods. CONCLUSIONS The potential of anti-cancer properties of statins seems to reach beyond their influence on cholesterol production. Further investigations are necessary to uncover their effects on cancer promoting signaling pathways. Given their distinct attributes, statins might emerge as promising contenders in the fight against tumorigenesis, as they appear to enhance the efficacy and address the limitations of conventional cancer treatments.
Collapse
Affiliation(s)
- Sneha Tripathi
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
| | - Ekta Gupta
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
| | - Sanjeev Galande
- Laboratory of Chromatin Biology & EpigeneticsIndian Institute of Science Education and ResearchPuneIndia
- Centre of Excellence in Epigenetics, Department of Life SciencesShiv Nadar Institution of EminenceGautam Buddha NagarIndia
| |
Collapse
|
15
|
Verrelle P, Gestraud P, Poyer F, Soria A, Tessier S, Lescure A, Anthony E, Corbé M, Heinrich S, Beauvineau C, Chaput L, Granzhan A, Piguel S, Perez F, Teulade-Fichou MP, Megnin-Chanet F, Del Nery E. Integrated High-Throughput Screening and Large-Scale Isobolographic Analysis to Accelerate the Discovery of Radiosensitizers With Greater Selectivity for Cancer Cells. Int J Radiat Oncol Biol Phys 2024; 118:1294-1307. [PMID: 37778425 DOI: 10.1016/j.ijrobp.2023.09.044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023]
Abstract
PURPOSE High-throughput screening (HTS) platforms have been widely used to identify candidate anticancer drugs and drug-drug combinations; however, HTS-based identification of new drug-ionizing radiation (IR) combinations has rarely been reported. Herein, we developed an integrated approach including cell-based HTS and computational large-scale isobolographic analysis to accelerate the identification of radiosensitizing compounds acting strongly and more specifically on cancer cells. METHODS AND MATERIALS In a 384-well plate format, 160 compounds likely to interfere with the cell response to radiation were screened on human glioblastoma (U251-MG) and cervix carcinoma (ME-180) cell lines, as well as on normal fibroblasts (CCD-19Lu). After drug exposure, cells were irradiated or not and short-term cell survival was assessed by high-throughput cell microscopy. Computational large-scale dose-response and isobolographic approach were used to identify promising synergistic drugs radiosensitizing cancer cells rather than normal cells. Synergy of a promising compound was confirmed on ME-180 cells by an independent 96-well assay protocol, and finally, by the gold-standard colony forming assay. RESULTS We retained 4 compounds synergistic at 2 isoeffects in U251-MG and ME-180 cell lines and 11 compounds synergistically effective in only one cancer cell line. Among these 15 promising radiosensitizers, 5 compounds showed limited toxicity combined or not with IR on normal fibroblasts. CONCLUSIONS Overall, this study demonstrated that HTS chemoradiation screening together with large-scale computational analysis is an efficient tool to identify synergistic drug-IR combinations, with concomitant assessment of unwanted toxicity on normal fibroblasts. It sparks expectations to accelerate the discovery of highly desired agents improving the therapeutic index of radiation therapy.
Collapse
Affiliation(s)
- Pierre Verrelle
- Radiation Oncology Department, Institut Curie Hospital, Paris, France; Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France.
| | - Pierre Gestraud
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Florent Poyer
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Adèle Soria
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Sarah Tessier
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Aurianne Lescure
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Elodie Anthony
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Maxime Corbé
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France
| | - Sophie Heinrich
- Experimental Radiotherapy Platform (RadeXp), Translational Research Department, Institut Curie, Orsay, France; Inserm U1021-CNRS UMR 3347, Institut Curie, Paris Saclay University
| | - Claire Beauvineau
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Ludovic Chaput
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Anton Granzhan
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Sandrine Piguel
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France; BioCIS UMR8076, Université Paris-Saclay, Faculté de Pharmacie, Orsay, France
| | - Franck Perez
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France; Cell Biology and Cancer UMR144, Institut Curie, PSL Research University, Paris, France
| | - Marie-Paule Teulade-Fichou
- Chemistry and Modelisation for the Biology of Cancer, CNRS UMR9187, INSERM U1196, Institut Curie, Université Paris Saclay, 91405 Orsay, France
| | - Frédérique Megnin-Chanet
- Bioinformatics and Computational Systems Biology of Cancer, PSL Research University, Mines Paris Tech, INSERM U900, Paris, France
| | - Elaine Del Nery
- Biophenics High-Content Screening Laboratory, Department of Translational Research, PSL Research University, PICT-IBiSa, Institut Curie Research Center, Paris, France.
| |
Collapse
|
16
|
Mostkowska A, Rousseau G, Raynal NJM. Repurposing of rituximab biosimilars to treat B cell mediated autoimmune diseases. FASEB J 2024; 38:e23536. [PMID: 38470360 DOI: 10.1096/fj.202302259rr] [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: 11/01/2023] [Revised: 02/12/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024]
Abstract
Rituximab, the first monoclonal antibody approved for the treatment of lymphoma, eventually became one of the most popular and versatile drugs ever in terms of clinical application and revenue. Since its patent expiration, and consequently, the loss of exclusivity of the original biologic, its repurposing as an off-label drug has increased dramatically, propelled by the development and commercialization of its many biosimilars. Currently, rituximab is prescribed worldwide to treat a vast range of autoimmune diseases mediated by B cells. Here, we present a comprehensive overview of rituximab repurposing in 115 autoimmune diseases across 17 medical specialties, sourced from over 1530 publications. Our work highlights the extent of its off-label use and clinical benefits, underlining the success of rituximab repurposing for both common and orphan immune-related diseases. We discuss the scientific mechanism associated with its clinical efficacy and provide additional indications for which rituximab could be investigated. Our study presents rituximab as a flagship example of drug repurposing owing to its central role in targeting cluster of differentiate 20 positive (CD20) B cells in 115 autoimmune diseases.
Collapse
Affiliation(s)
- Agata Mostkowska
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Guy Rousseau
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Noël J-M Raynal
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, Quebec, Canada
- Centre de recherche du CHU Sainte-Justine, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Weth FR, Hoggarth GB, Weth AF, Paterson E, White MPJ, Tan ST, Peng L, Gray C. Unlocking hidden potential: advancements, approaches, and obstacles in repurposing drugs for cancer therapy. Br J Cancer 2024; 130:703-715. [PMID: 38012383 PMCID: PMC10912636 DOI: 10.1038/s41416-023-02502-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 10/30/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2023] Open
Abstract
High rates of failure, exorbitant costs, and the sluggish pace of new drug discovery and development have led to a growing interest in repurposing "old" drugs to treat both common and rare diseases, particularly cancer. Cancer, a complex and heterogeneous disease, often necessitates a combination of different treatment modalities to achieve optimal outcomes. The intrinsic polygenicity of cancer, intricate biological signalling networks, and feedback loops make the inhibition of a single target frequently insufficient for achieving the desired therapeutic impact. As a result, addressing these complex or "smart" malignancies demands equally sophisticated treatment strategies. Combinatory treatments that target the multifaceted oncogenic signalling network hold immense promise. Repurposed drugs offer a potential solution to this challenge, harnessing known compounds for new indications. By avoiding the prohibitive costs and long development timelines associated with novel cancer drugs, this approach holds the potential to usher in more effective, efficient, and cost-effective cancer treatments. The pursuit of combinatory therapies through drug repurposing may hold the key to achieving superior outcomes for cancer patients. However, drug repurposing faces significant commercial, technological and regulatory challenges that need to be addressed. This review explores the diverse approaches employed in drug repurposing, delves into the challenges faced by the drug repurposing community, and presents innovative solutions to overcome these obstacles. By emphasising the significance of combinatory treatments within the context of drug repurposing, we aim to unlock the full potential of this approach for enhancing cancer therapy. The positive aspects of drug repurposing in oncology are underscored here; encompassing personalized treatment, accelerated development, market opportunities for shelved drugs, cancer prevention, expanded patient reach, improved patient access, multi-partner collaborations, increased likelihood of approval, reduced costs, and enhanced combination therapy.
Collapse
Affiliation(s)
- Freya R Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Georgia B Hoggarth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Anya F Weth
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
| | | | - Swee T Tan
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt, 5040, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
18
|
Fong ZV, Severs G, Moir J, White S, Qadan M, Tingle S. Calcium channel blockers are associated with improved survival in pancreatic cancer patients undergoing neoadjuvant chemotherapy and resection. HPB (Oxford) 2024; 26:418-425. [PMID: 38135550 DOI: 10.1016/j.hpb.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 10/16/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Repurposing existing drugs for use in oncology is more efficient, cost-effective and safe than novel drug discovery. Calcium signalling is increasingly recognised to have a key role in chemoresistance. This study assessed the impact of calcium channel blockers (CCB) in pancreatic cancer. METHODS Retrospective population study of patients undergoing resection (curative intent) of pancreatic ductal adenocarcinoma (SEER-Medicare, 2007-2017). Cox models were built to assess the impact on overall survival. As laboratory studies suggest a chemosensitising effect, the impact of CCB was assessed separately in patients receiving neoadjuvant chemotherapy. RESULTS 6,223 patients were included, of whom 660 were prescribed CCB. In total, 591 received neoadjuvant chemotherapy; in this cohort CCB prescription was associated with improved overall survival when adjusting for multiple prognostic factors (aHR = 0.715, 0.514-0.996, P = 0.047). This effect was not observed in patients not receiving neoadjuvant chemotherapy (aHR = 1.082, 0.982-1.191, P = 0.112). CONCLUSION CCB prescription was associated with improved overall survival in patients receiving neoadjuvant chemotherapy prior to pancreatic cancer resection. The association was specific to the group of patients receiving neoadjuvant chemotherapy, mirroring the chemosensitising effect in laboratory studies. This defines patients receiving neoadjuvant chemotherapy as a target population for prospective clinical trials of CCB in pancreatic cancer.
