1
|
De Silva MI, Gan HK, Bardy C. Repurposing trifluoperazine for glioblastoma treatment. Trends Pharmacol Sci 2025:S0165-6147(25)00045-8. [PMID: 40300936 DOI: 10.1016/j.tips.2025.03.005] [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: 02/10/2025] [Revised: 03/16/2025] [Accepted: 03/16/2025] [Indexed: 05/01/2025]
Abstract
Glioblastoma (GBM) remains a therapeutic challenge due to its heterogeneity and plasticity, which drive treatment resistance, especially when compounded by interactions with the brain microenvironment. Recent preclinical evidence indicates that trifluoperazine (TFP) inhibits treatment-induced malignant reprogramming of tumour cells, potentially helping to reduce tumour plasticity. TFP targets calmodulin, dopamine receptors, and stress-responsive proteins (nuclear protein 1, NUPR1). Through these mechanisms, TFP has been shown to reduce tumour growth, sensitise tumours to chemoradiotherapy, and prolong survival in xenograft animal models. The clinical safety profile of TFP is well known from its use as an antipsychotic, and recent preclinical evidence further indicates that TFP has low toxicity to healthy neurons and glia despite transient functional effects on dopamine receptors. This Opinion explores TFP mechanisms of action and clinical activity to assess its suitability as a repurposed therapeutic option for GBM.
Collapse
Affiliation(s)
- Manam Inushi De Silva
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia
| | - Hui K Gan
- Cancer Therapies and Biology Group, Centre of Research Excellence in Brain Tumours, Olivia Newton-John Cancer Wellness and Research Centre, Austin Hospital, Heidelberg, Melbourne, VIC, Australia; La Trobe University School of Cancer Medicine, and Department of Medicine, University of Melbourne, Heidelberg, Melbourne, VIC, Australia
| | - Cedric Bardy
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia; South Australian Health and Medical Research Institute (SAHMRI), Laboratory for Human Neurophysiology and Genetics, Adelaide, SA, Australia.
| |
Collapse
|
2
|
Wan Z, Sun X, Li Y, Chu T, Hao X, Cao Y, Zhang P. Applications of Artificial Intelligence in Drug Repurposing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411325. [PMID: 40047357 PMCID: PMC11984889 DOI: 10.1002/advs.202411325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 12/12/2024] [Indexed: 04/12/2025]
Abstract
Drug repurposing identifies new therapeutic uses for the existing drugs originally developed for different indications, aiming at capitalizing on the established safety and efficacy profiles of known drugs. Thus, it is beneficial to bypass of early stages of drug development, and to reduction of the time and cost associated with bringing new therapies to market. Traditional experimental methods are often time-consuming and expensive, making artificial intelligence (AI) a promising alternative due to its lower cost, computational advantages, and ability to uncover hidden patterns. This review focuses on the availability of AI algorithms in drug development, and their positive and specific roles in revealing repurposing of the existing drugs, especially being integrated with virtual screening. It is shown that the existing AI algorithms excel at analyzing large-scale datasets, identifying the complicated patterns of drug responses from these datasets, and making predictions for potential drug repurposing. Building on these insights, challenges remain in developing efficient AI algorithms and future research, including integrating drug-related data across databases for better repurposing, enhancing AI computational efficiency, and advancing personalized medicine.
Collapse
Affiliation(s)
- Zhaoman Wan
- State Key Laboratory of Common Mechanism Research for Major DiseasesSuzhou Institute of Systems MedicineChinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhouJiangsu215123China
| | - Xinran Sun
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100193China
| | - Yi Li
- Hunan Agriculture University College of Plant ProtectionChangshaHunan410128China
| | - Tianyao Chu
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Xueyu Hao
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| | - Yang Cao
- College of Life SciencesSichuan UniversityChengduSichuan610041China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth DefectsBeijing Pediatric Research InstituteMOE Key Laboratory of Major Diseases in ChildrenRare Disease CenterBeijing Children's HospitalCapital Medical UniversityNational Center for Children's HealthBeijing100045China
| |
Collapse
|
3
|
Owczarzy A, Kulig K, Morak-Młodawska B, Jeleń M, Muhammetoglu T, Rogóż W, Maciążek-Jurczyk M. Human Serum Albumin and Human Serum Albumin Nanoparticles as Carriers of 10-(2'-Pyrimidyl)-3,6-diazaphenothiazine: In Vitro Spectroscopic Studies. Molecules 2025; 30:315. [PMID: 39860185 PMCID: PMC11767950 DOI: 10.3390/molecules30020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 12/27/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Human serum albumin (HSA) plays a fundamental role in the human body, including the transport of exogenous and endogenous substances. HSA is also a biopolymer with a great medical and pharmaceutical potential. Due to nontoxicity and biocompatibility, this protein can be used as a nanocarrier. 10-(2'-Pyrimidyl)-3,6-diazaphenothiazine (10-Pyr-3,6-DAPT) is a phenothiazine showing high anticancer potential in vitro against glioma, melanoma and breast cancer cells. Additionally, this compound is characterized by selectivity of action towards MCF-7 breast cancer and has low cytotoxicity towards normal cells. Considering the promising pharmacological potential of this compound and using spectroscopic techniques, HSA and human serum albumin nanoparticles (HSA-NP) were tested as carriers of this molecule. Based on the obtained data and the appropriate mathematical models (Stern-Volmer and Klotz models), it can be concluded that 10-Pyr-3,6-DAPT probably forms a weak (Ka = (5.24 ± 0.57) × 104 and Ka = (4.67 ± 0.59) × 104) for excitation wavelengths λex 275 nm and λex 295 nm, respectively) static complex (kq > 1010) with HSA (at Sudlow site II (subdomain IIIA), and the phenomenon of it having both strong therapeutic and toxic effects is possible. High encapsulation efficiency of 10-Pyr-3,6-DAPT into the HSA-NPs was obtained, and the changes in albumin secondary structure due to the presence of 10-Pyr-3,6-DAPT were registered. Based on the data presented, it can be concluded that due to the high toxic effects of 10-Pyr-3,6-DAPT, a better carrier may be HSA-NPs.
Collapse
Affiliation(s)
- Aleksandra Owczarzy
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Karolina Kulig
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-005 Katowice, Poland; (B.M.-M.); (M.J.)
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-005 Katowice, Poland; (B.M.-M.); (M.J.)
| | | | - Wojciech Rogóż
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| | - Małgorzata Maciążek-Jurczyk
- Department of Physical Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 40-055 Katowice, Poland; (A.O.); (K.K.); (W.R.)
| |
Collapse
|
4
|
He L, Bhat K, Ioannidis A, Pajonk F. Effects of dopamine receptor antagonists and radiation on mouse neural stem/progenitor cells. Radiother Oncol 2024; 201:110562. [PMID: 39341503 PMCID: PMC11987595 DOI: 10.1016/j.radonc.2024.110562] [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: 02/21/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Dopamine receptor antagonists have recently been identified as potential anti-cancer agents in combination with radiation, and a first drug of this class is in clinical trials against pediatric glioma. Radiotherapy causes cognitive impairment primarily by eliminating neural stem/progenitor cells and subsequent loss of neurogenesis, along with inducing inflammation, vascular damage, and synaptic alterations. Here, we tested the combined effects of dopamine receptor antagonists and radiation on neural stem/progenitor cells. METHODS Using transgenic mice that report the presence of neural stem/progenitor cells through Nestin promoter-driven expression of EGFP, the effects of dopamine receptor antagonists alone or in combination with radiation on neural stem/progenitor cells were assessed in sphere-formation assays, extreme limiting dilution assays, flow cytometry and real-time PCR in vitro and in vivo in both sexes. RESULTS We report that hydroxyzine and trifluoperazine exhibited sex-dependent effects on murine newborn neural stem/progenitor cells in vitro. In contrast, amisulpride, nemonapride, and quetiapine, when combined with radiation, significantly increased the number of neural stem/progenitor cells in both sexes. In vivo, trifluoperazine showed sex-dependent effects on adult neural stem/progenitor cells, while amisulpride demonstrated significant effects in both sexes. Further, amisulpride increased sphere forming capacity and stem cell frequency in both sexes when compared to controls. CONCLUSION We conclude that a therapeutic window for dopamine receptor antagonists in combination with radiation potentially exists, making it a novel combination therapy against glioblastoma. Normal tissue toxicity following this treatment scheme likely differs depending on age and sex and should be taken into consideration when designing clinical trials.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States.
| | - Kruttika Bhat
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, United States; Jonsson Comprehensive Cancer Center at UCLA, United States; Department of Neurosurgery, David Geffen School of Medicine at UCLA, United States
| |
Collapse
|
5
|
Yuan D, Chen W, Jin S, Li W, Liu W, Liu L, Wu Y, Zhang Y, He X, Jiang J, Sun H, Liu X, Liu J. Co-expression of immune checkpoints in glioblastoma revealed by single-nucleus RNA sequencing and spatial transcriptomics. Comput Struct Biotechnol J 2024; 23:1534-1546. [PMID: 38633388 PMCID: PMC11021796 DOI: 10.1016/j.csbj.2024.04.014] [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: 12/05/2023] [Revised: 04/06/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
Glioblastoma (GBM) is one of the most malignant tumors of the central nervous system. The pattern of immune checkpoint expression in GBM remains largely unknown. We performed snRNA-Seq and spatial transcriptomic (ST) analyses on untreated GBM samples. 8 major cell types were found in both tumor and adjacent normal tissues, with variations in infiltration grade. Neoplastic cells_6 was identified in malignant cells with high expression of invasion and proliferator-related genes, and analyzed its interactions with microglia, MDM cells and T cells. Significant alterations in ligand-receptor interactions were observed, particularly between Neoplastic cells_6 and microglia, and found prominent expression of VISTA/VSIG3, suggesting a potential mechanism for evading immune system attacks. High expression of TIM-3, VISTA, PSGL-1 and VSIG-3 with similar expression patterns in GBM, may have potential as therapeutic targets. The prognostic value of VISTA expression was cross-validated in 180 glioma patients, and it was observed that patients with high VISTA expression had a poorer prognosis. In addition, multimodal cross analysis integrated SnRNA-seq and ST, revealing complex intracellular communication and mapping the GBM tumor microenvironment. This study reveals novel molecular characteristics of GBM, co-expression of immune checkpoints, and potential therapeutic targets, contributing to improving the understanding and treatment of GBM.
Collapse
Affiliation(s)
- Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Wenting Chen
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Wei Li
- Department of Neurosurgery, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Wanmei Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Yinhao Wu
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Yuxin Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Xiaoyu He
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Xiangyu Liu
- Department of Neurosurgery, the Affiliated Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Rodemer I, Vetter C, Kalder M, Hajek A, Kostev K. Negative Association Between Schizophrenia and Subsequent Cancer Diagnoses-A Retrospective Cohort Study from Germany. Eur J Investig Health Psychol Educ 2024; 14:2957-2965. [PMID: 39727502 DOI: 10.3390/ejihpe14120194] [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: 09/18/2024] [Revised: 11/13/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Since previous studies have reported contradictory findings regarding the relationship between schizophrenia and cancer, we evaluated the association between schizophrenia and cancer diagnoses. METHODS In this retrospective cohort study, the IQVIA Disease Analyzer database was utilized to examine the incidence of cancer among patients aged over 18 years diagnosed with schizophrenia in German general practices from 2005 to 2022. Patients with schizophrenia were compared with those without the condition, with adjustments made for age, sex, index year of diagnosis, average annual practitioners visit frequency, and comorbidity. Kaplan-Meier curves were used to analyze the 10-year cumulative incidence of schizophrenia and cancer in total amongst patients with and without schizophrenia. Univariate Cox regression analysis was performed to calculate Hazard Ratios (HR) of cancer risk and their 95% confidence intervals (CI) of cancer in total and of specific cancer types. RESULTS Patients with schizophrenia (N = 13.711) had a lower incidence of cancer diagnosis compared to those without (N = 68.555). Specifically, 10.4% of patients with schizophrenia and 12.5% of patients without the condition were diagnosed with cancer (p < 0.001). Cox regression analysis showed a significant association between schizophrenia and subsequent cancer in the total population (HR: 0.82; 95% CI: 0.76-0.90), and among men (HR: 0.70; 95% CI: 0.61-0.80), but not among women (HR: 0.94, 95% CI: 0.84-1.04). Analyses stratified by cancer type and sex revealed a strong and significant association between schizophrenia and a decreased risk of prostate cancer in men (HR: 0.38; 95% CI: 0.24-0.61). Furthermore, there was also a negative association between schizophrenia and colorectal cancer risk in men, but statistical significance was not reached (HR: 0.58; 95% CI: 0.37-0.93). CONCLUSIONS This study demonstrates negative associations between schizophrenia and subsequent cancer, and more specifically in men for prostate and colorectal cancer. However, further research is required to explore the underlying reasons for these associations.