Collapse
Affiliation(s)
- Zhi V Fong
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - George Severs
- Department of HPB Surgery, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - John Moir
- Department of HPB Surgery, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK
| | - Steve White
- Department of HPB Surgery, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Samuel Tingle
- Department of HPB Surgery, Freeman Hospital, Newcastle upon Tyne, NE7 7DN, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
19
|
Gouveia MJ, Ribeiro E, Vale N. A Surprising Repurposing of Central Nervous System Drugs against Squamous Cell Carcinoma of the Bladder, UM-UC-5. Pharmaceutics 2024; 16:212. [PMID: 38399266 PMCID: PMC10892655 DOI: 10.3390/pharmaceutics16020212] [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: 11/30/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The potential benefits of drug repurposing have gained attention as an alternative to developing de novo drugs. The potential of using central nervous system (CNS) drugs as anticancer drugs has been explored in several types of human cancers, such as breast and colon cancer, among others. Here, we examine the effect of the CNS drugs sertraline, paroxetine, and chlorpromazine on human squamous carcinoma cells of the bladder (UM-UC-5). After exposing UM-UC-5 cells to increased concentrations of each drug for 48 h, we assessed their metabolic activity using an MTT assay. Based on those results, we calculated cell viability and the half-maximal inhibitory concentration (IC50) values. The results suggest that the CNS drugs were effective against UM-UC-5 in the order of potency of sertraline > chlorpromazine > paroxetine. Interestingly, sertraline was more potent than 5-fluorouracil (5-FU), a widely used anticancer drug. This study demonstrated, for the first time, the promising anticancer activity of CNS drugs on human bladder cancer cells in vitro and supports the repurposing of CNS drugs to improve cancer treatment. Nevertheless, further studies are necessary to understand their mechanism of action and in vivo activity.
Collapse
Affiliation(s)
- Maria João Gouveia
- Center for the Study in Animal Science (CECA/ICETA), University of Porto, Rua de D. Manuel II, Apt 55142, 4051-401 Porto, Portugal;
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, Rua Alexandre Herculano 321, 4000-055 Porto, Portugal
| | - Eduarda Ribeiro
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- ICBAS—School of Medicine and Biomedical Sciences, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| |
Collapse
|
20
|
Fatemi N, Karimpour M, Bahrami H, Zali MR, Chaleshi V, Riccio A, Nazemalhosseini-Mojarad E, Totonchi M. Current trends and future prospects of drug repositioning in gastrointestinal oncology. Front Pharmacol 2024; 14:1329244. [PMID: 38239190 PMCID: PMC10794567 DOI: 10.3389/fphar.2023.1329244] [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: 10/28/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
Gastrointestinal (GI) cancers comprise a significant number of cancer cases worldwide and contribute to a high percentage of cancer-related deaths. To improve survival rates of GI cancer patients, it is important to find and implement more effective therapeutic strategies with better prognoses and fewer side effects. The development of new drugs can be a lengthy and expensive process, often involving clinical trials that may fail in the early stages. One strategy to address these challenges is drug repurposing (DR). Drug repurposing is a developmental strategy that involves using existing drugs approved for other diseases and leveraging their safety and pharmacological data to explore their potential use in treating different diseases. In this paper, we outline the existing therapeutic strategies and challenges associated with GI cancers and explore DR as a promising alternative approach. We have presented an extensive review of different DR methodologies, research efforts and examples of repurposed drugs within various GI cancer types, such as colorectal, pancreatic and liver cancers. Our aim is to provide a comprehensive overview of employing the DR approach in GI cancers to inform future research endeavors and clinical trials in this field.
Collapse
Affiliation(s)
- Nayeralsadat Fatemi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Karimpour
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hoda Bahrami
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Vahid Chaleshi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Andrea Riccio
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Institute of Genetics and Biophysics (IGB) “Adriano Buzzati-Traverso”, Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Ehsan Nazemalhosseini-Mojarad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Totonchi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), Università degli Studi della Campania “Luigi Vanvitelli”, Caserta, Italy
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
21
|
Kumari P, Dang S. Evaluation of Enhanced Cytotoxicity Effect of Repurposed Drug Simvastatin/Thymoquinone Combination against Breast Cancer Cell Line. Cardiovasc Hematol Agents Med Chem 2024; 22:348-366. [PMID: 37907488 DOI: 10.2174/0118715257259037231012182741] [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: 05/09/2023] [Revised: 08/02/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Repurposing of drugs for their anticancer potential is gaining a lot of importance in drug discovery. AIMS The present study aims to explore the potential of Simvastatin (SIM), a drug used in the treatment of high cholesterol, and Thymoquinone (Nigella Sativa) (THY) for its anti-cancer activity on breast cancer cell lines. Thymoquinone is reported to have many potential medicinal properties exhibiting antioxidant, antiinflammatory, anti-cancer, activities like inhibition of tissue growth and division. METHODS In this analysis, we explored the inhibitory effects of the combination of Simvastatin ad Thymoquinone on two breast cancer cell lines viz MCF-7 and MDA-MB-231 cells. The combined effect of Simvastatin and Thymoquinone on Cell viability, Colony formation, Cell migration, and orientation of more programmed cell death in vitro was studied. Cell cycle arrest in the G2/M phase was concomitant with the combined effect of SIM and THY persuading apoptosis and generating reactive oxygen species (ROS). RESULTS The cell cycle arrest with combined treatment was observed that only 1.8% and 1.1% cells gated in G2/M phase in MCF-7 & MDA-MB-231 cell. An increased apoptosis was observed when cells were treated in combination which was about 76.20% and 58.15% respectively for MCF-7 and MDA-MB-231 cells. CONCLUSION It was concluded that the combined effect of simvastatin and thymoquinone stimulates apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Pallavi Kumari
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| |
Collapse
|
22
|
Rai S, Shukla S, Scotti L, Mani A. Drug Repurposing against Novel Therapeutic Targets in Plasmodium falciparum for Malaria: The Computational Perspective. Curr Med Chem 2024; 31:6272-6287. [PMID: 37550911 DOI: 10.2174/0929867331666230807151708] [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: 04/06/2023] [Revised: 05/28/2023] [Accepted: 07/01/2023] [Indexed: 08/09/2023]
Abstract
Malaria remains one of the most challenging tropical diseases. Since malaria cases are reportedly alarming in terms of infections and mortality, urgent attention is needed for addressing the issues of drug resistance in falciparum malaria. High throughput screening methods have paved way for rapid identification of anti-malarial. Furthermore, drug repurposing helps in shortening the time required for drug safety approvals. Hence, discovery of new antimalarials by drug repurposing is a promising approach for combating the disease. This article summarizes the recent computational approaches used for identifying novel antimalarials by using drug target interaction tools followed by pharmacokinetic studies.
Collapse
Affiliation(s)
- Shweta Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| | - Shruti Shukla
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| | - Luciana Scotti
- Postgraduate Programa in Natural and Synthetic Bioactive Compounds, University Hospital, Federal University of Paraíba-Campus I, 58051-970, João Pessoa, PB, Brazil
| | - Ashutosh Mani
- Department of Biotechnology, Motilal Nehru National Institute of Technology, Allahabad, 211004 India
| |
Collapse
|
23
|
Almeida-Nunes DL, Silvestre R, Dinis-Oliveira RJ, Ricardo S. Enhancing Immunotherapy in Ovarian Cancer: The Emerging Role of Metformin and Statins. Int J Mol Sci 2023; 25:323. [PMID: 38203494 PMCID: PMC10779012 DOI: 10.3390/ijms25010323] [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: 11/25/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
Ovarian cancer metastization is accompanied by the development of malignant ascites, which are associated with poor prognosis. The acellular fraction of this ascitic fluid contains tumor-promoting soluble factors, bioactive lipids, cytokines, and extracellular vesicles, all of which communicate with the tumor cells within this peritoneal fluid. Metabolomic profiling of ovarian cancer ascites has revealed significant differences in the pathways of fatty acids, cholesterol, glucose, and insulin. The proteins involved in these pathways promote tumor growth, resistance to chemotherapy, and immune evasion. Unveiling the key role of this liquid tumor microenvironment is crucial for discovering more efficient treatment options. This review focuses on the cholesterol and insulin pathways in ovarian cancer, identifying statins and metformin as viable treatment options when combined with standard chemotherapy. These findings are supported by clinical trials showing improved overall survival with these combinations. Additionally, statins and metformin are associated with the reversal of T-cell exhaustion, positioning these drugs as potential combinatory strategies to improve immunotherapy outcomes in ovarian cancer patients.
Collapse
Affiliation(s)
- Diana Luísa Almeida-Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal;
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal;
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal;
- UCIBIO-REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4169-007 Porto, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4169-007 Porto, Portugal
- FOREN—Forensic Science Experts, 1400-136 Lisboa, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal;
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116 Gandra, Portugal;
- Faculty of Medicine, University of Porto, 4169-007 Porto, Portugal
| |
Collapse
|
24
|
Zhang N, Li Y, Sundquist J, Sundquist K, Ji J. Identifying actionable druggable targets for breast cancer: Mendelian randomization and population-based analyses. EBioMedicine 2023; 98:104859. [PMID: 38251461 PMCID: PMC10628347 DOI: 10.1016/j.ebiom.2023.104859] [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: 05/26/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Drug repurposing provides a cost-effective approach to address the need for breast cancer prevention and therapeutics. We aimed to identify actionable druggable targets using Mendelian randomization (MR) and then validate the candidate drugs using population-based analyses. METHODS We identified genetic instruments for 1406 actionable targets of approved non-oncological drugs based on gene expression, DNA methylation, and protein expression quantitative trait loci (eQTL, mQTL, and pQTL, respectively). Genome-wide association study (GWAS) summary statistics were obtained from the Breast Cancer Association Consortium (122,977 cases, 105,974 controls). We further conducted a nested case-control study using data retrieved from Swedish registers to validate the candidate drugs that were identified from MR analyses. FINDINGS We identified six significant MR associations with gene expression levels (TUBB, MDM2, CSK, ULK3, MC1R and KCNN4) and two significant associations with gene methylation levels across 21 CpG islands (RPS23 and MAPT). Results from the nested case-control study showed that the use of raloxifene (targeting MAPT) was associated with 35% reduced breast cancer risk (odds ratio, OR, 0.65; 95% confidence interval, CI, 0.51-0.83). However, usage of estradiol, tolterodine, and nitrofurantoin (also targeting MAPT) was associated with increased breast cancer risk, with adjusted ORs and 95% CI of 1.10 (1.07-1.13), 1.16 (1.09-1.24), and 1.09 (1.05-1.13), respectively. The effect of raloxifene and nitrofurantoin lost significance in further validation analyses using active-comparator and new-user design. INTERPRETATION This large-scale MR analysis, combined with population-based validation, identified eight druggable target genes for breast cancer and suggested that raloxifene is an effective chemoprevention against breast cancer. FUNDING Swedish Research Council, Cancerfonden, Crafoordska Stiftelsen, Allmänna Sjukhusets i Malmö Stiftelsen för bekämpande av cancer, 111 Project and MAS cancer.
Collapse
Affiliation(s)
- Naiqi Zhang
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden.
| | - Yanni Li
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
| | - Jan Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, USA; Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Japan
| | - Kristina Sundquist
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden; Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, USA; Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Japan
| | - Jianguang Ji
- Center for Primary Health Care Research, Department of Clinical Sciences Malmö, Lund University, Sweden
| |
Collapse
|
25
|
Beaudry A, Jacques-Ricard S, Darracq A, Sgarioto N, Garcia A, García TR, Lemieux W, Béland K, Haddad E, Cordeiro P, Duval M, McGraw S, Richer C, Caron M, Marois F, St-Onge P, Sinnett D, Banquy X, Raynal NJM. Repurposing disulfiram, an alcohol-abuse drug, in neuroblastoma causes KAT2A downregulation and in vivo activity with a water/oil emulsion. Sci Rep 2023; 13:16443. [PMID: 37777587 PMCID: PMC10543387 DOI: 10.1038/s41598-023-43219-2] [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: 03/09/2023] [Accepted: 09/21/2023] [Indexed: 10/02/2023] Open
Abstract
Neuroblastoma, the most common type of pediatric extracranial solid tumor, causes 10% of childhood cancer deaths. Despite intensive multimodal treatment, the outcomes of high-risk neuroblastoma remain poor. We urgently need to develop new therapies with safe long-term toxicity profiles for rapid testing in clinical trials. Drug repurposing is a promising approach to meet these needs. Here, we investigated disulfiram, a safe and successful chronic alcoholism treatment with known anticancer and epigenetic effects. Disulfiram efficiently induced cell cycle arrest and decreased the viability of six human neuroblastoma cell lines at half-maximal inhibitory concentrations up to 20 times lower than its peak clinical plasma level in patients treated for chronic alcoholism. Disulfiram shifted neuroblastoma transcriptome, decreasing MYCN levels and activating neuronal differentiation. Consistently, disulfiram significantly reduced the protein level of lysine acetyltransferase 2A (KAT2A), drastically reducing acetylation of its target residues on histone H3. To investigate disulfiram's anticancer effects in an in vivo model of high-risk neuroblastoma, we developed a disulfiram-loaded emulsion to deliver the highly liposoluble drug. Treatment with the emulsion significantly delayed neuroblastoma progression in mice. These results identify KAT2A as a novel target of disulfiram, which directly impacts neuroblastoma epigenetics and is a promising candidate for repurposing to treat pediatric neuroblastoma.
Collapse
Affiliation(s)
- Annie Beaudry
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Simon Jacques-Ricard
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Anaïs Darracq
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Nicolas Sgarioto
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Araceli Garcia
- Faculté de Pharmacie, Université de Montréal, Montreal, QC, Canada
| | | | - William Lemieux
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Kathie Béland
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Elie Haddad
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Paulo Cordeiro
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Michel Duval
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Serge McGraw
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Chantal Richer
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Maxime Caron
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - François Marois
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Pascal St-Onge
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
| | - Daniel Sinnett
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada
- Département de Pédiatrie, Université de Montréal, Montreal, QC, Canada
| | - Xavier Banquy
- Faculté de Pharmacie, Université de Montréal, Montreal, QC, Canada
| | - Noël J-M Raynal
- Sainte-Justine University Hospital Research Center, 3175, Chemin de la Côte-Sainte-Catherine, Montreal, QC, H3T 1C5, Canada.
- Département de Pharmacologie et de Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada.
| |
Collapse
|
26
|
Mohi-Ud-Din R, Chawla A, Sharma P, Mir PA, Potoo FH, Reiner Ž, Reiner I, Ateşşahin DA, Sharifi-Rad J, Mir RH, Calina D. Repurposing approved non-oncology drugs for cancer therapy: a comprehensive review of mechanisms, efficacy, and clinical prospects. Eur J Med Res 2023; 28:345. [PMID: 37710280 PMCID: PMC10500791 DOI: 10.1186/s40001-023-01275-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 08/08/2023] [Indexed: 09/16/2023] Open
Abstract
Cancer poses a significant global health challenge, with predictions of increasing prevalence in the coming years due to limited prevention, late diagnosis, and inadequate success with current therapies. In addition, the high cost of new anti-cancer drugs creates barriers in meeting the medical needs of cancer patients, especially in developing countries. The lengthy and costly process of developing novel drugs further hinders drug discovery and clinical implementation. Therefore, there has been a growing interest in repurposing approved drugs for other diseases to address the urgent need for effective cancer treatments. The aim of this comprehensive review is to provide an overview of the potential of approved non-oncology drugs as therapeutic options for cancer treatment. These drugs come from various chemotherapeutic classes, including antimalarials, antibiotics, antivirals, anti-inflammatory drugs, and antifungals, and have demonstrated significant antiproliferative, pro-apoptotic, immunomodulatory, and antimetastatic properties. A systematic review of the literature was conducted to identify relevant studies on the repurposing of approved non-oncology drugs for cancer therapy. Various electronic databases, such as PubMed, Scopus, and Google Scholar, were searched using appropriate keywords. Studies focusing on the therapeutic potential, mechanisms of action, efficacy, and clinical prospects of repurposed drugs in cancer treatment were included in the analysis. The review highlights the promising outcomes of repurposing approved non-oncology drugs for cancer therapy. Drugs belonging to different therapeutic classes have demonstrated notable antitumor effects, including inhibiting cell proliferation, promoting apoptosis, modulating the immune response, and suppressing metastasis. These findings suggest the potential of these repurposed drugs as effective therapeutic approaches in cancer treatment. Repurposing approved non-oncology drugs provides a promising strategy for addressing the urgent need for effective and accessible cancer treatments. The diverse classes of repurposed drugs, with their demonstrated antiproliferative, pro-apoptotic, immunomodulatory, and antimetastatic properties, offer new avenues for cancer therapy. Further research and clinical trials are warranted to explore the full potential of these repurposed drugs and optimize their use in treating various cancer types. Repurposing approved drugs can significantly expedite the process of identifying effective treatments and improve patient outcomes in a cost-effective manner.