Collapse
Affiliation(s)
- Ira Rodemer
- Epidemiology, IQVIA, 60549 Frankfurt, Germany
| | | | - Matthias Kalder
- Department of Gynecology and Obstetrics, Philipps-University of Marburg, 35043 Marburg, Germany
| | - André Hajek
- Department of Health Economics and Health Services Research, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Karel Kostev
- Epidemiology, IQVIA, 60549 Frankfurt, Germany
- Department of Gynecology and Obstetrics, Philipps-University of Marburg, 35043 Marburg, Germany
| |
Collapse
|
7
|
Zhong Y, Geng F, Mazik L, Yin X, Becker AP, Mohammed S, Su H, Xing E, Kou Y, Chiang CY, Fan Y, Guo Y, Wang Q, Li PK, Mo X, Lefai E, He L, Cheng X, Zhang X, Chakravarti A, Guo D. Combinatorial targeting of glutamine metabolism and lysosomal-based lipid metabolism effectively suppresses glioblastoma. Cell Rep Med 2024; 5:101706. [PMID: 39236712 PMCID: PMC11524980 DOI: 10.1016/j.xcrm.2024.101706] [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: 12/08/2023] [Revised: 06/07/2024] [Accepted: 08/08/2024] [Indexed: 09/07/2024]
Abstract
Antipsychotic drugs have been shown to have antitumor effects but have had limited potency in the clinic. Here, we unveil that pimozide inhibits lysosome hydrolytic function to suppress fatty acid and cholesterol release in glioblastoma (GBM), the most lethal brain tumor. Unexpectedly, GBM develops resistance to pimozide by boosting glutamine consumption and lipogenesis. These elevations are driven by SREBP-1, which we find upregulates the expression of ASCT2, a key glutamine transporter. Glutamine, in turn, intensifies SREBP-1 activation through the release of ammonia, creating a feedforward loop that amplifies both glutamine metabolism and lipid synthesis, leading to drug resistance. Disrupting this loop via pharmacological targeting of ASCT2 or glutaminase, in combination with pimozide, induces remarkable mitochondrial damage and oxidative stress, leading to GBM cell death in vitro and in vivo. Our findings underscore the promising therapeutic potential of effectively targeting GBM by combining glutamine metabolism inhibition with lysosome suppression.
Collapse
Affiliation(s)
- Yaogang Zhong
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Feng Geng
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Logan Mazik
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Xinmin Yin
- Department of Chemistry, Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Aline Paixao Becker
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Shabber Mohammed
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy at The Ohio State University, Columbus, OH 43210, USA
| | - Huali Su
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Enming Xing
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy at The Ohio State University, Columbus, OH 43210, USA
| | - Yongjun Kou
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Cheng-Yao Chiang
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yunzhou Fan
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Yongchen Guo
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Qiang Wang
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Pui-Kai Li
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy at The Ohio State University, Columbus, OH 43210, USA
| | - Xiaokui Mo
- Center for Biostatistics, Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Etienne Lefai
- Human Nutrition Unit, French National Research Institute for Agriculture, Food and Environment, University Clermont Auvergne, 63122 Clermont-Ferrand, France
| | - Liqing He
- Department of Chemistry, Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Xiaolin Cheng
- Division of Medicinal Chemistry & Pharmacognosy, College of Pharmacy at The Ohio State University, Columbus, OH 43210, USA; Translational Data Analytics Institute at The Ohio State University, Columbus, OH 43210, USA
| | - Xiang Zhang
- Department of Chemistry, Center for Regulatory and Environmental Analytical Metabolomics, University of Louisville, Louisville, KY 40208, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital and Richard J. Solove Research Institute, and College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center for Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| |
Collapse
|
8
|
Tausk F. Finally, successful interventions to ameliorate cutaneous infestations. J DERMATOL TREAT 2024; 35:2326655. [PMID: 38465790 DOI: 10.1080/09546634.2024.2326655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Affiliation(s)
- Francisco Tausk
- Department of Dermatology, Allergy, Immunology and Rheumatology University of Rochester, Rochester, NY, USA
| |
Collapse
|
9
|
Martula E, Morak-Młodawska B, Jeleń M, Okechukwu PN, Balachandran A, Tehirunavukarasu P, Anamalay K, Ulaganathan V. Synthesis and Structural Characterization of Novel Dimers of Dipyridothiazine as Promising Antiproliferative Agents. Molecules 2023; 28:7662. [PMID: 38005384 PMCID: PMC10674446 DOI: 10.3390/molecules28227662] [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: 11/02/2023] [Revised: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Many new isomeric dipyridothiazine dimers have been presented as molecules with anticancer potential. These compounds were obtained in efficient syntheses of 1,6-, 1,8-, 2,7- and 3,6-diazaphenothiazines with selected alkylaromatic linkers. The structures of these compounds has been proven with two-dimensional spectroscopic techniques (COSY, NOESY, HSQC and HMBC) and high-resolution mass spectrometry (HRMS). In silico analyses of probable molecular targets were performed using the Way2Drug server. All new dimers were tested for anticancer activity against breast cancer line MCF7 and colon cancer line SW480. Cytotoxicity was assessed on normal L6 muscle cells. The tested dimers had high anticancer potential expressed as IC50 and the selectivity index SI. The most active derivative, 4c, showed an IC50 activity of less than 1 µM and an SI selectivity index higher than 100. Moreover, the compounds were characterized by low toxicity towards normal cells, simultaneously indicating a high cytostatic potential.
Collapse
Affiliation(s)
- Emilia Martula
- Doctoral School of The Medical University of Silesia, 40-055 Katowice, Poland;
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland;
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland;
| | - Patrick N. Okechukwu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Abbirami Balachandran
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Prethika Tehirunavukarasu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Kirthani Anamalay
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia; (P.N.O.); (A.B.); (P.T.); (K.A.)
| | - Vaidehi Ulaganathan
- Department of Food Science and Nutrition, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
10
|
Vanneste M, Venzke A, Guin S, Fuller AJ, Jezewski AJ, Beattie SR, Krysan DJ, Meyers MJ, Henry MD. The anti-cancer efficacy of a novel phenothiazine derivative is independent of dopamine and serotonin receptor inhibition. Front Oncol 2023; 13:1295185. [PMID: 37909019 PMCID: PMC10613967 DOI: 10.3389/fonc.2023.1295185] [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: 09/15/2023] [Accepted: 10/02/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction An attractive, yet unrealized, goal in cancer therapy is repurposing psychiatric drugs that can readily penetrate the blood-brain barrier for the treatment of primary brain tumors and brain metastases. Phenothiazines (PTZs) have demonstrated anti-cancer properties through a variety of mechanisms. However, it remains unclear whether these effects are entirely separate from their activity as dopamine and serotonin receptor (DR/5-HTR) antagonists. Methods In this study, we evaluated the anti-cancer efficacy of a novel PTZ analog, CWHM-974, that was shown to be 100-1000-fold less potent against DR/5-HTR than its analog fluphenazine (FLU). Results CWHM-974 was more potent than FLU against a panel of cancer cell lines, thus clearly demonstrating that its anti-cancer effects were independent of DR/5-HTR signaling. Our results further suggested that calmodulin (CaM) binding may be necessary, but not sufficient, to explain the anti-cancer effects of CWHM-974. While both FLU and CWHM-974 induced apoptosis, they induced distinct effects on the cell cycle (G0/G1 and mitotic arrest respectively) suggesting that they may have differential effects on CaM-binding proteins involved in cell cycle regulation. Discussion Altogether, our findings indicated that the anti-cancer efficacy of the CWHM-974 is separable from DR/5-HTR antagonism. Thus, reducing the toxicity associated with phenothiazines related to DR/5-HTR antagonism may improve the potential to repurpose this class of drugs to treat brain tumors and/or brain metastasis.
Collapse
Affiliation(s)
- Marion Vanneste
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Anita Venzke
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Soumitra Guin
- Department of Chemistry, Saint Louis University, Saint Louis, MO, United States
| | - Andrew J. Fuller
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Andrew J. Jezewski
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Sarah R. Beattie
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Damian J. Krysan
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Marvin J. Meyers
- Department of Chemistry, Saint Louis University, Saint Louis, MO, United States
| | - Michael D. Henry
- Department of Molecular Physiology and Biophysics, Carver College of Medicine, University of Iowa, Iowa City IA, United States
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
11
|
Liu H, Huang Q, Fan Y, Li B, Liu X, Hu C. Dissecting the novel abilities of aripiprazole: The generation of anti-colorectal cancer effects by targeting G αq via HTR2B. Acta Pharm Sin B 2023; 13:3400-3413. [PMID: 37655314 PMCID: PMC10465950 DOI: 10.1016/j.apsb.2023.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 09/02/2023] Open
Abstract
Colorectal cancer (CRC) is a type of malignant tumor that seriously threatens human health and life, and its treatment has always been a difficulty and hotspot in research. Herein, this study for the first time reports that antipsychotic aripiprazole (Ari) against the proliferation of CRC cells both in vitro and in vivo, but with less damage in normal colon cells. Mechanistically, the results showed that 5-hydroxytryptamine 2B receptor (HTR2B) and its coupling protein G protein subunit alpha q (Gαq) were highly distributed in CRC cells. Ari had a strong affinity with HTR2B and inhibited HTR2B downstream signaling. Blockade of HTR2B signaling suppressed the growth of CRC cells, but HTR2B was not found to have independent anticancer activity. Interestingly, the binding of Gαq to HTR2B was decreased after Ari treatment. Knockdown of Gαq not only restricted CRC cell growth, but also directly affected the anti-CRC efficacy of Ari. Moreover, an interaction between Ari and Gαq was found in that the mutation at amino acid 190 of Gαq reduced the efficacy of Ari. Thus, these results confirm that Gαq coupled to HTR2B was a potential target of Ari in mediating CRC proliferation. Collectively, this study provides a novel effective strategy for CRC therapy and favorable evidence for promoting Ari as an anticancer agent.
Collapse
Affiliation(s)
- Haowei Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Qiuming Huang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yunqi Fan
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Bo Li
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xuemei Liu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic Substances, Chongqing Institute for Food and Drug Control, Chongqing 401121, China
| | - Changhua Hu
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic Substances, Chongqing Institute for Food and Drug Control, Chongqing 401121, China
| |
Collapse
|
12
|
Błaszczyk M, Kozioł A, Palko-Łabuz A, Środa-Pomianek K, Wesołowska O. Modulators of cellular cholesterol homeostasis as antiproliferative and model membranes perturbing agents. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2023; 1865:184163. [PMID: 37172710 DOI: 10.1016/j.bbamem.2023.184163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 04/14/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023]
Abstract
Cholesterol is an important component of mammalian cell membranes affecting their fluidity and permeability. Together with sphingomyelin, cholesterol forms microdomains, called lipid rafts. They play important role in signal transduction forming platforms for interaction of signal proteins. Altered levels of cholesterol are known to be strongly associated with the development of various pathologies (e.g., cancer, atherosclerosis and cardiovascular diseases). In the present work, the group of compounds that share the property of affecting cellular homeostasis of cholesterol was studied. It contained antipsychotic and antidepressant drugs, as well as the inhibitors of cholesterol biosynthesis, simvastatin, betulin, and its derivatives. All compounds were demonstrated to be cytotoxic to colon cancer cells but not to non-cancerous cells. Moreover, the most active compounds decreased the level of free cellular cholesterol. The interaction of drugs with raft-mimicking model membranes was visualized. All compounds reduced the size of lipid domains, however, only some affected their number and shape. Membrane interactions of betulin and its novel derivatives were characterized in detail. Molecular modeling indicated that high dipole moment and significant lipophilicity were characteristic for the most potent antiproliferative agents. The importance of membrane interactions of cholesterol homeostasis-affecting compounds, especially betulin derivatives, for their anticancer potency was suggested.