Collapse
Affiliation(s)
- Roohi Mohi-Ud-Din
- Department of General Medicine, Sher-I-Kashmir Institute of Medical Sciences (SKIMS), Srinagar, Jammu and Kashmir, 190001, India
| | - Apporva Chawla
- Khalsa College of Pharmacy, G.T. Road, Amritsar, Punjab, 143001, India
| | - Pooja Sharma
- Khalsa College of Pharmacy, G.T. Road, Amritsar, Punjab, 143001, India
| | - Prince Ahad Mir
- Khalsa College of Pharmacy, G.T. Road, Amritsar, Punjab, 143001, India
| | - Faheem Hyder Potoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, 1982, 31441, Dammam, Saudi Arabia
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, Zagreb, Croatia
| | - Ivan Reiner
- Department of Nursing Sciences, Catholic University of Croatia, Ilica 242, 10000, Zagreb, Croatia
| | - Dilek Arslan Ateşşahin
- Baskil Vocational School, Department of Plant and Animal Production, Fırat University, 23100, Elazıg, Turkey
| | | | - Reyaz Hassan Mir
- Pharmaceutical Chemistry Division, Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar, Kashmir, 190006, India.
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| |
Collapse
|
27
|
Aroosa M, Malik JA, Ahmed S, Bender O, Ahemad N, Anwar S. The evidence for repurposing anti-epileptic drugs to target cancer. Mol Biol Rep 2023; 50:7667-7680. [PMID: 37418080 PMCID: PMC10460753 DOI: 10.1007/s11033-023-08568-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/31/2023] [Indexed: 07/08/2023]
Abstract
Antiepileptic drugs are versatile drugs with the potential to be used in functional drug formulations with drug repurposing approaches. In the present review, we investigated the anticancer properties of antiepileptic drugs and interlinked cancer and epileptic pathways. Our focus was primarily on those drugs that have entered clinical trials with positive results and those that provided good results in preclinical studies. Many contributing factors make cancer therapy fail, like drug resistance, tumor heterogeneity, and cost; exploring all alternatives for efficient treatment is important. It is crucial to find new drug targets to find out new antitumor molecules from the already clinically validated and approved drugs utilizing drug repurposing methods. The advancements in genomics, proteomics, and other computational approaches speed up drug repurposing. This review summarizes the potential of antiepileptic drugs in different cancers and tumor progression in the brain. Valproic acid, oxcarbazepine, lacosamide, lamotrigine, and levetiracetam are the drugs that showed potential beneficial outcomes against different cancers. Antiepileptic drugs might be a good option for adjuvant cancer therapy, but there is a need to investigate further their efficacy in cancer therapy clinical trials.
Collapse
Affiliation(s)
- Mir Aroosa
- Department of Pharmacology, Jamia Hamdard, New Delhi, India
| | - Jonaid Ahmad Malik
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Rupnagar, Punjab, India
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Ropar, Ropar, India
| | - Sakeel Ahmed
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Ahmedabad, Gujarat, India
| | - Onur Bender
- Biotechnology Institute, Ankara University, Ankara, Turkey
| | - Nafees Ahemad
- School of Pharmacy, Monash University Malaysia, Jalan lagoon selatan, Petaling Jaya, Selangor, DE, Malaysia.
| | - Sirajudheen Anwar
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail, Saudi Arabia.
| |
Collapse
|
28
|
Gao X, Wang B, Huang Y, Wu M, Li Y, Li Y, Zhu X, Wu M. Role of the Nrf2 Signaling Pathway in Ovarian Aging: Potential Mechanism and Protective Strategies. Int J Mol Sci 2023; 24:13327. [PMID: 37686132 PMCID: PMC10488162 DOI: 10.3390/ijms241713327] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The ovary holds a significant role as a reproductive endocrine organ in women, and its aging process bears implications such as menopause, decreased fertility, and long-term health risks including osteoporosis, cardiovascular disorders, and cognitive decline. The phenomenon of oxidative stress is tightly linked to the aging metabolic processes. More and more studies have demonstrated that oxidative stress impacts both physiologic and pathologic ovarian aging, and the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway plays a crucial role in regulating the antioxidant response. Furthermore, various therapeutic approaches have been identified to ameliorate ovarian aging by modulating the Nrf2 pathway. This review summarizes the important role of the Nrf2/ Kelch-like ECH-associated protein 1 (Keap1) signaling pathway in regulating oxidative stress and influencing ovarian aging. Additionally, it highlights the therapeutic strategies aimed at targeting the Nrf2/Keap1 pathway.
Collapse
Affiliation(s)
- Xiaofan Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Bo Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Yibao Huang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Meng Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Yuting Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Yinuo Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Xiaoran Zhu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| | - Mingfu Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.G.); (B.W.); (Y.H.); (M.W.); (Y.L.); (Y.L.); (X.Z.)
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Wuhan 430030, China
| |
Collapse
|
29
|
Leblond P, Tresch-Bruneel E, Probst A, Néant N, Solas C, Sterin A, Boulanger T, Aerts I, Faure-Conter C, Bertozzi AI, Chastagner P, Entz-Werlé N, De Carli E, Deley MCL, Bouche G, André N. Phase I Study of a Combination of Fluvastatin and Celecoxib in Children with Relapsing/Refractory Low-Grade or High-Grade Glioma (FLUVABREX). Cancers (Basel) 2023; 15:cancers15072020. [PMID: 37046681 PMCID: PMC10093481 DOI: 10.3390/cancers15072020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Preclinical data support the activity of celecoxib and fluvastatin in high-grade (HGG) and low-grade gliomas (LGG). A phase I trial (NCT02115074) was designed to evaluate the safety of this combination in children with refractory/relapsed HGG and LGG using four dose levels of fluvastatin with a fixed daily dose of celecoxib. A Continual Reassessment Method was used for fluvastatin dose escalation. Dose-limiting toxicities (DLT) were determined on the first treatment cycle. Twenty patients were included. Ten LGG and ten HGG patients received a median of 3.5 treatment cycles. Two DLTs were reported: one grade 3 maculopapular rash (4 mg/kg dose level) and one grade 4 increase of Creatine Phospho-Kinase (6 mg/kg dose level). We identified the dose of 6 mg/kg/day as the recommended phase II dose (RP2D) of fluvastatin with celecoxib. Four patients with LGG continued treatment beyond 12 cycles because of stable disease, including one patient who received 23 treatment cycles. In children with refractory/relapsed glioma, the RP2D of fluvastatin with celecoxib is 6 mg/kg/day. The long-term stable diseases observed in LGG suggest a possible role of the combination in a maintenance setting, given its good tolerance and low cost for children living in low- and middle-income countries.
Collapse
|
30
|
Orecchioni S, Falvo P, Talarico G, Mitola G, Bravetti G, Mancuso P, Nicoli P, Bertolini F. Vinorelbine and Intermittent Cyclophosphamide Sensitize an Aggressive Myc-Driven B-Cell Lymphoma to Anti-PD-1 by an Immunological Memory Effective against Tumor Re-Challenge. J Clin Med 2023; 12:jcm12072535. [PMID: 37048617 PMCID: PMC10095342 DOI: 10.3390/jcm12072535] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/14/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
We have previously shown in triple-negative breast cancer (TNBC) models that a triple therapy (TT) including intermittent cyclophosphamide (C), vinorelbine (V), and anti-PD-1 activates antigen-presenting cells (APC) and generates stem like-T cells able to control local and metastatic tumor progression. In the present manuscript, we report the generation of a highly aggressive, anti-PD-1 resistant model of a high-grade, Myc-driven B-cell non-Hodgkin’s lymphoma (NHL) that can be controlled in vivo by TT but not by other chemotherapeutic agents, including cytarabine (AraC), platinum (P), and doxorubicin (D). The immunological memory elicited in tumor-bearing mice by TT (but not by other treatments) can effectively control NHL re-challenge even at very high inoculum doses. TT re-shaped the landscape of circulating innate NK cells and adaptive immune cells, including B and T cells, and significantly reduced exhausted CD4+ and CD8+ TIM3+PD-1+ T cells in the spleens of treated mice.
Collapse
Affiliation(s)
- Stefania Orecchioni
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Paolo Falvo
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Giovanna Talarico
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Giulia Mitola
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Giulia Bravetti
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Patrizia Mancuso
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
| | - Paola Nicoli
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Via Adamello 16, 20137 Milan, Italy
| | - Francesco Bertolini
- Laboratory of Hematology-Oncology, European Institute of Oncology IRCCS, Via Ripamonti 435, 20141 Milan, Italy
- Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, 20141 Milan, Italy
- Correspondence:
| |
Collapse
|
31
|
Islam S, Rahaman MH, Yu M, Noll B, Martin JH, Wang S, Head R. Anti-Leukaemic Activity of Rilpivirine Is Mediated by Aurora A Kinase Inhibition. Cancers (Basel) 2023; 15:cancers15041044. [PMID: 36831387 PMCID: PMC9954146 DOI: 10.3390/cancers15041044] [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: 12/23/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/10/2023] Open
Abstract
Acute myeloid leukaemia (AML) affects predominantly elderly people and has an incidence of 1% of all cancers and 2% of all cancer deaths. Despite using intensive chemotherapy and allogeneic stem cell transplantation, the treatment options for AML remain open for innovation. Thus, there is a need to explore alternative therapies such as less toxic targeted therapies in AML. Aurora A kinase is a well-established target for the treatment of various cancers, including AML. This kinase plays a pivotal role in the cell-division cycle, particularly in different stages of mitosis, and is also involved in many other cellular regulatory processes. In a previous study, we demonstrated that the anti-viral drug rilpivirine is an Aurora A kinase inhibitor. In the current study, we have further explored the selectivity of rilpivirine for Aurora A kinase inhibition by testing this drug against a panel of 429 kinases. Concurrently, we demonstrated that rilpivirine significantly inhibited the proliferation of AML cells in a time- and concentration-dependent manner that was preceded by G2/M cell-cycle arrest leading to the induction of apoptosis. Consistent with its kinase inhibitory role, rilpivirine modulated the expression of critical proteins in the Aurora A kinase-signalling pathway. Importantly, orally administered rilpivirine significantly inhibited tumour growth in an HL-60 xenograft model without showing body weight changes or other clinical signs of toxicity. Furthermore, rilpivirine enhanced the anti-proliferative efficacy of the conventional anti-leukaemic chemotherapeutic agent cytarabine. Collectively, these findings provide the stimulus to explore further the anti-leukaemic activity of the anti-viral drug rilpivirine.