Collapse
Affiliation(s)
- Maria Błaszczyk
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chalubinskiego 3a, 50-368 Wroclaw, Poland.
| | - Agata Kozioł
- Department of Biochemistry and Immunochemistry, Wroclaw Medical University, Sklodowskiej-Curie 48/50, 50-369 Wroclaw, Poland.
| | - Anna Palko-Łabuz
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chalubinskiego 3a, 50-368 Wroclaw, Poland.
| | - Kamila Środa-Pomianek
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chalubinskiego 3a, 50-368 Wroclaw, Poland.
| | - Olga Wesołowska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, ul. Chalubinskiego 3a, 50-368 Wroclaw, Poland.
| |
Collapse
|
13
|
Moura C, Vale N. The Role of Dopamine in Repurposing Drugs for Oncology. Biomedicines 2023; 11:1917. [PMID: 37509555 PMCID: PMC10377204 DOI: 10.3390/biomedicines11071917] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/04/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Dopamine is a neurotransmitter that plays an important role within the brain by regulating a wide variety of cognitive and emotional processes. In cancer, its role is distinct and uncertain, but it is characterized by the interaction with its receptors that may be in the tumor cells; we have examples of different types of cancer with this characteristic, of which breast and colon cancer stand out. It is believed that dopamine and some of its receptors also influence other cellular processes such as cell proliferation, survival, migration, and invasion. The potential of these receptors has allowed the exploration of existing drugs, originally developed for non-oncological purposes, for the possible treatment of cancer. However, regarding the repurposing of drugs for cancer treatment, the role of dopamine is not so straightforward and needs to be clarified. For this reason, this review intends to present concepts associated with twelve drugs reused for oncology based on dopamine and its receptors. Some of them can behave as antagonists and inhibit tumor cell growth leading to cell death. Attention to this group of drugs may enhance the study of other pharmacological conditions such as signaling pathways related to cell proliferation and migration. Modulation of these pathways using drugs originally developed for other conditions may offer potential therapeutic opportunities in oncology. It is important to note that while the repurposing of oncology drugs based on dopamine signaling is promising, further studies are still needed to fully understand the mechanisms involved and determine the clinical efficacy and safety of these approaches.
Collapse
Affiliation(s)
- Catarina Moura
- OncoPharma 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
- OncoPharma 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, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
14
|
Rončević A, Koruga N, Soldo Koruga A, Rončević R, Rotim T, Šimundić T, Kretić D, Perić M, Turk T, Štimac D. Personalized Treatment of Glioblastoma: Current State and Future Perspective. Biomedicines 2023; 11:1579. [PMID: 37371674 DOI: 10.3390/biomedicines11061579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive glial tumor of the central nervous system. Despite intense scientific efforts, patients diagnosed with GBM and treated with the current standard of care have a median survival of only 15 months. Patients are initially treated by a neurosurgeon with the goal of maximal safe resection of the tumor. Obtaining tissue samples during surgery is indispensable for the diagnosis of GBM. Technological improvements, such as navigation systems and intraoperative monitoring, significantly advanced the possibility of safe gross tumor resection. Usually within six weeks after the surgery, concomitant radiotherapy and chemotherapy with temozolomide are initiated. However, current radiotherapy regimens are based on population-level studies and could also be improved. Implementing artificial intelligence in radiotherapy planning might be used to individualize treatment plans. Furthermore, detailed genetic and molecular markers of the tumor could provide patient-tailored immunochemotherapy. In this article, we review current standard of care and possibilities of personalizing these treatments. Additionally, we discuss novel individualized therapeutic options with encouraging results. Due to inherent heterogeneity of GBM, applying patient-tailored treatment could significantly prolong survival of these patients.
Collapse
Affiliation(s)
- Alen Rončević
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Nenad Koruga
- Department of Neurosurgery, University Hospital Center Osijek, 31000 Osijek, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | - Anamarija Soldo Koruga
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Neurology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Robert Rončević
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tatjana Rotim
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tihana Šimundić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Nephrology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Domagoj Kretić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Marija Perić
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Cytology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Tajana Turk
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Diagnostic and Interventional Radiology, University Hospital Center Osijek, 31000 Osijek, Croatia
| | - Damir Štimac
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Department of Radiology, National Memorial Hospital Vukovar, 32000 Vukovar, Croatia
| |
Collapse
|
15
|
Yoon S, Kim HS. First-Line Combination Treatment with Low-Dose Bipolar Drugs for ABCB1-Overexpressing Drug-Resistant Cancer Populations. Int J Mol Sci 2023; 24:ijms24098389. [PMID: 37176096 PMCID: PMC10179254 DOI: 10.3390/ijms24098389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/25/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
Tumors include a heterogeneous population, of which a small proportion includes drug-resistant cancer (stem) cells. In drug-sensitive cancer populations, first-line chemotherapy reduces tumor volume via apoptosis. However, it stimulates drug-resistant cancer populations and finally results in tumor recurrence. Recurrent tumors are unresponsive to chemotherapeutic drugs and are primarily drug-resistant cancers. Therefore, increased apoptosis in drug-resistant cancer cells in heterogeneous populations is important in first-line chemotherapeutic treatments. The overexpression of ABCB1 (or P-gp) on cell membranes is an important characteristic of drug-resistant cancer cells; therefore, first-line combination treatments with P-gp inhibitors could delay tumor recurrence. Low doses of bipolar drugs showed P-gp inhibitory activity, and their use as a combined therapy sensitized drug-resistant cancer cells. FDA-approved bipolar drugs have been used in clinics for a long period of time, and their toxicities are well reported. They can be easily applied as first-line combination treatments for targeting resistant cancer populations. To apply bipolar drugs faster in first-line combination treatments, knowledge of their complete information is crucial. This review discusses the use of low-dose bipolar drugs in sensitizing ABCB1-overexpressing, drug-resistant cancers. We believe that this review will contribute to facilitating first-line combination treatments with low-dose bipolar drugs for targeting drug-resistant cancer populations. In addition, our findings may aid further investigations into targeting drug-resistant cancer populations with low-dose bipolar drugs.
Collapse
Affiliation(s)
- Sungpil Yoon
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon 16419, Republic of Korea
| |
Collapse
|
16
|
Servidei T, Sgambato A, Lucchetti D, Navarra P, Ruggiero A. Drug Repurposing in Pediatric Brain Tumors: Posterior Fossa Ependymoma and Diffuse Midline Glioma under the Looking Glass. FRONT BIOSCI-LANDMRK 2023; 28:77. [PMID: 37114548 DOI: 10.31083/j.fbl2804077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/29/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023]
Abstract
Tumors of the Central Nervous System (CNS) represent the leading cause of cancer-related deaths in children. Current treatment options are not curative for most malignant histologies, and intense preclinical and clinical research is needed to develop more effective therapeutic interventions against these tumors, most of which meet the FDA definition for orphan diseases. Increased attention is being paid to the repositioning of already-approved drugs for new anticancer indications as a fast-tracking strategy for identifying new and more effective therapies. Two pediatric CNS tumors, posterior fossa ependymoma (EPN-PF) type A and diffuse midline glioma (DMG) H3K27-altered, share loss of H3K27 trimethylation as a common epigenetic hallmark and display early onset and poor prognosis. These features suggest a potentially common druggable vulnerability. Successful treatment of these CNS tumors raises several challenges due to the location of tumors, chemoresistance, drug blood-brain barrier penetration, and the likelihood of adverse side effects. Recently, increasing evidence demonstrates intense interactions between tumor cell subpopulations and supportive tumor microenvironments (TMEs) including nerve, metabolic, and inflammatory TMEs. These findings suggest the use of drugs, and/or multi-drug combinations, that attack both tumor cells and the TME simultaneously. In this work, we present an overview of the existing evidence concerning the most preclinically validated noncancer drugs with antineoplastic activity. These drugs belong to four pharmacotherapeutic classes: antiparasitic, neuroactive, metabolic, and anti-inflammatory. Preclinical evidence and undergoing clinical trials in patients with brain tumors, with special emphasis on pediatric EPN-PF and DMG, are summarized and critically discussed.
Collapse
Affiliation(s)
- Tiziana Servidei
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandro Sgambato
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Donatella Lucchetti
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore -- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
17
|
Jeleń M, Pluta K, Szmielew M, Morak-Młodawska B, Herman K, Giercuszkiewicz K, Kasprzycka A, Skonieczna M. 14-Substituted Diquinothiazines as a New Group of Anticancer Agents. Molecules 2023; 28:molecules28073248. [PMID: 37050010 PMCID: PMC10096123 DOI: 10.3390/molecules28073248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023] Open
Abstract
A series of novel double-angularly condensed diquinothiazines with aminoalkyl, amidoalkyl, sulfonamidoalkyl, and substituted phenyl groups was designed, synthesized, and evaluated for their anticancer activity against four selected human tumor cell lines (HTC116, SH-SY5Y, A549, and H1299). The cytotoxicity of the novel diquinothiazines was investigated against BEAS-2B cells. The activities of the compounds were compared to etoposide. Among them, compounds with aminoalkyl and phenyl groups showed excellent broad-spectrum anticancer activity. The most active 14-(methylthiophenyl)diquinothiazine, 3c, showed low cytotoxicity against BEAS-2B cells and high activity against tumor cell lines HTC116, SH-SY5Y, A549, and H1299, with IC50 values of 2.3 µM, 2.7 µM, 17.2 µM, and 2.7 µM, respectively (etopiside 8.6 µM, 3.9 µM, 44.8 µM, and 0.6, respectively). Live long-term microscopic observations of cell survival using the starting molecule M0 were also performed. Flow cytometry showed the proapoptotic effects of the studied diquinothiazines. Inhibition of the cell cycle in the S phase was observed, which is associated with damage to nucleic acids and connected to DNA replication arrest.