Collapse
Affiliation(s)
- Saiful Islam
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Muhammed H. Rahaman
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Mingfeng Yu
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Benjamin Noll
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Jennifer H. Martin
- Centre for Human Drug Repurposing and Medicines Research, University of Newcastle, Newcastle, NSW 2305, Australia
| | - Shudong Wang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Richard Head
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
- Correspondence:
| |
Collapse
|
32
|
Roundhill EA, Pantziarka P, Liddle DE, Shaw LA, Albadrani G, Burchill SA. Exploiting the Stemness and Chemoresistance Transcriptome of Ewing Sarcoma to Identify Candidate Therapeutic Targets and Drug-Repurposing Candidates. Cancers (Basel) 2023; 15:cancers15030769. [PMID: 36765727 PMCID: PMC9913297 DOI: 10.3390/cancers15030769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/08/2023] [Accepted: 01/17/2023] [Indexed: 01/28/2023] Open
Abstract
Outcomes for most patients with Ewing sarcoma (ES) have remained unchanged for the last 30 years, emphasising the need for more effective and tolerable treatments. We have hypothesised that using small-molecule inhibitors to kill the self-renewing chemotherapy-resistant cells (Ewing sarcoma cancer stem-like cells; ES-CSCs) responsible for progression and relapse could improve outcomes and minimise treatment-induced morbidities. For the first time, we demonstrate that ABCG1, a potential oncogene in some cancers, is highly expressed in ES-CSCs independently of CD133. Using functional models, transcriptomics and a bespoke in silico drug-repurposing pipeline, we have prioritised a group of tractable small-molecule inhibitors for further preclinical studies. Consistent with the cellular origin of ES, 21 candidate molecular targets of pluripotency, stemness and chemoresistance were identified. Small-molecule inhibitors to 13 of the 21 molecular targets (62%) were identified. POU5F1/OCT4 was the most promising new therapeutic target in Ewing sarcoma, interacting with 10 of the 21 prioritised molecular targets and meriting further study. The majority of small-molecule inhibitors (72%) target one of two drug efflux proteins, p-glycoprotein (n = 168) or MRP1 (n = 13). In summary, we have identified a novel cell surface marker of ES-CSCs and cancer/non-cancer drugs to targets expressed by these cells that are worthy of further preclinical evaluation. If effective in preclinical models, these drugs and drug combinations might be repurposed for clinical evaluation in patients with ES.
Collapse
Affiliation(s)
- Elizabeth Ann Roundhill
- Children’s Cancer Research Group, Leeds Institute of Medical Research, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK
- Correspondence: (E.A.R.); (S.A.B.)
| | - Pan Pantziarka
- Anticancer Fund, Brusselsesteenweg 11, 1860 Meise, Belgium
| | - Danielle E. Liddle
- Children’s Cancer Research Group, Leeds Institute of Medical Research, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Lucy A. Shaw
- Children’s Cancer Research Group, Leeds Institute of Medical Research, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Ghadeer Albadrani
- Children’s Cancer Research Group, Leeds Institute of Medical Research, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK
| | - Susan Ann Burchill
- Children’s Cancer Research Group, Leeds Institute of Medical Research, St James’s University Hospital, Beckett Street, Leeds LS9 7TF, UK
- Correspondence: (E.A.R.); (S.A.B.)
| |
Collapse
|
33
|
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] [Download PDF] [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
|
34
|
Shankar E, Subramaniam V, Allimuthu D. Editorial: Adopting drug repurposing to overcome drug resistance in cancer. Front Cell Dev Biol 2023; 11:1191682. [PMID: 37187616 PMCID: PMC10175806 DOI: 10.3389/fcell.2023.1191682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023] Open
Affiliation(s)
- Eswar Shankar
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- *Correspondence: Dharmaraja Allimuthu, ; Eswar Shankar, ; Vish Subramaniam,
| | - Vish Subramaniam
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States
- Department of Mechanical and Aerospace Engineering, The Ohio State University, Columbus, OH, United States
- *Correspondence: Dharmaraja Allimuthu, ; Eswar Shankar, ; Vish Subramaniam,
| | - Dharmaraja Allimuthu
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur, Uttar Pradesh, India
- *Correspondence: Dharmaraja Allimuthu, ; Eswar Shankar, ; Vish Subramaniam,
| |
Collapse
|
35
|
Mteremko D, Chilongola J, Paluch AS, Chacha M. Targeting human thymidylate synthase: Ensemble-based virtual screening for drug repositioning and the role of water. J Mol Graph Model 2023; 118:108348. [PMID: 36257147 DOI: 10.1016/j.jmgm.2022.108348] [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: 06/07/2022] [Revised: 09/21/2022] [Accepted: 09/23/2022] [Indexed: 11/29/2022]
Abstract
A drug repositioning computational approach was carried to search inhibitors for human thymidylate synthase. An ensemble-based virtual screening of FDA-approved drugs showed the drugs Imatinib, Lumacaftor and Naldemedine to be likely candidates for repurposing. The role of water in the drug-receptor interactions was revealed by the application of an extended AutoDock scoring function that included the water forcefield. The binding affinity scores when hydrated ligands were docked were improved in the drugs considered. Further binding free energy calculations based on the Molecular Mechanics Poisson-Boltzmann Surface Area method revealed that Imatinib, Lumacaftor and Naldemedine scored -130.7 ± 28.1, -210.6 ± 29.9 and -238.0 ± 25.4 kJ/mol, respectively, showing good binding affinity for the candidates considered. Overall, the analysis of the molecular dynamics trajectory of the receptor-drug complexes revealed stable structures for Imatinib, Lumacaftor and Naldemedine, for the entire simulation time.
Collapse
Affiliation(s)
- Denis Mteremko
- The Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania.
| | - Jaffu Chilongola
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Andrew S Paluch
- Department of Chemical, Paper, and Biomedical Engineering, Miami University, Oxford, OH, 45056, USA
| | - Musa Chacha
- The Nelson Mandela African Institution of Science and Technology, Arusha, Tanzania; Arusha Technical College, Arusha, Tanzania
| |
Collapse
|
36
|
Repurposing Drugs in Small Animal Oncology. Animals (Basel) 2022; 13:ani13010139. [PMID: 36611747 PMCID: PMC9817697 DOI: 10.3390/ani13010139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/18/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Repurposing drugs in oncology consists of using off-label drugs that are licensed for various non-oncological medical conditions to treat cancer. Repurposing drugs has the advantage of using drugs that are already commercialized, with known mechanisms of action, proven safety profiles, and known toxicology, pharmacokinetics and pharmacodynamics, and posology. These drugs are usually cheaper than new anti-cancer drugs and thus more affordable, even in low-income countries. The interest in repurposed anti-cancer drugs has led to numerous in vivo and in vitro studies, with some promising results. Some randomized clinical trials have also been performed in humans, with certain drugs showing some degree of clinical efficacy, but the true clinical benefit for most of these drugs remains unknown. Repurposing drugs in veterinary oncology is a very new concept and only a few studies have been published so far. In this review, we summarize both the benefits and challenges of using repurposed anti-cancer drugs; we report and discuss the most relevant studies that have been previously published in small animal oncology, and we suggest potential drugs that could be clinically investigated for anti-cancer treatment in dogs and cats.