Collapse
Affiliation(s)
- Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Krystian Pluta
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Małgorzata Szmielew
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Kinga Herman
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego Street 4, 44-100 Gliwice, Poland
| | - Klaudia Giercuszkiewicz
- Department of Systems Biology and Engineering, The Silesian University of Technology, Akademicka Street 16, 44–100 Gliwice, Poland
| | - Anna Kasprzycka
- Department of Organic Chemistry, Bioorganic Chemistry and Biotechnology, Faculty of Chemistry, Silesian University of Technology, Krzywoustego Street 4, 44-100 Gliwice, Poland
- Centre of Biotechnology, Silesian University of Technology, Krzywoustego Street 8, 44-100 Gliwice, Poland
| | - Magdalena Skonieczna
- Department of Systems Biology and Engineering, The Silesian University of Technology, Akademicka Street 16, 44–100 Gliwice, Poland
- Centre of Biotechnology, Silesian University of Technology, Krzywoustego Street 8, 44-100 Gliwice, Poland
| |
Collapse
|
18
|
de Bartolomeis A, Ciccarelli M, De Simone G, Mazza B, Barone A, Vellucci L. Canonical and Non-Canonical Antipsychotics' Dopamine-Related Mechanisms of Present and Next Generation Molecules: A Systematic Review on Translational Highlights for Treatment Response and Treatment-Resistant Schizophrenia. Int J Mol Sci 2023; 24:ijms24065945. [PMID: 36983018 PMCID: PMC10051989 DOI: 10.3390/ijms24065945] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Schizophrenia is a severe psychiatric illness affecting almost 25 million people worldwide and is conceptualized as a disorder of synaptic plasticity and brain connectivity. Antipsychotics are the primary pharmacological treatment after more than sixty years after their introduction in therapy. Two findings hold true for all presently available antipsychotics. First, all antipsychotics occupy the dopamine D2 receptor (D2R) as an antagonist or partial agonist, even if with different affinity; second, D2R occupancy is the necessary and probably the sufficient mechanism for antipsychotic effect despite the complexity of antipsychotics' receptor profile. D2R occupancy is followed by coincident or divergent intracellular mechanisms, implying the contribution of cAMP regulation, β-arrestin recruitment, and phospholipase A activation, to quote some of the mechanisms considered canonical. However, in recent years, novel mechanisms related to dopamine function beyond or together with D2R occupancy have emerged. Among these potentially non-canonical mechanisms, the role of Na2+ channels at the dopamine at the presynaptic site, dopamine transporter (DAT) involvement as the main regulator of dopamine concentration at synaptic clefts, and the putative role of antipsychotics as chaperones for intracellular D2R sequestration, should be included. These mechanisms expand the fundamental role of dopamine in schizophrenia therapy and may have relevance to considering putatively new strategies for treatment-resistant schizophrenia (TRS), an extremely severe condition epidemiologically relevant and affecting almost 30% of schizophrenia patients. Here, we performed a critical evaluation of the role of antipsychotics in synaptic plasticity, focusing on their canonical and non-canonical mechanisms of action relevant to the treatment of schizophrenia and their subsequent implication for the pathophysiology and potential therapy of TRS.
Collapse
Affiliation(s)
- Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences and Dentistry, University Medical School of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
19
|
Repurposing Antidepressants and Phenothiazine Antipsychotics as Efflux Pump Inhibitors in Cancer and Infectious Diseases. Antibiotics (Basel) 2023; 12:antibiotics12010137. [PMID: 36671340 PMCID: PMC9855052 DOI: 10.3390/antibiotics12010137] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/29/2022] [Accepted: 01/05/2023] [Indexed: 01/13/2023] Open
Abstract
Multidrug resistance (MDR) is a major obstacle in the therapy of infectious diseases and cancer. One of the major mechanisms of MDR is the overexpression of efflux pumps (EPs) that are responsible for extruding antimicrobial and anticancer agents. EPs have additional roles of detoxification that may aid the development of bacterial infection and the progression of cancer. Therefore, targeting EPs may be an attractive strategy to treat bacterial infections and cancer. The development and discovery of a new drug require a long timeline and may come with high development costs. A potential alternative to reduce the time and costs of drug development is to repurpose already existing drugs. Antidepressants and antipsychotic agents are widely used in clinical practice in the treatment of psychiatric disorders and some somatic diseases. Antidepressants and antipsychotics have demonstrated various beneficial activities that may be utilized in the treatment of infections and cancer. This review aims to provide a brief overview of antibacterial and anticancer effects of selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs) and phenothiazine antipsychotics, while focusing on EPs. However, it should be noted that the antimicrobial activity of a traditionally non-antibiotic drug may have clinical implications regarding dysbiosis and bacterial MDR.
Collapse
|
20
|
Tri- and Pentacyclic Azaphenothiazine as Pro-Apoptotic Agents in Lung Carcinoma with a Protective Potential to Healthy Cell Lines. Molecules 2022; 27:molecules27165255. [PMID: 36014495 PMCID: PMC9413739 DOI: 10.3390/molecules27165255] [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: 06/16/2022] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 12/24/2022] Open
Abstract
The phenothiazine derivatives, tricyclic 10H-3,6-diazaphenothiazine (DPT-1) and pentacyclic 7-(3′-dimethylaminopropyl)diquinothiazine (DPT-2), have recently been shown to exhibit promising anticancer activities in vitro. In this report, we demonstrated that DPT-1 and DPT-2 could be pro-apoptotic agents in lung carcinoma, the human lung carcinoma A549 and non-small lung carcinoma H1299, in the range of IC50 = 1.52–12.89 µM, with a protective potential to healthy cell lines BEAS-2B and NHDF. The compounds showed higher activity in the range of the tested concentrations and low cytotoxicity in relation to normal healthy cells than doxorubicin, used as the reference drug. The cytostatic potential of DPT-1 and DPT-2 was demonstrated with the use of MTT assay. Cell cycle analysis via flow cytometry using Annexin-V assay showed the pro-apoptotic and pro-necrotic role of the studied diazaphenothiazines in the cell cycle. DPT-1 and DPT-2 initiated a biological response in the investigated cancer models with a different mechanism and at a different rate. Based on these findings, it can be concluded that DPT-1 and DPT-2 have potential as chemotherapeutic agents.
Collapse
|
21
|
Brown JS. Treatment of cancer with antipsychotic medications: Pushing the boundaries of schizophrenia and cancer. Neurosci Biobehav Rev 2022; 141:104809. [PMID: 35970416 DOI: 10.1016/j.neubiorev.2022.104809] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 10/15/2022]
Abstract
Over a century ago, the phenothiazine dye, methylene blue, was discovered to have both antipsychotic and anti-cancer effects. In the 20th-century, the first phenothiazine antipsychotic, chlorpromazine, was found to inhibit cancer. During the years of elucidating the pharmacology of the phenothiazines, reserpine, an antipsychotic with a long historical background, was likewise discovered to have anti-cancer properties. Research on the effects of antipsychotics on cancer continued slowly until the 21st century when efforts to repurpose antipsychotics for cancer treatment accelerated. This review examines the history of these developments, and identifies which antipsychotics might treat cancer, and which cancers might be treated by antipsychotics. The review also describes the molecular mechanisms through which antipsychotics may inhibit cancer. Although the overlap of molecular pathways between schizophrenia and cancer have been known or suspected for many years, no comprehensive review of the subject has appeared in the psychiatric literature to assess the significance of these similarities. This review fills that gap and discusses what, if any, significance the similarities have regarding the etiology of schizophrenia.
Collapse
|
22
|
Siddiqui S, Deshmukh AJ, Mudaliar P, Nalawade AJ, Iyer D, Aich J. Drug repurposing: re-inventing therapies for cancer without re-entering the development pipeline—a review. J Egypt Natl Canc Inst 2022; 34:33. [PMID: 35934727 PMCID: PMC9358112 DOI: 10.1186/s43046-022-00137-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/10/2022] [Indexed: 11/25/2022] Open
Abstract
While majority of the current treatment approaches for cancer remain expensive and are associated with several side effects, development of new treatment modalities takes a significant period of research, time, and expenditure. An alternative novel approach is drug repurposing that focuses on finding new applications for the previously clinically approved drugs. The process of drug repurposing has also been facilitated by current advances in the field of proteomics, genomics, and information computational biology. This approach not only provides cheaper, effective, and potentially safer drugs with less side effects but also increases the processing pace of drug development. In this review, we wish to highlight some recent developments in the area of drug repurposing in cancer with a specific focus on the repurposing potential of anti-psychotic, anti-inflammatory and anti-viral drugs, anti-diabetic, antibacterial, and anti-fungal drugs.
Collapse
|
23
|
Tibor Fekete J, Győrffy B. A unified platform enabling biomarker ranking and validation for 1,562 drugs using transcriptomic data of 1,250 cancer cell lines. Comput Struct Biotechnol J 2022; 20:2885-2894. [PMID: 35765648 PMCID: PMC9198269 DOI: 10.1016/j.csbj.2022.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/01/2022] [Accepted: 06/01/2022] [Indexed: 12/21/2022] Open
Abstract
Intro In vitro cell line models provide a valuable resource to investigate compounds useful in the systemic chemotherapy of cancer. However, the due to the dispersal of the data into several different databases, the utilization of these resources is limited. Here, our aim was to establish a platform enabling the validation of chemoresistance-associated genes and the ranking of available cell line models. Methods We processed four independent databases, DepMap, GDSC1, GDSC2, and CTRP. The gene expression data was quantile normalized and HUGO gene names were assigned to have unambiguous identification of the genes. Resistance values were exported for all agents. The correlation between gene expression and therapy resistance is computed using ROC test. Results We combined four datasets with chemosensitivity data of 1562 agents and transcriptome-level gene expression of 1250 cancer cell lines. We have set up an online tool utilizing this database to correlate available cell line sensitivity data and treatment response in a uniform analysis pipeline (www.rocplot.com/cells). We employed the established pipeline to by rank genes related to resistance against afatinib and lapatinib, two inhibitors of the tyrosine-kinase domain of ERBB2. Discussion The computational tool is useful 1) to correlate gene expression with resistance, 2) to identify and rank resistant and sensitive cell lines, and 3) to rank resistance associated genes, cancer hallmarks, and gene ontology pathways. The platform will be an invaluable support to speed up cancer research by validating gene-resistance correlations and by selecting the best cell line models for new experiments.
Collapse
Affiliation(s)
- János Tibor Fekete
- Semmelweis University, Department of Bioinformatics and 2 Department of Pediatrics, Budapest H-1094, Hungary
- Research Center for Natural Sciences, Institute of Enzymology, Momentum Cancer Biomarker Research Group, Magyar tudósok körútja 2., Budapest H-1117, Hungary
| | - Balázs Győrffy
- Semmelweis University, Department of Bioinformatics and 2 Department of Pediatrics, Budapest H-1094, Hungary
- Research Center for Natural Sciences, Institute of Enzymology, Momentum Cancer Biomarker Research Group, Magyar tudósok körútja 2., Budapest H-1117, Hungary
| |
Collapse
|
24
|
Osacka J, Kiss A, Pirnik Z. Possible involvement of apoptosis in the antipsychotics side effects: A minireview. Clin Exp Pharmacol Physiol 2022; 49:836-847. [PMID: 35575958 DOI: 10.1111/1440-1681.13671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 01/04/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022]
Abstract
Antipsychotics are used in the treatment of schizophrenia and other psychiatric disorders. Generally they are divided into typical and atypical ones, according to the fact that atypical antipychotics induce less side effects and are more effective in terms of social and cognitive improvements. Their pharmacological effects are mediated via broad range of receptors that consequently influence different cellular signaling pathways. Antipsychotics produce udesirable side effects that range from relatively minor to life-threatening ones. In vitro and in vivo studies have pointed to neurotoxic effect exerted by some antipsychotics and have shown that apoptosis might play role in some side effects induced by antipsyschotics, including tardive dyskinesia, weight gain, agranulocytosis, osteoporosis, myocarditis, etc. Although cumulative data have suggested safety of atypical antipsychotics use during pregnancy some of them have been shown to induce apoptotic neurodegenerative and structural changes in fetal brains with long-lasting impact on cognitive impairment of offsprings. Typical antipsychotics seem to be more cytotoxic than atypical ones. Recently, epidemiological studies have shown lower incidence of cancer in schizophrenic patients what suggest ability of antipsychotics to suppress risk of cancer development. Some antipsychotics have been reported to inhibit cancer cell proliferation and induce their apoptosis. Thus, antipsychotics apoptotic effect may be used as a tool in the treatmnet of some types of cancer, especially in combinatorial therapies. In this minireview, we focused on pro- and anti-apototic or "Dr. Jekyll and Mr. Hyde" effects of antipsychotics, which can be involved in their side effects, as well as their promising therapeutical indications. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jana Osacka
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Alexander Kiss
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia
| | - Zdenko Pirnik
- Biomedical Research Center, Slovak Academy of Sciences, Institute of Experimental Endocrinology, Dubravska cesta 9, Bratislava, Slovakia.,Institute of Physiology, Faculty of Medicine Comenius University in Bratislava, Sasinkova 2, Bratislava, Slovakia.,Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam., 2Prague, Czech Republic
| |
Collapse
|
25
|
Timilsina S, Rajamanickam S, Rao A, Subbarayalu P, Nirzhor S, Abdelfattah N, Viswanadhapalli S, Chen Y, Jatoi I, Brenner A, Rao MK, Vadlamudi R, Kaklamani V. The antidepressant imipramine inhibits breast cancer growth by targeting estrogen receptor signaling and DNA repair events. Cancer Lett 2022; 540:215717. [PMID: 35568265 DOI: 10.1016/j.canlet.2022.215717] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/11/2022] [Accepted: 04/28/2022] [Indexed: 11/29/2022]
Abstract
Aberrant activities of various cell cycle and DNA repair proteins promote cancer growth and progression and render them resistant to therapies. Here, we demonstrate that the anti-depressant imipramine blocks growth of triple-negative (TNBC) and estrogen receptor-positive (ER+) breast cancers by inducing cell cycle arrest and by blocking heightened homologous recombination (HR) and non-homologous end joining-mediated (NHEJ) DNA repair activities. Our results reveal that imipramine inhibits the expression of several cell cycle- and DNA repair-associated proteins including E2F1, CDK1, Cyclin D1, and RAD51. In addition, we show that imipramine inhibits the growth of ER + breast cancers by inhibiting the estrogen receptor- α (ER-α) signaling. Our studies in preclinical mouse models and ex vivo explants from breast cancer patients show that imipramine sensitizes TNBC to the PARP inhibitor olaparib and endocrine resistant ER + breast cancer to anti-estrogens. Our studies suggest that repurposing imipramine could enhance routine care for breast cancer patients. Based on these results, we designed an ongoing clinical trial, where we are testing the efficacy of imipramine for treating patients with triple-negative and estrogen receptor-positive breast cancer. Since aberrant DNA repair activity is used by many cancers to survive and become resistant to therapy, imipramine could be used alone and/or with currently used drugs for treating many aggressive cancers.
Collapse
Affiliation(s)
- Santosh Timilsina
- Department of Cell Systems and Anatomy, UT Health, San Antonio, USA; Greehey Children's Cancer Research Institute, USA
| | - Subapriya Rajamanickam
- Greehey Children's Cancer Research Institute, USA; Department of Molecular Medicine, UT Health, San Antonio, USA
| | - Arhan Rao
- Health Careers High School, San Antonio, TX, USA
| | - Panneerdoss Subbarayalu
- Department of Cell Systems and Anatomy, UT Health, San Antonio, USA; Greehey Children's Cancer Research Institute, USA
| | - Saif Nirzhor
- Department of Cell Systems and Anatomy, UT Health, San Antonio, USA; Greehey Children's Cancer Research Institute, USA
| | | | | | - Yidong Chen
- Greehey Children's Cancer Research Institute, USA; Department of Epidemiology and Statistics, UT Health, San Antonio, USA
| | - Ismail Jatoi
- Department of Surgery, UT Health, San Antonio, USA
| | | | - Manjeet K Rao
- Department of Cell Systems and Anatomy, UT Health, San Antonio, USA; Greehey Children's Cancer Research Institute, USA.
| | - Ratna Vadlamudi
- Department of Obstetrics and Gynecology, UT Health, San Antonio, USA.
| | | |
Collapse
|
26
|
Lin J, Liu B, Zhang Y, Lv L, Cheng D, Zhang W, Shi Y, Jiang X, Tang L, Yuan Y, Zhai H, Shen Q, Xiong Q, Jin Z, Chen Y, Yang C. Gemin6 promotes c-Myc stabilisation and non-small cell lung cancer progression via accelerating AURKB mRNA maturation. Clin Transl Med 2022; 12:e811. [PMID: 35452176 PMCID: PMC9029014 DOI: 10.1002/ctm2.811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 01/03/2023] Open
Affiliation(s)
- Jie Lin
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Baiyang Liu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yong Zhang
- Department of Pathology, Cancer Hospital of China Medical University, Shenyang, Liaoning, China
| | - Li Lv
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | | | - Wenhui Zhang
- The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yulin Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | - Xiulin Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Lin Tang
- Kunming Medical University, Kunming, China
| | | | - Haoqing Zhai
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qiushuo Shen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China
| | | | | | - Yongbin Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Cuiping Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, Kunming Institute of Zoology, Kunming, Yunnan, China.,Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China.,The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| |
Collapse
|
27
|
Lin WZ, Liu YC, Lee MC, Tang CT, Wu GJ, Chang YT, Chu CM, Shiau CY. From GWAS to drug screening: repurposing antipsychotics for glioblastoma. J Transl Med 2022; 20:70. [PMID: 35120529 PMCID: PMC8815269 DOI: 10.1186/s12967-021-03209-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/19/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Glioblastoma is currently an incurable cancer. Genome-wide association studies have demonstrated that 41 genetic variants are associated with glioblastoma and may provide an option for drug development. METHODS We investigated FDA-approved antipsychotics for their potential treatment of glioblastoma based on genome-wide association studies data using a 'pathway/gene-set analysis' approach. RESULTS The in-silico screening led to the discovery of 12 candidate drugs. DepMap portal revealed that 42 glioma cell lines show higher sensitivities to 12 candidate drugs than to Temozolomide, the current standard treatment for glioblastoma. CONCLUSION In particular, cell lines showed significantly higher sensitivities to Norcyclobenzaprine and Protriptyline which were predicted to bind targets to disrupt a certain molecular function such as DNA repair, response to hormones, or DNA-templated transcription, and may lead to an effect on survival-related pathways including cell cycle arrest, response to ER stress, glucose transport, and regulation of autophagy. However, it is recommended that their mechanism of action and efficacy are further determined.
Collapse
Affiliation(s)
- Wei-Zhi Lin
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| | - Yen-Chun Liu
- School of Medicine, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| | - Meng-Chang Lee
- School of Public Health, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| | - Chi-Tun Tang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, No. 325, Sec. 2, Chenggong Rd., Neihu District, Taipei, 11490 Taiwan
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
- Department of Obstetrics and Gynecology, Tri-Service General Hospital, No. 325, Sec. 2, Chenggong Rd., Neihu District, Taipei, 11490 Taiwan
| | - Yu-Tien Chang
- School of Public Health, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| | - Chi-Ming Chu
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
- School of Public Health, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| | - Chia-Yang Shiau
- Graduate Institute of Life Sciences, National Defense Medical Center, No.161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
- Fidelity Regulation Therapeutics Inc., 161, Sec. 6, Minquan E. Rd., Neihu Dist., Taipei City, 11490 Taiwan
| |
Collapse
|
28
|
Fu L, Jin W, Zhang J, Zhu L, Lu J, Zhen Y, Zhang L, Ouyang L, Liu B, Yu H. Repurposing non-oncology small-molecule drugs to improve cancer therapy: Current situation and future directions. Acta Pharm Sin B 2022; 12:532-557. [PMID: 35256933 PMCID: PMC8897051 DOI: 10.1016/j.apsb.2021.09.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/05/2021] [Accepted: 08/27/2021] [Indexed: 12/25/2022] Open
Abstract
Drug repurposing or repositioning has been well-known to refer to the therapeutic applications of a drug for another indication other than it was originally approved for. Repurposing non-oncology small-molecule drugs has been increasingly becoming an attractive approach to improve cancer therapy, with potentially lower overall costs and shorter timelines. Several non-oncology drugs approved by FDA have been recently reported to treat different types of human cancers, with the aid of some new emerging technologies, such as omics sequencing and artificial intelligence to overcome the bottleneck of drug repurposing. Therefore, in this review, we focus on summarizing the therapeutic potential of non-oncology drugs, including cardiovascular drugs, microbiological drugs, small-molecule antibiotics, anti-viral drugs, anti-inflammatory drugs, anti-neurodegenerative drugs, antipsychotic drugs, antidepressants, and other drugs in human cancers. We also discuss their novel potential targets and relevant signaling pathways of these old non-oncology drugs in cancer therapies. Taken together, these inspiring findings will shed new light on repurposing more non-oncology small-molecule drugs with their intricate molecular mechanisms for future cancer drug discovery.
Collapse
|
29
|
Persico M, Abbruzzese C, Matteoni S, Matarrese P, Campana AM, Villani V, Pace A, Paggi MG. Tackling the Behavior of Cancer Cells: Molecular Bases for Repurposing Antipsychotic Drugs in the Treatment of Glioblastoma. Cells 2022; 11:263. [PMID: 35053377 PMCID: PMC8773942 DOI: 10.3390/cells11020263] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 02/01/2023] Open
Abstract
Glioblastoma (GBM) is associated with a very dismal prognosis, and current therapeutic options still retain an overall unsatisfactorily efficacy in clinical practice. Therefore, novel therapeutic approaches and effective medications are highly needed. Since the development of new drugs is an extremely long, complex and expensive process, researchers and clinicians are increasingly considering drug repositioning/repurposing as a valid alternative to the standard research process. Drug repurposing is also under active investigation in GBM therapy, since a wide range of noncancer and cancer therapeutics have been proposed or investigated in clinical trials. Among these, a remarkable role is played by the antipsychotic drugs, thanks to some still partially unexplored, interesting features of these agents. Indeed, antipsychotic drugs have been described to interfere at variable incisiveness with most hallmarks of cancer. In this review, we analyze the effects of antipsychotics in oncology and how these drugs can interfere with the hallmarks of cancer in GBM. Overall, according to available evidence, mostly at the preclinical level, it is possible to speculate that repurposing of antipsychotics in GBM therapy might contribute to providing potentially effective and inexpensive therapies for patients with this disease.
Collapse
Affiliation(s)
- Michele Persico
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Claudia Abbruzzese
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Silvia Matteoni
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| | - Paola Matarrese
- Center for Gender Specific Medicine, Istituto Superiore di Sanità, 00162 Rome, Italy;
| | - Anna Maria Campana
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY 10032, USA;
| | - Veronica Villani
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Andrea Pace
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (V.V.); (A.P.)
| | - Marco G. Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS-Regina Elena National Cancer Institute, 00144 Rome, Italy; (M.P.); (C.A.); (S.M.)
| |
Collapse
|
30
|
Vincent-Chong VK, Seshadri M. Adrenergic-Angiogenic Crosstalk in Head and Neck Cancer: Mechanisms and Therapeutic Implications. FRONTIERS IN ORAL HEALTH 2021; 2. [PMID: 34790909 PMCID: PMC8594278 DOI: 10.3389/froh.2021.689482] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are loco-regionally aggressive tumors that often lead to debilitating changes in appearance, speech, swallowing and respiratory function in patients. It is therefore critical to develop novel targeted treatment strategies that can effectively target multiple components within the tumor microenvironment. In this regard, there has been an increased recognition of the role of neural signaling networks as mediators of disease progression in HNSCC. Here, we summarize the current knowledge on the mechanisms of adrenergic signaling in HNSCC specifically focusing on neurovascular crosstalk and the potential of targeting the adrenergic-angiogenic axis through repurposing of FDA-approved drugs against HNSCC.
Collapse
Affiliation(s)
| | - Mukund Seshadri
- Center for Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.,Department of Dentistry and Maxillofacial Prosthetics Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
31
|
|
32
|
Lianos GD, Alexiou GA, Rausei S, Galani V, Mitsis M, Kyritsis AP. Repurposing antipsychotic drugs for cancer treatment: current evidence and future perspectives. Expert Rev Anticancer Ther 2021; 22:131-134. [PMID: 34612130 DOI: 10.1080/14737140.2022.1987221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Georgios D Lianos
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - George A Alexiou
- Department of Neurosurgery, University Hospital of Ioannina, Ioannina, Greece
| | - Stefano Rausei
- Department of Surgery, ASST Valle Olona, Gallarate, Italy
| | - Vasiliki Galani
- Neurosurgical Institute, University of Ioannina, Ioannina, Greece
| | - Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | | |
Collapse
|
33
|
Antoszczak M, Markowska A, Markowska J, Huczyński A. Antidepressants and Antipsychotic Agents as Repurposable Oncological Drug Candidates. Curr Med Chem 2021; 28:2137-2174. [PMID: 32895037 DOI: 10.2174/0929867327666200907141452] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/26/2020] [Accepted: 06/10/2020] [Indexed: 11/22/2022]
Abstract
Drug repurposing, also known as drug repositioning/reprofiling, is a relatively new strategy for the identification of alternative uses of well-known therapeutics that are outside the scope of their original medical indications. Such an approach might entail a number of advantages compared to standard de novo drug development, including less time needed to introduce the drug to the market, and lower costs. The group of compounds that could be considered as promising candidates for repurposing in oncology include the central nervous system drugs, especially selected antidepressant and antipsychotic agents. In this article, we provide an overview of some antidepressants (citalopram, fluoxetine, paroxetine, sertraline) and antipsychotics (chlorpromazine, pimozide, thioridazine, trifluoperazine) that have the potential to be repurposed as novel chemotherapeutics in cancer treatment, as they have been found to exhibit preventive and/or therapeutic action in cancer patients. Nevertheless, although drug repurposing seems to be an attractive strategy to search for oncological drugs, we would like to clearly indicate that it should not replace the search for new lead structures, but only complement de novo drug development.