Collapse
|
37
|
Jamialahmadi O, Salehabadi E, Hashemi-Najafabadi S, Motamedian E, Bagheri F, Mancina RM, Romeo S. Cellular Genome-Scale Metabolic Modeling Identifies New Potential Drug Targets Against Hepatocellular Carcinoma. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2022; 26:671-682. [PMID: 36508280 DOI: 10.1089/omi.2022.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Genome-scale metabolic modeling (GEM) is one of the key approaches to unpack cancer metabolism and for discovery of new drug targets. In this study, we report the Transcriptional Regulated Flux Balance Analysis-CORE (TRFBA-), an algorithm for GEM using key growth-correlated reactions using hepatocellular carcinoma (HCC), an important global health burden, as a case study. We generated a HepG2 cell-specific GEM by integrating this cell line transcriptomic data with a generic human metabolic model to forecast potential drug targets for HCC. A total of 108 essential genes for growth were predicted by the TRFBA-CORE. These genes were enriched for metabolic pathways involved in cholesterol, sterol, and steroid biosynthesis. Furthermore, we silenced a predicted essential gene, 11-beta dehydrogenase hydroxysteroid type 2 (HSD11B2), in HepG2 cells resulting in a reduction in cell viability. To further identify novel potential drug targets in HCC, we examined the effect of nine drugs targeting the essential genes, and observed that most drugs inhibited the growth of HepG2 cells. Some of these drugs in this model performed better than Sorafenib, the first-line therapeutic against HCC. A HepG2 cell-specific GEM highlights sterol metabolism to be essential for cell growth. HSD11B2 downregulation results in lower cell growth. Most of the compounds, selected by drug repurposing approach, show a significant inhibitory effect on cell growth in a wide range of concentrations. These findings offer new molecular leads for drug discovery for hepatic cancer while also illustrating the importance of GEM and drug repurposing in cancer therapeutics innovation.
Collapse
Affiliation(s)
- Oveis Jamialahmadi
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Salehabadi
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Sameereh Hashemi-Najafabadi
- Department of Biomedical Engineering, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Motamedian
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology and Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Rosellina Margherita Mancina
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Stefano Romeo
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.,Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy.,Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
38
|
Capistrano I R, Paul S, Boere I, Pantziarka P, Chopra S, Nout RA, Bouche G. Drug repurposing as a potential source of innovative therapies in cervical cancer. Int J Gynecol Cancer 2022; 32:1377-1386. [PMID: 36137575 PMCID: PMC9664115 DOI: 10.1136/ijgc-2022-003585] [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: 04/22/2022] [Accepted: 08/19/2022] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE Drug repurposing is an alternative development pathway that utilizes the properties of drugs approved for other diseases and builds on available safety and pharmacological data to develop the drug as a potential treatment for other diseases. A literature-based approach was performed to identify drug repurposing opportunities in cervical cancer to inform future research and trials. METHODS We queried PubMed for each drug included in two databases (ReDO_DB and CDcervix_DB, which include 300+ non-cancer drugs and 200+ cancer drugs not used in cervical cancer, respectively) and manually assessed all abstracts for relevance and activity in cervix cancer, and type of evidence. Subsequently, we also performed a search of clinical trial databases where we generated a list of registered trials in cervical cancer with all drugs from our databases. RESULTS Of the 534 drugs from both databases, 174 (33%) had at least one relevant abstract or registered trial in cervical cancer. 94 (18%) drugs had at least human data available, and 52 (10%) drugs were evaluated in registered trials. To prioritize drugs to consider for future trials, all 174 drugs were further assessed for strength of scientific rationale, feasibility for integration in cervical cancer standard of care, evidence of radiosensitization, and potential mechanism of action. Out of the 174 drugs, 38 (22%) potential drug candidates were selected. CONCLUSION This study resulted in a list of candidate drugs for potential evaluation in cervical cancer. Many drugs might warrant additional (pre)clinical investigation, which could be done in a coordinated manner using platform trials.
Collapse
Affiliation(s)
| | - Sonz Paul
- Department of Radiation Oncology, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Ingrid Boere
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Pan Pantziarka
- Anticancer Fund, Meise, Belgium
- The George Pantziarka TP53 Trust, London, UK
| | - Supriya Chopra
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Homi Bhabha National Institute, Navi Mumbai, India
| | - Remi A Nout
- Department of Radiotherapy, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, Netherlands
| | | |
Collapse
|
39
|
Ren J, Wang B, Wu Q, Wang G. Combination of niclosamide and current therapies to overcome resistance for cancer: New frontiers for an old drug. Biomed Pharmacother 2022; 155:113789. [DOI: 10.1016/j.biopha.2022.113789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/02/2022] Open
|
40
|
Sultana T, Jan U, Lee H, Lee H, Lee JI. Exceptional Repositioning of Dog Dewormer: Fenbendazole Fever. Curr Issues Mol Biol 2022; 44:4977-4986. [PMID: 36286053 PMCID: PMC9600184 DOI: 10.3390/cimb44100338] [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: 09/24/2022] [Revised: 10/11/2022] [Accepted: 10/11/2022] [Indexed: 12/02/2022] Open
Abstract
Fenbendazole (FZ) is a benzimidazole carbamate drug with broad-spectrum antiparasitic activity in humans and animals. The mechanism of action of FZ is associated with microtubular polymerization inhibition and glucose uptake blockade resulting in reduced glycogen stores and decreased ATP formation in the adult stages of susceptible parasites. A completely cured case of lung cancer became known globally and greatly influenced the cancer community in South Korea. Desperate Korean patients with cancer began self-administering FZ without their physician’s knowledge, which interfered with the outcome of the cancer treatment planned by their oncologists. On the basis of presented evidence, this review provides valuable information from PubMed, Naver, Google Scholar, and Social Network Services (SNS) on the effects of FZ in a broad range of preclinical studies on cancer. In addition, we suggest investigating the self-administration of products, including supplements, herbs, or bioactive compounds, by patients to circumvent waiting for long and costly FZ clinical trials.
Collapse
Affiliation(s)
- Tania Sultana
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Umair Jan
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Hyunsu Lee
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Hyejin Lee
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
| | - Jeong Ik Lee
- Regenerative Medicine Laboratory, Center for Stem Cell Research, Department of Biomedical Science and Technology, Institute of Biomedical Science and Technology, Konkuk University, Seoul 05029, Korea
- Department of Veterinary Obstetrics and Theriogenology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Korea
- Correspondence: ; Tel.: +82-2-2049-6234
| |
Collapse
|
41
|
Patil KS, Hajare AA, Manjappa AS, More HN, Disouza JI. Design, Development, In Silico, and In Vitro Characterization of Camptothecin-Loaded Mixed Micelles: In Vitro Testing of Verapamil and Ranolazine for Repurposing as Coadjuvant Therapy in Cancer. J Pharm Innov 2022. [DOI: 10.1007/s12247-022-09688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Li CL, Fang ZX, Wu Z, Hou YY, Wu HT, Liu J. Repurposed itraconazole for use in the treatment of malignancies as a promising therapeutic strategy. Biomed Pharmacother 2022; 154:113616. [PMID: 36055112 DOI: 10.1016/j.biopha.2022.113616] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/24/2022] [Accepted: 08/27/2022] [Indexed: 02/05/2023] Open
Abstract
Understanding cancer biology and the development of novel agents for cancer treatment has always been the goal of cancer researchers. However, the research and development of new drugs is hindered by its long development time, exorbitant cost, high regulatory hurdles, and staggering failure rates. Given the challenges involved drug development for cancer therapies, alternative strategies, in particular the repurposing of 'old' drugs that have been approved for other indications, are attractive. Itraconazole is an FDA-approved anti-fungal drug of the triazole class, and has been used clinically for more than 30 years. Recent drug repurposing screens revealed itraconazole exerts anti-cancer activity via inhibiting angiogenesis and multiple oncogenic signaling pathways. To explore the potential utilization of itraconazole in different types of malignancies, we retrieved the published literature relating to itraconazole in cancer and reviewed the mechanisms of itraconazole in preclinical and clinical cancer studies. Current research predicts the hedgehog signaling pathway as the main target by which itraconazole inhibits a variety of solid and hematological cancers. As clinical trial results become available, itraconazole could emerge as a new antitumor drug that can be used in combination with first-line antitumor drugs.
Collapse
Affiliation(s)
- Chun-Lan Li
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, China; Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou 515041, China
| | - Ze-Xuan Fang
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, China; Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou 515041, China
| | - Zheng Wu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, China; Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou 515041, China
| | - Yan-Yu Hou
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, China; Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou 515041, China
| | - Hua-Tao Wu
- Department of General Surgery, First Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
| | - Jing Liu
- Guangdong Provincial Key Laboratory for Diagnosis and Treatment of Breast Cancer, Cancer Hospital of Shantou University Medical College, Shantou 515041, China
- Department of Physiology/Changjiang Scholar's Laboratory, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
43
|
Islam S, Teo T, Kumarasiri M, Slater M, Martin JH, Wang S, Head R. Combined In Silico and In Vitro Evidence Supporting an Aurora A Kinase Inhibitory Role of the Anti-Viral Drug Rilpivirine and an Anti-Proliferative Influence on Cancer Cells. Pharmaceuticals (Basel) 2022; 15:ph15101186. [PMID: 36297298 PMCID: PMC9607353 DOI: 10.3390/ph15101186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/20/2022] Open
Abstract
The global burden of cancer necessitates rapid and ongoing development of effective cancer therapies. One promising approach in this context is the repurposing of existing non-cancer drugs for cancer indications. A key to this approach is selecting the cellular targets against which to identify novel repurposed drugs for pre-clinical analysis. Protein kinases are highly sought-after anticancer drug targets since dysregulation of kinases is the hallmark of cancer. To identify potential kinase-targeted drug candidates from the existing portfolio of non-cancer therapeutics, we used combined in silico and in vitro approaches, including ligand-based 3D screening followed by biochemical and cellular assessments. This strategy revealed that the anti-viral drug rilpivirine is an Aurora A kinase inhibitor. In view of previous findings implicating Aurora A kinase in abnormal cell cycle regulation, we also examined the influence of rilpivirine on the growth of T47D breast cancer cells. Herein, we detail the identification of rilpivirine as an Aurora A kinase inhibitor, its molecular basis of inhibitory activity towards this kinase, and its Aurora A-mediated anticancer mechanisms in T47D cells. Our results illustrate the value of integrated in silico and in vitro screening strategies in identifying repurposed drug candidates and provide a scientific basis for further exploring the potential anticancer properties of the anti-viral drug rilpivirine.