Collapse
Affiliation(s)
- Michał Antoszczak
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| | - Anna Markowska
- \Department of Perinatology and Women's Diseases, Poznań University of Medical Sciences, Poznan, Poland
| | - Janina Markowska
- Department of Oncology, Poznań University of Medical Sciences, Poznan, Poland
| | - Adam Huczyński
- Department of Medical Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Poznan, Poland
| |
Collapse
|
34
|
Cui Y, Wu H, Yang L, Huang T, Li J, Gong X, Li L, Sun X, Mao F, Wang Y. Chlorpromazine Sensitizes Progestin-Resistant Endometrial Cancer Cells to MPA by Upregulating PRB. Front Oncol 2021; 11:665832. [PMID: 33937078 PMCID: PMC8087176 DOI: 10.3389/fonc.2021.665832] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/29/2021] [Indexed: 12/30/2022] Open
Abstract
Medroxyprogesterone acetate (MPA) is the main conservative treatment for endometrial cancer (EC) patients desirable to preserve fertility and those who cannot suffer from surgery. Considering the high incidence of progestin resistance and recurrence of MPA treatment, we reproposed antipsychotics chlorpromazine (CPZ) as a new strategy for both progestin-sensitive and -resistant endometrial cancer. Cytobiology experiments indicated that CPZ could significantly suppress proliferation, migration/invasion and induce apoptosis in Ishikawa (ISK) and KLE EC cell lines. And xenograft mouse models were constructed to validate the antitumor effect and toxicity of CPZ in-vivo. CPZ inhibited the growth at a low dose of 3mg/kg and the mice exhibited no signs of toxicity. Next, concomitant treatment and sequential treatment with CPZ and MPA were proceeded to analysis the synergistic effect in EC cells. Concomitant treatment only performed a limited synergistic effect on apoptosis in ISK and KLE cells. Nevertheless, sequential treatment showed favorable synergistic effects in progestin-resistant KLE cells. Finally, a stable MPA-resistant cell line shRNA was established to explore the mechanism of CPZ reversing progestin resistance. Immunoblot data showed that CPZ inhibited the activation of PI3K/AKT signal in ISK and KLE cells and upregulated PRB expression in progestin-resistant cells, by which CPZ overcame progestin resistance to MPA. Thus, CPZ might act as a candidate drug for conservative treatment and sequential treatment with CPZ and MPA could be a suitable therapeutic option for progestin resistant patients.
Collapse
Affiliation(s)
- Yunxia Cui
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Huiwen Wu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Linlin Yang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Huang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China.,College of Pharmacy and Chemistry, Dali University, Dali, China.,Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lijuan Li
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Sun
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
| | - Fei Mao
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yudong Wang
- Department of Gynecologic Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Municipal Key Clinical Specialty, Female Tumor Reproductive Specialty, Shanghai, China
| |
Collapse
|
35
|
Kang L, Wang F, Zhang J, Yang H, Xia C, Qian J, Jiang G. High Chemo-/Stereoselectivity for Synthesis of Polysubstituted Monofluorinated Pyrimidyl Enol Ether Derivatives. Org Lett 2021; 23:1669-1674. [PMID: 33599505 DOI: 10.1021/acs.orglett.1c00092] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A novel intramolecular Smiles rearrangement of α-fluoro-β-keto-pyrimidylsulfones (usually used as a carbon nucleophile) was developed, providing a versatile avenue for synthesis of tri/tetra-substituted monofluorinated pyrimidyl enol ethers. Among these, diverse (Z)-monofluorovinylsulfones and sulfinates were efficiently assembled by adding extra electrophile and fine-tuning reaction conditions. The process is triggered by a keto-enol tautomerism from enol oxyanion to pyrimidine 2-carbon, completely different from the classical carbon nucleophilic addition reaction approach.
Collapse
Affiliation(s)
- Lei Kang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Fang Wang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Jinlong Zhang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Huameng Yang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Chungu Xia
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Jinlong Qian
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| | - Gaoxi Jiang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, P. R. China
| |
Collapse
|
36
|
Morak-Młodawska B, Jeleń M, Pluta K. Phenothiazines Modified with the Pyridine Ring as Promising Anticancer Agents. Life (Basel) 2021; 11:206. [PMID: 33807874 PMCID: PMC8001302 DOI: 10.3390/life11030206] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/29/2022] Open
Abstract
Azaphenothiazines are the largest and most perspective group of modified phenothiazines, and they exhibit variety of biological activities. The review sums up the current knowledge on the anticancer activity of isomeric pyridobenzothiazines and dipyridothiazines, which are modified azaphenothiazines with one and two pyridine rings, respectively, against 10 types of cancer cell lines. Some 10-substituted dipyridothiazines and even 10-unsubstituted parent compounds, such as 10H-1,9-diazaphenothiazine and 10H-3,6-diazaphenothiazine, exhibited very potent action with the IC50 values less than 1 µg/mL and 1 µM against selected cancer cell lines. The strength of the anticancer action depends both on the tricyclic ring scaffolds and the substituents at the thiazine nitrogen atom. The review discusses the kind of the substituents, nature of tricyclic ring scaffolds with the location of the azine nitrogen atoms, the types of the cancer cell lines, and the mechanism of action.
Collapse
Affiliation(s)
- Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland; (M.J.); (K.P.)
| | | | | |
Collapse
|
37
|
Matteoni S, Matarrese P, Ascione B, Buccarelli M, Ricci-Vitiani L, Pallini R, Villani V, Pace A, Paggi MG, Abbruzzese C. Anticancer Properties of the Antipsychotic Drug Chlorpromazine and Its Synergism With Temozolomide in Restraining Human Glioblastoma Proliferation In Vitro. Front Oncol 2021; 11:635472. [PMID: 33718225 PMCID: PMC7952964 DOI: 10.3389/fonc.2021.635472] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/14/2021] [Indexed: 01/06/2023] Open
Abstract
The extremely poor prognosis of patients affected by glioblastoma (GBM, grade IV glioma) prompts the search for new and more effective therapies. In this regard, drug repurposing or repositioning can represent a safe, swift, and inexpensive way to bring novel pharmacological approaches from bench to bedside. Chlorpromazine, a medication used since six decades for the therapy of psychiatric disorders, shows in vitro several features that make it eligible for repositioning in cancer therapy. Using six GBM cell lines, three of which growing as patient-derived neurospheres and displaying stem-like properties, we found that chlorpromazine was able to inhibit viability in an apoptosis-independent way, induce hyperdiploidy, reduce cloning efficiency as well as neurosphere formation and downregulate the expression of stemness genes in all these cell lines. Notably, chlorpromazine synergized with temozolomide, the first-line therapeutic in GBM patients, in hindering GBM cell viability, and both drugs strongly cooperated in reducing cloning efficiency and inducing cell death in vitro for all the GBM cell lines assayed. These results prompted us to start a Phase II clinical trial on GBM patients (EudraCT # 2019-001988-75; ClinicalTrials.gov Identifier: NCT04224441) by adding chlorpromazine to temozolomide in the adjuvant phase of the standard first-line therapeutic protocol.
Collapse
Affiliation(s)
- Silvia Matteoni
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Paola Matarrese
- Center for Gender Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Barbara Ascione
- Center for Gender Specific Medicine, Oncology Unit, Istituto Superiore di Sanità, Rome, Italy
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Roberto Pallini
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Institute of Neurosurgery, Catholic University School of Medicine, Rome, Italy
| | - Veronica Villani
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Andrea Pace
- Neuro-Oncology, IRCCS-Regina Elena National Cancer Institute, Rome, Italy
| | - Marco G Paggi
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Claudia Abbruzzese
- Cellular Networks and Molecular Therapeutic Targets, Proteomics Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
38
|
Ghouzlani A, Rafii S, Karkouri M, Lakhdar A, Badou A. The Promising IgSF11 Immune Checkpoint Is Highly Expressed in Advanced Human Gliomas and Associates to Poor Prognosis. Front Oncol 2021; 10:608609. [PMID: 33604291 PMCID: PMC7884863 DOI: 10.3389/fonc.2020.608609] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
Glioma is the most prevalent primary brain tumor. Immune checkpoint blockade has made a great stride in mending patient’s clinical outcome for multiple types of cancers. However, PD-1, CTLA-4, or VEGF blockade exhibited only poor outcome in glioma patients. This study aimed to explore the expression and role of IgSF11, an emerging immune checkpoint and a ligand of VISTA, in human gliomas. IgSF11 mRNA expression was assessed in human glioma patients at different grades using 2 independent cohorts, a set of 52 Moroccan samples, including 20 glioma tissues, 22 PBMC samples taken before and 10 PBMC samples taken after surgery; and a series of 667 patients from TCGA. In parallel, immunohistochemistry was performed to evaluate IgSF11 protein staining. IgSF11 gene expression was significantly upregulated in high grade glioma tissues, compared to low grade. IgSF11 protein also showed a significant expression in low and high-grade gliomas. Interestingly, IgSF11 expression seemed to correlate positively with other critical immune checkpoints such as PD1, PDL-1, VISTA, and surprisingly negatively with CTLA-4. Although, T cell markers appeared higher in advanced gliomas, T cell-produced pro-inflammatory genes showed similar expression levels, highly likely because of the potent immunosuppressive microenvironment. Indeed, increased expression of IgSF11 in advanced human gliomas associated with a poor overall survival. Our data strongly suggest that IgSF11 is an immune checkpoint, which is upregulated in advanced human gliomas and contributes to the immunosuppressive state resulting in a poor clinical outcome in glioma patients. IgSF11 could be considered as a possible promising therapeutic target in advanced human gliomas.
Collapse
Affiliation(s)
- Amina Ghouzlani
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Soumaya Rafii
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mehdi Karkouri
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco.,Department of Pathology, CHU Ibn Rochd, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Department of Neurosurgery, UHC Ibn Rochd, Casablanca, Morocco.,Laboratory of Research on Neurologic, Neurosensorial Diseases and Handicap, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| |
Collapse
|
39
|
Stip E, Rizvi TA, Mustafa F, Javaid S, Aburuz S, Ahmed NN, Abdel Aziz K, Arnone D, Subbarayan A, Al Mugaddam F, Khan G. The Large Action of Chlorpromazine: Translational and Transdisciplinary Considerations in the Face of COVID-19. Front Pharmacol 2020; 11:577678. [PMID: 33390948 PMCID: PMC7772402 DOI: 10.3389/fphar.2020.577678] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a severe acute respiratory syndrome (SARS) in humans that is caused by SARS-associated coronavirus type 2 (SARS-CoV-2). In the context of COVID-19, several aspects of the relations between psychiatry and the pandemic due to the coronavirus have been described. Some drugs used as antiviral medication have neuropsychiatric side effects, and conversely some psychotropic drugs have antiviral properties. Chlorpromazine (CPZ, Largactil®) is a well-established antipsychotic medication that has recently been proposed to have antiviral activity against SARS-CoV-2. This review aims to 1) inform health care professionals and scientists about the history of CPZ use in psychiatry and its potential anti- SARS-CoV-2 activities 2) inform psychiatrists about its potential anti-SARS-CoV-2 activities, and 3) propose a research protocol for investigating the use of CPZ in the treatment of COVID-19 during the potential second wave. The history of CPZ's discovery and development is described in addition to the review of literature from published studies within the discipline of virology related to CPZ. The early stages of infection with coronavirus are critical events in the course of the viral cycle. In particular, viral entry is the first step in the interaction between the virus and the cell that can initiate, maintain, and spread the infection. The possible mechanism of action of CPZ is related to virus cell entry via clathrin-mediated endocytosis. Therefore, CPZ could be useful to treat COVID-19 patients provided that its efficacy is evaluated in adequate and well-conducted clinical trials. Interestingly, clinical trials of very good quality are in progress. However, more information is still needed about the appropriate dosage regimen. In short, CPZ repositioning is defined as a new use beyond the field of psychiatry.
Collapse
Affiliation(s)
- Emmanuel Stip
- Department of Psychiatry, University of Montréal, Montréal, QC, Canada
- Department of Psychiatry and Behavioral Science, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Tahir A. Rizvi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Farah Mustafa
- Department of Biochemistry, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Syed Javaid
- Department of Psychiatry and Behavioral Science, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Salahdein Aburuz
- Department of Pharmacology and Therapeutics, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Nahida Nayaz Ahmed
- Ambulatory Healthcare Services, Al Maqtaa Healthcare Center, Middle Regions Clinics Division, SEHA, Abu Dhabi, United Arab Emirates
| | - Karim Abdel Aziz
- Department of Psychiatry and Behavioral Science, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Danilo Arnone
- Department of Psychiatry and Behavioral Science, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
- Kings’ College London, Institute of Psychiatry, Psychology, Neuroscience, Department of Psychological Medicine, Centre for Affective Disorders, London, United Kingdom
| | - Aravinthan Subbarayan
- Behavioral Sciences Institute (BSI), Al Ain Hospital, SEHA, Al Ain, United Arab Emirates
| | - Fadwa Al Mugaddam
- Department of Psychiatry, University of Montréal, Montréal, QC, Canada
| | - Gulfaraz Khan
- Department of Medical Microbiology and Immunology, College of Medicine and Health Science, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
40
|
Haloperidol Induced Cell Cycle Arrest and Apoptosis in Glioblastoma Cells. Biomedicines 2020; 8:biomedicines8120595. [PMID: 33322363 PMCID: PMC7763579 DOI: 10.3390/biomedicines8120595] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 12/18/2022] Open
Abstract
Although several antipsychotic drugs have been shown to possess anticancer activities, haloperidol, a “first-generation” antipsychotic drug, has not been extensively evaluated for potential antineoplastic properties. The aim of this study was to investigate the antitumoral effects of haloperidol in glioblastoma (GBM) U87, U251 and T98 cell lines, and the effects of combined treatment with temozolomide (TMZ) and/or radiotherapy, using 4 Gy of irradiation. The viability and proliferation of the cells were evaluated with trypan blue exclusion assay and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis, using the annexin-propidium iodide (PI), and cell cycle, cluster of differentiation (CD) expression and caspase-8 activation were measured using flow cytometry. Treatment with haloperidol significantly reduced cell viability in U87, U251 and T98 GBM cell lines. Haloperidol induced apoptosis in a dose-dependent manner, inhibited cell migration and produced an alteration in the expression of CD24/CD44. The additional effect of haloperidol, combined with temozolomide and radiation therapy, increased tumor cell death. Haloperidol was observed to induce apoptosis and to increase caspase-8 activation. In conclusion, haloperidol may represent an innovative strategy for the treatment of GBM and further studies are warranted in glioma xenograft models and other malignancies.
Collapse
|
41
|
Varalda M, Antona A, Bettio V, Roy K, Vachamaram A, Yellenki V, Massarotti A, Baldanzi G, Capello D. Psychotropic Drugs Show Anticancer Activity by Disrupting Mitochondrial and Lysosomal Function. Front Oncol 2020; 10:562196. [PMID: 33194631 PMCID: PMC7604408 DOI: 10.3389/fonc.2020.562196] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 09/15/2020] [Indexed: 12/24/2022] Open
Abstract
Background and Purpose: Drug repositioning is a promising strategy for discovering new therapeutic strategies for cancer therapy. We investigated psychotropic drugs for their antitumor activity because of several epidemiological studies reporting lower cancer incidence in individuals receiving long term drug treatment. Experimental Approach: We investigated 27 psychotropic drugs for their cytotoxic activity in colorectal carcinoma, glioblastoma and breast cancer cell lines. Consistent with the cationic amphiphilic structure of the most cytotoxic compounds, we investigated their effect on mitochondrial and lysosomal compartments. Results: Penfluridol, ebastine, pimozide and fluoxetine, fluspirilene and nefazodone showed significant cytotoxicity, in the low micromolar range, in all cell lines tested. In MCF7 cells these drugs caused mitochondrial membrane depolarization, increased the acidic vesicular compartments and induced phospholipidosis. Both penfluridol and spiperone induced AMPK activation and autophagy. Neither caspase nor autophagy inhibitors rescued cells from death induced by ebastine, fluoxetine, fluspirilene and nefazodone. Treatment with 3-methyladenine partially rescued cell death induced by pimozide and spiperone, whereas enhanced the cytotoxic activity of penfluridol. Conversely, inhibition of lysosomal cathepsins significantly reduced cell death induced by ebastin, penfluridol, pimozide, spiperone and mildly in fluoxetine treated cells. Lastly, Spiperone cytotoxicity was restricted to colorectal cancer and breast cancer and caused apoptotic cell death in MCF7 cells. Conclusions: The cytotoxicity of psychotropic drugs with cationic amphiphilic structures relied on simultaneous mitochondrial and lysosomal disruption and induction of cell death that not necessarily requires apoptosis. Since dual targeting of lysosomes and mitochondria constitutes a new promising therapeutic approach for cancer, particularly those in which the apoptotic machinery is defective, these data further support their clinical development for cancer therapy.
Collapse
Affiliation(s)
- Marco Varalda
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Annamaria Antona
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Valentina Bettio
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| | - Konkonika Roy
- Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Ajay Vachamaram
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Vaibhav Yellenki
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy
| | - Alberto Massarotti
- Department Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| | - Gianluca Baldanzi
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,Center for Translational Research on Allergic and Autoimmune Diseases (CAAD), University of Piemonte Orientale, Novara, Italy
| | - Daniela Capello
- Department of Translational Medicine, Centre of Excellence in Aging Sciences, University of Piemonte Orientale, Novara, Italy.,UPO Biobank, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
42
|
Sikora J, Podsiedlik M, Pietras T, Kosmalski M, Matłoka M, Moszczyński-Petkowski R, Wieczorek M, Markowicz-Piasecka M. Quetiapine and novel PDE10A inhibitors potentiate the anti-BuChE activity of donepezil. J Enzyme Inhib Med Chem 2020; 35:1743-1750. [PMID: 32938236 PMCID: PMC7534323 DOI: 10.1080/14756366.2020.1818739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The symptoms of Alzheimer’s disease (AD) do not include only memory loss and cognitive decline but also neuropsychiatric manifestation. These AD-related symptoms are usually treated with the aid of antipsychotics; however, their effects on cognition and safety remain unexplored. The present study determines the effects of quetiapine, an atypical antipsychotic, and two imidazo[1,2-a]pyrimidine-based inhibitors of PDE10A on the activity of human cholinesterases. Quetiapine moderately inhibited BuChE (IC50 = 6.08 ± 1.64 µmol/L) but improved the anti-BuChE properties of donepezil by decreasing its IC50 value. Both PDE10A inhibitors were found to possess moderate anti-AChE properties. The combined mixtures of donepezil and imidazo[1,2-a]pyrimidine analogues produce a synergistic anti-BuChE effect which was greater than either compound alone, improving the IC50 value by approximately six times. These favourable interactions between quetiapine, PDE10A inhibitors and clinically approved donepezil, resulting in improved anti-BuChE activity, can lead to a wider variety of potent AD treatment options.
Collapse
Affiliation(s)
- Joanna Sikora
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, Lodz, Poland
| | - Maria Podsiedlik
- Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Pietras
- Department of Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Marcin Kosmalski
- Department of Clinical Pharmacology, Medical University of Lodz, Lodz, Poland
| | - Mikołaj Matłoka
- Research and Development Department, Celon Pharma S.A, Łomianki, Poland
| | | | - Maciej Wieczorek
- Research and Development Department, Celon Pharma S.A, Łomianki, Poland
| | - Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
43
|
Avendaño-Félix M, Aguilar-Medina M, Bermudez M, Lizárraga-Verdugo E, López-Camarillo C, Ramos-Payán R. Refocusing the Use of Psychiatric Drugs for Treatment of Gastrointestinal Cancers. Front Oncol 2020; 10:1452. [PMID: 32923398 PMCID: PMC7456997 DOI: 10.3389/fonc.2020.01452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022] Open
Abstract
Gastrointestinal cancers (GICs) are the most common human tumors worldwide. Treatments have limited effects, and increasing global cancer burden makes it necessary to investigate alternative strategies such as drug repurposing. Interestingly, it has been found that psychiatric drugs (PDs) are promising as a new generation of cancer chemotherapies due to their anti-neoplastic properties. This review compiles the state of the art about how PDs have been redirected for cancer therapeutics in GICs. PDs, especially anti-psychotics, anti-depressants and anti-epileptic drugs, have shown effects on cell viability, cell growth, inhibition of proliferation (cell cycle arrest), apoptosis promotion by caspases activation or cytochrome C release, production of reactive oxygen species (ROS) and nuclear fragmentation over esophageal, gastric, colorectal, liver and pancreatic cancers. Additionally, PDs can inhibit neovascularization, invasion and metastasis in a dose-dependent manner. Moreover, they can induce chemosensibilization to 5-fluorouracil and cisplatin and can act synergistically with anti-neoplastic drugs such as gemcitabine, paclitaxel and oxaliplatin. All anti-cancer activities are given by activation or inhibition of pathways such as HDAC1/PTEN/Akt, EGFR/ErbB2/ErbB3, and PI3K/Akt; PI3K-AK-mTOR, HDAC1/PTEN/Akt; Wnt/β-catenin. Further investigations and clinical trials are needed to elucidate all molecular mechanisms involved on anti-cancer activities as well as adverse effects on patients.
Collapse
Affiliation(s)
- Mariana Avendaño-Félix
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Mercedes Bermudez
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - Erik Lizárraga-Verdugo
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Rosalío Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán, Mexico
| |
Collapse
|
44
|
Zhang H, Fan F, Yu Y, Wang Z, Liu F, Dai Z, Zhang L, Liu Z, Cheng Q. Clinical characterization, genetic profiling, and immune infiltration of TOX in diffuse gliomas. J Transl Med 2020; 18:305. [PMID: 32762688 PMCID: PMC7409670 DOI: 10.1186/s12967-020-02460-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/26/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Immunotherapies targeting glioblastoma (GBM) have led to significant improvements in patient outcomes. TOX is closely associated with the immune environment surrounding tumors, but its role in gliomas is not fully understood. METHODS Using data from The Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA), we analyzed the transcriptomes of 1691 WHO grade I-IV human glioma samples. The R language was used to perform most of the statistical analyses. Somatic mutations and somatic copy number variation (CNV) were analyzed using GISTIC 2.0. RESULTS TOX was down-regulated in malignant gliomas compared to low grade gliomas, and upregulated in the proneural and IDH mutant subtypes of GBM. TOXlow tumours are associated with the loss of PTEN and amplification of EGFR, while TOXhigh tumours harbor frequent mutations in IDH1 (91%). TOX was highly expressed in leading edge regions of tumours. Gene ontology and pathway analyses demonstrated that TOX was enriched in multiple immune related processes including lymphocyte migration in GBM. Finally, TOX had a negative association with the infiltration of several immune cell types in the tumour microenvironment. CONCLUSION TOX has the potential to be a new prognostic marker for GBM.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fan Fan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- Center for Medical Genetics and Hunan Provincial Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuanqiang Yu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
- Clinical Diagnosis and Therapeutic Center of Glioma, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, People's Republic of China.