Collapse
Affiliation(s)
- Saiful Islam
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Theodosia Teo
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Malika Kumarasiri
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Martin Slater
- Cresset Discovery, New Cambridge House, Litlington, Royston SG8 0SS, UK
| | - Jennifer H. Martin
- Centre for Human Drug Repurposing and Medicines Research, University of Newcastle, Newcastle, NSW 2305, Australia
| | - Shudong Wang
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Richard Head
- Drug Discovery and Development, Clinical and Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
- Correspondence:
| |
Collapse
|
44
|
Simoens S, Abdallah K, Barbier L, Lacosta TB, Blonda A, Car E, Claessens Z, Desmet T, De Sutter E, Govaerts L, Janssens R, Lalova T, Moorkens E, Saesen R, Schoefs E, Vandenplas Y, Van Overbeeke E, Verbaanderd C, Huys I. How to balance valuable innovation with affordable access to medicines in Belgium? Front Pharmacol 2022; 13:960701. [PMID: 36188534 PMCID: PMC9523170 DOI: 10.3389/fphar.2022.960701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Background: Countries are struggling to provide affordable access to medicines while supporting the market entry of innovative, expensive products. This Perspective aims to discuss challenges and avenues for balancing health care system objectives of access, affordability and innovation related to medicines in Belgium (and in other countries). Methods: This Perspective focuses on the R&D, regulatory approval and market access phases, with particular attention to oncology medicines, precision medicines, orphan medicines, advanced therapies, repurposed medicines, generics and biosimilars. The authors conducted a narrative review of the peer-reviewed literature, of the grey literature (such as policy documents and reports of consultancy agencies), and of their own research. Results: Health care stakeholders need to consider various initiatives for balancing innovation with access to medicines, which relate to clinical and non-clinical outcomes (e.g. supporting the conduct of pragmatic clinical trials, treatment optimisation and patient preference studies, optimising the use of real-world evidence in market access decision making), value assessment (e.g. increasing the transparency of the reimbursement system and criteria, tailoring the design of managed entry agreements to specific types of uncertainty), affordability (e.g. harnessing the role of generics and biosimilars in encouraging price competition, maximising opportunities for personalising and repurposing medicines) and access mechanisms (e.g. promoting collaboration and early dialogue between stakeholders including patients). Conclusion: Although there is no silver bullet that can balance valuable innovation with affordable access to medicines, (Belgian) policy and decision makers should continue to explore initiatives that exploit the potential of both the on-patent and off-patent pharmaceutical markets.
Collapse
Affiliation(s)
- Steven Simoens
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Khadidja Abdallah
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Liese Barbier
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | | | - Alessandra Blonda
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Elif Car
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Zilke Claessens
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Thomas Desmet
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Evelien De Sutter
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Laurenz Govaerts
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Rosanne Janssens
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Teodora Lalova
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- KU Leuven Centre for IT & IP Law (CiTiP), Leuven, Belgium
| | - Evelien Moorkens
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Robbe Saesen
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- European Organisation for Research and Treatment of Cancer, Brussels, Belgium
| | - Elise Schoefs
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Yannick Vandenplas
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Eline Van Overbeeke
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| | - Ciska Verbaanderd
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
- Anticancer Fund, Strombeek-Bever, Brussels, Belgium
| | - Isabelle Huys
- KU Leuven Department of Pharmaceutical and Pharmacological Sciences, Leuven, Belgium
| |
Collapse
|
45
|
Solernó LM, Sobol NT, Gottardo MF, Capobianco CS, Ferrero MR, Vásquez L, Alonso DF, Garona J. Propranolol blocks osteosarcoma cell cycle progression, inhibits angiogenesis and slows xenograft growth in combination with cisplatin-based chemotherapy. Sci Rep 2022; 12:15058. [PMID: 36075937 PMCID: PMC9458647 DOI: 10.1038/s41598-022-18324-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Osteosarcoma is still associated with limited response to standard-of-care therapy and alarmingly elevated mortality rates, especially in low- and middle-income countries. Despite multiple efforts to repurpose β-blocker propranolol in oncology, its potential application in osteosarcoma management remains largely unexplored. Considering the unsatisfied clinical needs of this aggressive disease, we evaluated the antitumoral activity of propranolol using different in vitro and in vivo osteosarcoma preclinical models, alone or in addition to chemotherapy. Propranolol significantly impaired cellular growth in β2-adrenergic receptor-expressing MG-63 and U-2OS cells, and was capable of blocking growth-stimulating effects triggered by catecholamines. siRNA-mediated ADRB2 knockdown in MG-63 cells was associated with decreased cell survival and a significant attenuation of PPN anti-osteosarcoma activity. Direct cytostatic effects of propranolol were independent of apoptosis induction and were associated with reduced mitosis, G0/G1 cell cycle arrest and a significant down-regulation of cell cycle regulator Cyclin D1. Moreover, colony formation, 3D spheroid growth, cell chemotaxis and capillary-like tube formation were drastically impaired after propranolol treatment. Interestingly, anti-migratory activity of β-blocker was associated with altered actin cytoskeleton dynamics. In vivo, propranolol treatment (10 mg/kg/day i.p.) reduced the early angiogenic response triggered by MG-63 cells in nude mice. Synergistic effects were observed in vitro after combining propranolol with chemotherapeutic agent cisplatin. Sustained administration of propranolol (10 mg/kg/day i.p., five days a week), alone and especially in addition to low-dose metronomic cisplatin (2 mg/kg/day i.p., three times a week), markedly reduced xenograft progression. After histological analysis, propranolol and cisplatin combination resulted in low tumor mitotic index and increased tumor necrosis. β-blockade using propranolol seems to be an achievable and cost-effective therapeutic approach to modulate osteosarcoma aggressiveness. Further translational studies of propranolol repurposing in osteosarcoma are warranted.
Collapse
Affiliation(s)
- Luisina M Solernó
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Natasha T Sobol
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - María F Gottardo
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina
| | - Carla S Capobianco
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina
| | - Maximiliano R Ferrero
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany.,Biomedicine Research Institute of Buenos Aires (IBioBA), Buenos Aires, Argentina
| | - Liliana Vásquez
- Precision Medicine Research Center, School of Medicine, University of San Martín de Porres, Lima, Perú
| | - Daniel F Alonso
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina.,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina.,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina
| | - Juan Garona
- Center for Molecular and Translational Oncology (COMTra), Science and Technology Department, National University of Quilmes, Buenos Aires, Argentina. .,Center for Translational Medicine, El Cruce "Néstor Kirchner" Hospital, Buenos Aires, Argentina. .,National Council of Scientific and Technical Research (CONICET), Buenos Aires, Argentina.
| |
Collapse
|
46
|
Pitavastatin and Ivermectin Enhance the Efficacy of Paclitaxel in Chemoresistant High-Grade Serous Carcinoma. Cancers (Basel) 2022; 14:cancers14184357. [PMID: 36139522 PMCID: PMC9496819 DOI: 10.3390/cancers14184357] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/01/2022] [Accepted: 09/03/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary The main challenge in high-grade serous carcinoma management is to unveil therapeutic approaches to overcome chemoresistance. Drug combinations and repurposing of non-oncological agents are attractive strategies that allow for higher efficacy, decreased toxicity, and the overcoming of chemoresistance. Several non-oncological drugs display an effective anti-cancer activity and have been studied to be repurposed in multi-drug resistant neoplasms. The purpose of our study was to explore whether combining Paclitaxel with repurposed drugs (Pitavastatin, Metformin, Ivermectin, Itraconazole and Alendronate) led to a therapeutic benefit. Our results showed that the combination of Paclitaxel with Pitavastatin or Ivermectin demonstrates the highest cytotoxic effect and the strongest synergism among all combinations for two chemoresistant cell lines. Thus, the combination of these repurposed drugs with Paclitaxel could be a particularly valuable strategy to treat ovarian cancer patients with intrinsic or acquired chemoresistance. Abstract Chemotherapy is a hallmark in high-grade serous carcinoma management; however, chemoresistance and side effects lead to therapeutic interruption. Combining repurposed drugs with chemotherapy has the potential to improve antineoplastic efficacy, since drugs can have independent mechanisms of action and suppress different pathways simultaneously. This study aimed to explore whether the combination of Paclitaxel with repurposed drugs led to a therapeutic benefit. Thus, we evaluated the cytotoxic effects of Paclitaxel alone and in combination with several repurposed drugs (Pitavastatin, Metformin, Ivermectin, Itraconazole and Alendronate) in two tumor chemoresistant (OVCAR8 and OVCAR8 PTX R P) and a non-tumoral (HOSE6.3) cell lines. Cellular viability was assessed using Presto Blue assay, and the synergistic interactions were evaluated using Chou–Talalay, Bliss Independence and Highest Single Agent reference models. The combination of Paclitaxel with Pitavastatin or Ivermectin showed the highest cytotoxic effect and the strongest synergism among all combinations for both chemoresistant cell lines, resulting in a chemotherapeutic effect superior to both drugs alone. Almost all the repurposed drugs in combination with Paclitaxel presented a safe pharmacological profile in non-tumoral cells. Overall, we suggest that Pitavastatin and Ivermectin could act synergistically in combination with Paclitaxel, being promising two-drug combinations for high-grade serous carcinoma management.