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
45
|
Long S, Li M, Liu J, Yang Y, Li G. Identification of immunologic subtype and prognosis of GBM based on TNFSF14 and immune checkpoint gene expression profiling. Aging (Albany NY) 2020; 12:7112-7128. [PMID: 32310827 PMCID: PMC7202515 DOI: 10.18632/aging.103065] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/24/2020] [Indexed: 02/03/2023]
Abstract
Immune-checkpoint therapy has failed to show significant benefit in glioblastoma (GBM) patients. Immunologic subtypes of GBM are necessary to identify patients who might benefit from immune-checkpoint therapy. This study reviewed 152 GBM samples from The Cancer Genome Atlas (TCGA) and 214 GBM samples from Chinese Glioma Genome Atlas (CGGA). Correlation analysis showed that immune checkpoint genes (ICGs) were mainly positively correlated. The prognostic analysis of the overall survival showed that there was a significant correlation between the overall survival (OS) and the prognosis of ICGs, in which the TNFSF14 gene was a significant adverse prognostic factor. Combined with TMB and neoantigens, we found that TNFSF9 and CD27 were significantly negatively correlated with TMB and neoantigens. The association between adaptive immune pathway genes and ICG expression showed that they were positively correlated with ICGs, indicating that adaptive immune pathway genes have a certain regulatory effect on the expression of ICGs. The analysis of clinical features of the samples showed that the higher the expression of ICGs, the more likely to be correlated with mutant isocitrate dehydrogenase (IDH), while the lower the expression level of IDH, the more likely to be significantly correlated with the primary GBM. Survival analysis showed that low expression of PD-L1, IDO1, or CTLA4 with TNFSF14 in the low expression group had the best prognosis, while high expression of IDO1 or CD274 with TNFSF14 in the high expression group and low expression of CTLA4 with TNFSF14 in the high expression group had the worst prognosis. We conclude that TNFSF14 is a biomarker to identify immunologic subtype and prognosis with other ICGs in GBM and may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Shengrong Long
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Mingdong Li
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jia Liu
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi Yang
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guangyu Li
- Department of Neurosurgery, First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
46
|
Pluta K, Jeleń M, Morak-Młodawska B. The Smiles rearrangement in the syntheses of azaphenothiazines. Part I. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127501] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
47
|
Choi J, Lee YJ, Yoon YJ, Kim CH, Park SJ, Kim SY, Doo Kim N, Cho Han D, Kwon BM. Pimozide suppresses cancer cell migration and tumor metastasis through binding to ARPC2, a subunit of the Arp2/3 complex. Cancer Sci 2019; 110:3788-3801. [PMID: 31571309 PMCID: PMC6890432 DOI: 10.1111/cas.14205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
ARPC2 is a subunit of the Arp2/3 complex, which is essential for lamellipodia, invadopodia and filopodia, and ARPC2 has been identified as a migrastatic target molecule. To identify ARPC2 inhibitors, we generated an ARPC2 knockout DLD-1 human colon cancer cell line using the clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) system and explored gene signature-based strategies, such as a connectivity map (CMap) using the gene expression profiling data of ARPC2 knockout and knockdown cells. From the CMap-based drug discovery strategy, we identified pimozide (a clinically used antipsychotic drug) as a migrastatic drug and ARPC2 inhibitor. Pimozide inhibited the migration and invasion of various cancer cells. Through drug affinity responsive target stability (DARTS) analysis and cellular thermal shift assay (CETSA), it was confirmed that pimozide directly binds to ARPC2. Pimozide increased the lag phase of Arp2/3 complex-dependent actin polymerization and inhibited the vinculin-mediated recruitment of ARPC2 to focal adhesions in cancer cells. To validate the likely binding of pimozide to ARPC2, mutant cells, including ARPC2F225A , ARPC2F247A and ARPC2Y250F cells, were prepared using ARPC2 knockout cells prepared by gene-editing technology. Pimozide strongly inhibited the migration of mutant cells because the mutated ARPC2 likely has a larger binding pocket than the wild-type ARPC2. Therefore, pimozide is a potential ARPC2 inhibitor, and ARPC2 is a new molecular target. Taken together, the results of the present study provide new insights into the molecular mechanism and target that are responsible for the antitumor and antimetastatic activity of pimozide.
Collapse
Affiliation(s)
- Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yae Jin Yoon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Cheol-Hee Kim
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Seung-Jin Park
- Korea Research Institute of Bioscience and Biotechnology, Personalized Genomic Medicine Research Center, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Seon-Young Kim
- Korea Research Institute of Bioscience and Biotechnology, Personalized Genomic Medicine Research Center, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| |
Collapse
|
48
|
In Vitro Activity of Sertraline, an Antidepressant, Against Antibiotic-Susceptible and Antibiotic-Resistant Helicobacter pylori Strains. Pathogens 2019; 8:pathogens8040228. [PMID: 31717683 PMCID: PMC6963513 DOI: 10.3390/pathogens8040228] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/02/2019] [Accepted: 11/08/2019] [Indexed: 12/19/2022] Open
Abstract
Antibiotic resistance of Helicobacter pylori, a spiral bacterium associated with gastric diseases, is a topic that has been intensively discussed in last decades. Recent discoveries indicate promising antimicrobial and antibiotic-potentiating properties of sertraline (SER), an antidepressant substance. The aim of the study, therefore, was to determine the antibacterial activity of SER in relation to antibiotic-sensitive and antibiotic-resistant H. pylori strains. The antimicrobial tests were performed using a diffusion-disk method, microdilution method, and time-killing assay. The interaction between SER and antibiotics (amoxicillin, clarithromycin, tetracycline, and metronidazole) was determined by using a checkerboard method. In addition, the study was expanded to include observations by light, fluorescence, and scanning electron microscopy. The growth inhibition zones were in the range of 19–37 mm for discs impregnated with 2 mg of SER. The minimal inhibitory concentrations (MICs) and minimal bactericidal concentrations (MBCs) counted for 2–8 µg/mL and 4–8 µg/mL, respectively. The time-killing assay showed the time-dependent and concentration-dependent bactericidal activity of SER. Bacteria exposed to MBCs (but not sub-MICs and MICs ≠ MBCs) underwent morphological transformation into coccoid forms. This mechanism, however, was not protective because these cells after a 24-h incubation had a several-fold reduced green/red fluorescence ratio compared to the control. Using the checkerboard assay, a synergistic/additive interaction of SER with all four antibiotics tested was demonstrated. These results indicate that SER may be a promising anti-H. pylori compound.
Collapse
|
49
|
Zhuo C, Xun Z, Hou W, Ji F, Lin X, Tian H, Zheng W, Chen M, Liu C, Wang W, Chen C. Surprising Anticancer Activities of Psychiatric Medications: Old Drugs Offer New Hope for Patients With Brain Cancer. Front Pharmacol 2019; 10:1262. [PMID: 31695618 PMCID: PMC6817617 DOI: 10.3389/fphar.2019.01262] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 09/30/2019] [Indexed: 12/17/2022] Open
Abstract
Despite decades of research and major efforts, malignant brain tumors remain among the deadliest of all cancers. Recently, an increasing number of psychiatric drugs has been proven to possess suppressing activities against brain tumors, and rapid progress has been made in understanding the potential mechanisms of action of these drugs. In particular, the traditional mood stabilizer valproic acid, the widely used antidepressants fluoxetine and escitalopram oxalate, and the atypical psychiatric drug aripiprazole have demonstrated promise for application in brain tumor treatment strategies through multiple lines of laboratory, preclinical, and clinical evidence. The unexpected discovery of the anticancer properties of these drugs has ignited interest in the repurposing of other psychiatric drugs to combat brain cancer. In this review, we synthesize recent progress in understanding the potential molecular mechanisms underlying the brain cancer-killing activities of representative psychiatric drugs. We also identify key limitations in the repurposing of these medications that must be overcome to enhance our ability to successfully prevent and treat brain cancer, especially in the most vulnerable groups of patients, such as children and adolescents, pregnant women, and those with unfavorable genetic variants. Moreover, we propose perspectives that may guide future research and provide long-awaited new hope to patients with brain cancer and their families.
Collapse
Affiliation(s)
- Chuanjun Zhuo
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China.,Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China.,Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China.,Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Zhiyuan Xun
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weihong Hou
- Department of Biochemistry and Molecular Biology, Zhengzhou University, Zhengzhou, China.,Department of Biology, University of North Carolina at Charlotte, Charlotte, NC, United States
| | - Feng Ji
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Xiaodong Lin
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Hongjun Tian
- Department of Psychiatric-Neuroimaging-Genetics and Morbidity Laboratory (PNGC-Lab), Nankai University Affiliated Anding Hospital, Tianjin Mental Health Center, Mental Health Teaching Hospital, Tianjin Medical University, Tianjin, China
| | - Weifang Zheng
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| | - Min Chen
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Chuanxin Liu
- Department of Psychiatry, School of Mental Health, Psychiatric Genetics Laboratory (PSYG-Lab), Jining Medical University, Jining, China
| | - Wenqiang Wang
- Department of China-Canada Biological Psychiatry Lab, Xiamen Xianyue Hospital, Xiamen, China
| | - Ce Chen
- Department of Psychiatry, Wenzhou Seventh People's Hospital, Wenzhou, China
| |
Collapse
|
50
|
HASSANI M, GHASSEMI-BARGHI N, MODANLOO M, MOHAMMADPOUR A, SHOKRZADEH M. CYTOTOXIC EFFECTS OF DULOXETINE ON MKN45 AND NIH3T3 CELL LINES AND GENOTOXIC EFFECTS ON HUMAN PERIPHERAL BLOOD LYMPHOCYTES. ARQUIVOS DE GASTROENTEROLOGIA 2019; 56:372-376. [DOI: 10.1590/s0004-2803.201900000-71] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
Abstract
ABSTRACT BACKGROUND: Gastric cancer is the second leading cause of cancer-related death globally. Unfortunately, the survival rate of the gastric cancer patients who underwent chemotherapy following surgery has been less than a half. Besides, chemotherapy has many side effects. Current evidence suggests that some antidepressants like duloxetine have growth-inhibiting effects against a number of cancer cell lines. OBJECTIVE: Thus, the aim of this study was to determine the cytotoxic and genotoxic effects of duloxetine on gastric cancer. METHODS: In this regard, the cytotoxicity and genotoxicity of duloxetine were investigated in MKN45 and NIH3T3 cell lines by MTT assay and on peripheral blood lymphocytes by MN assay. For this purpose, cells were cultured in 96 wells plate. Stock solutions of duloxetine and cisplatin were prepared. After cell incubation with different concentrations of duloxetine (1, 10, 25, 50, 100 and 200 μL), MTT solution was added. For micronucleus assay fresh blood was added to RPMI culture medium 1640 supplemented, and different concentrations of duloxetine (1, 10, 25, 50, 100 and 200 μL) were added. RESULTS: The cytotoxicity of duloxetine on MKN45 cancer cell line and NIH3T3 normal cell line were studied followed by MTT assay. duloxetine exhibited higher IC50 in the MKN45 cells in comparison with the NIH3T3 cells. In addition, genotoxic effect of duloxetine was evaluated by micronucleus assay. The results revealed that duloxetine induced more DNA damage at 100 and 200 μM and no significant difference at 200 μM with respect to cisplatin, but it had less genotoxic effects at 100 and 50 μM concentrations. CONCLUSION: Although, in this study, duloxetine had less genotoxicity than cisplatin in concentrations under 200 μM and showed cytotoxic effects as well, due to its IC50, it cannot be considered as a better choice for gastric cancer therapies with respect to cisplatin as a common anticancer drug.
Collapse
|