Collapse
|
47
|
Gao Y, Kruithof-de Julio M, Peng RW, Dorn P. Organoids as a Model for Precision Medicine in Malignant Pleural Mesothelioma: Where Are We Today? Cancers (Basel) 2022; 14:3758. [PMID: 35954422 PMCID: PMC9367391 DOI: 10.3390/cancers14153758] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 12/10/2022] Open
Abstract
MPM is an aggressive tumor originating from pleural mesothelial cells. A characteristic feature of the disease is the dominant prevalence of therapeutically intractable inactivating alterations in TSGs, making MPM one of the most difficult cancers to treat and the epitome of a cancer characterized by a significant lack of therapy options and an extremely poor prognosis (5-year survival rate of only 5% to 10%). Extensive interpatient heterogeneity poses another major challenge for targeted therapy of MPM, warranting stratified therapy for specific subgroups of MPM patients. Accurate preclinical models are critical for the discovery of new therapies and the development of personalized medicine. Organoids, an in vitro 'organ-like' 3D structure derived from patient tumor tissue that faithfully mimics the biology and complex architecture of cancer and largely overcomes the limitations of other existing models, are the next-generation tumor model. Although organoids have been successfully produced and used in many cancers, the development of MPM organoids is still in its infancy. Here, we provide an overview of recent advances in cancer organoids, focusing on the progress and challenges in MPM organoid development. We also elaborate the potential of MPM organoids for understanding MPM pathobiology, discovering new therapeutic targets, and developing personalized treatments for MPM patients.
Collapse
Affiliation(s)
- Yanyun Gao
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| | - Marianna Kruithof-de Julio
- Urology Research Laboratory, Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland;
- Department for BioMedical Research (DBMR), Translation Organoid Research, University of Bern, 3008 Bern, Switzerland
- Department of Urology, Inselspital, Bern University Hospital, 3008 Bern, Switzerland
| | - Ren-Wang Peng
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, University of Bern, 3008 Bern, Switzerland;
- Department of BioMedical Research (DBMR), Oncology-Thoracic Malignancies (OTM), University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
48
|
Rossi M, Talbot J, Piris P, Grand ML, Montero MP, Matteudi M, Agavnian-Couquiaud E, Appay R, Keime C, Williamson D, Buric D, Bourgarel V, Padovani L, Clifford SC, Ayrault O, Pasquier E, André N, Carré M. Beta-blockers disrupt mitochondrial bioenergetics and increase radiotherapy efficacy independently of beta-adrenergic receptors in medulloblastoma. EBioMedicine 2022; 82:104149. [PMID: 35816899 PMCID: PMC9283511 DOI: 10.1016/j.ebiom.2022.104149] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 11/03/2022] Open
Abstract
Background Medulloblastoma is the most frequent brain malignancy of childhood. The current multimodal treatment comes at the expense of serious and often long-lasting side effects. Drug repurposing is a strategy to fast-track anti-cancer therapy with low toxicity. Here, we showed the ability of β-blockers to potentiate radiotherapy in medulloblastoma with bad prognosis. Methods Medulloblastoma cell lines, patient-derived xenograft cells, 3D spheroids and an innovative cerebellar organotypic model were used to identify synergistic interactions between β-blockers and ionising radiations. Gene expression profiles of β-adrenergic receptors were analysed in medulloblastoma samples from 240 patients. Signaling pathways were explored by RT-qPCR, RNA interference, western blotting and RNA sequencing. Medulloblastoma cell bioenergetics were evaluated by measuring the oxygen consumption rate, the extracellular acidification rate and superoxide production. Findings Low concentrations of β-blockers significantly potentiated clinically relevant radiation protocols. Although patient biopsies showed detectable expression of β-adrenergic receptors, the ability of the repurposed drugs to potentiate ionising radiations did not result from the inhibition of the canonical signaling pathway. We highlighted that the efficacy of the combinatorial treatment relied on a metabolic catastrophe that deprives medulloblastoma cells of their adaptive bioenergetics capacities. This led to an overproduction of superoxide radicals and ultimately to an increase in ionising radiations-mediated DNA damages. Interpretation These data provide the evidence of the efficacy of β-blockers as potentiators of radiotherapy in medulloblastoma, which may help improve the treatment and quality of life of children with high-risk brain tumours. Funding This study was funded by institutional grants and charities.
Collapse
|
49
|
Kammoun AK, Hegazy MA, Khedr A, Awan ZA, Khayat MT, Al-Sawahli MM. Etodolac Fortified Sodium Deoxycholate Stabilized Zein Nanoplatforms for Augmented Repositioning Profile in Human Hepatocellular Carcinoma: Assessment of Bioaccessibility, Anti-Proliferation, Pro-Apoptosis and Oxidant Potentials in HepG2 Cells. Pharmaceuticals (Basel) 2022; 15:916. [PMID: 35893740 PMCID: PMC9331642 DOI: 10.3390/ph15080916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/29/2022] [Accepted: 07/08/2022] [Indexed: 02/04/2023] Open
Abstract
This work aimed to enhance the purposing profile of Etodolac (ETD) in Human Hepatocellular Carcinoma (HCC) HepG2 cells using sodium deoxycholate stabilized zein nanospheres (ETD-SDZN NSs). ETD-SDZN NSs were formulated using the nan-precipitation method and were characterized, in particular, in terms of mean particle size, zeta potential, encapsulation efficiency, colloidal stability and bioaccessibility. Estimations of cytotoxicity, cellular uptake, cell cycle progression, Annexin-V staining, mRNA expression of apoptotic genes and oxidative stress evaluations were conducted. The ETD-SDZN NSs selected formula obtained an average particle size of 113.6 ± 7.4 nm, a zeta potential value of 32.7 ± 2.3 mV, an encapsulation efficiency of 93.3 ± 5.2%, enhanced bioaccessibility and significantly reduced IC50 against HepG2 cells, by approximately 13 times. There was also enhanced cellular uptake, accumulation in G2-M phase and elevated percentage cells in pre-G1 phase, significant elevated mRNA expression of P53, significant reduced expression of Cyclin-dependent kinase 1 (CDK1) and Cyclooxygenase-2 (COX-2) with enhanced oxidative stress by reducing glutathione reductase (GR) level, ameliorated reactive oxygen species (ROS) generation and lipid peroxidation outputs. ETD-SDZN NSs obtained a supreme cell death-inducing profile toward HepG2 cells compared to free ETD. The method of formulation was successful in acquiring the promising profile of ETD in HCC as a therapeutic molecule due to ameliorated cellular uptake, proapoptotic and oxidant potentials.
Collapse
Affiliation(s)
- Ahmed K. Kammoun
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia; (A.K.K.); (A.K.); (M.T.K.)
| | - Maha A. Hegazy
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt;
| | - Alaa Khedr
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia; (A.K.K.); (A.K.); (M.T.K.)
| | - Zuhier Ahmed Awan
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Maan T. Khayat
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia; (A.K.K.); (A.K.); (M.T.K.)
| | | |
Collapse
|
50
|
Liu Q, Zeng A, Liu Z, Wu C, Song L. Liver organoids: From fabrication to application in liver diseases. Front Physiol 2022; 13:956244. [PMID: 35923228 PMCID: PMC9340459 DOI: 10.3389/fphys.2022.956244] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022] Open
Abstract
As the largest internal organ, the liver is the key hub for many physiological processes. Previous research on the liver has been mainly conducted on animal models and cell lines, in which not only there are deficiencies in species variability and retention of heritable material, but it is also difficult for primary hepatocytes to maintain their metabolic functions after in vitro expansion. Because of the increased burden of liver disease worldwide, there is a growing demand for 3D in vitro liver models—Liver Organoids. Based on the type of initiation cells, the liver organoid can be classified as PSC-derived or ASC-derived. Liver organoids originated from ASC or primary sclerosing cholangitis, which are co-cultured in matrix gel with components such as stromal cells or immune cells, and eventually form three-dimensional structures in the presence of cytokines. Liver organoids have already made progress in drug screening, individual medicine and disease modeling with hereditary liver diseases, alcoholic or non-alcoholic liver diseases and primary liver cancer. In this review, we summarize the generation process of liver organoids and the current clinical applications, including disease modeling, drug screening and individual medical treatment, which provide new perspectives for liver physiology and disease research.
Collapse
Affiliation(s)
- Qianglin Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, China
| | - Zibo Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chunjie Wu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunjie Wu, ; Linjiang Song,
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Chunjie Wu, ; Linjiang Song,
| |
Collapse
|