101
|
Telmisartan restricts Chikungunya virus infection in vitro and in vivo through the AT1/PPAR-γ/MAPKs pathways. Antimicrob Agents Chemother 2021; 66:e0148921. [PMID: 34748384 DOI: 10.1128/aac.01489-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chikungunya virus (CHIKV) has re-emerged as a global public health threat. The inflammatory pathways of RAS and PPAR-γ are usually involved in viral infections. Thus, Telmisartan (TM) with known capacity to block AT1 receptor and activate PPAR-γ, was investigated against CHIKV. The anti-CHIKV effect of TM was investigated in vitro (Vero, RAW 264.7 cells and hPBMCs) and in vivo (C57BL/6 mice). TM was found to abrogate CHIKV infection efficiently (IC50 of 15.34-20.89μM in the Vero and RAW 264.7 cells respectively). Viral RNA and proteins were reduced remarkably. Additionally, TM interfered in the early and late stages of CHIKV life cycle with efficacy in both pre and post-treatment assay. Moreover, the agonist of AT1 receptor and antagonist of PPAR-γ increased CHIKV infection suggesting TM's anti-viral potential by modulating host factors. Besides, reduced activation of all major MAPKs, NF-κB (p65) and cytokines by TM through the inflammatory axis supported the fact that the anti-CHIKV efficacy of TM is partly mediated through the AT1/PPAR-γ/MAPKs pathways. Interestingly, at the human equivalent dose, TM abrogated CHIKV infection and inflammation significantly leading to reduced clinical score and complete survival of C57BL/6 mice. Additionally, TM reduced infection in hPBMC derived monocyte-macrophage populations in vitro. Hence, TM was found to reduce CHIKV infection by targeting both viral and host factors. Considering its safety and in vivo efficacy, it can be a suitable candidate in future for repurposing against CHIKV.
Collapse
|
102
|
Bhattacharya D, Becker C, Readhead B, Goossens N, Novik J, Fiel MI, Cousens LP, Magnusson B, Backmark A, Hicks R, Dudley JT, Friedman SL. Repositioning of a novel GABA-B receptor agonist, AZD3355 (Lesogaberan), for the treatment of non-alcoholic steatohepatitis. Sci Rep 2021; 11:20827. [PMID: 34675338 PMCID: PMC8531016 DOI: 10.1038/s41598-021-99008-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 09/14/2021] [Indexed: 01/02/2023] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is a rising health challenge, with no approved drugs. We used a computational drug repositioning strategy to uncover a novel therapy for NASH, identifying a GABA-B receptor agonist, AZD3355 (Lesogaberan) previously evaluated as a therapy for esophageal reflux. AZD3355's potential efficacy in NASH was tested in human stellate cells, human precision cut liver slices (hPCLS), and in vivo in a well-validated murine model of NASH. In human stellate cells AZD3355 significantly downregulated profibrotic gene and protein expression. Transcriptomic analysis of these responses identified key regulatory nodes impacted by AZD3355, including Myc, as well as MAP and ERK kinases. In PCLS, AZD3355 down-regulated collagen1α1, αSMA and TNF-α mRNAs as well as secreted collagen1α1. In vivo, the drug significantly improved histology, profibrogenic gene expression, and tumor development, which was comparable to activity of obeticholic acid in a robust mouse model of NASH, but awaits further testing to determine its relative efficacy in patients. These data identify a well-tolerated clinical stage asset as a novel candidate therapy for human NASH through its hepatoprotective, anti-inflammatory and antifibrotic mechanisms of action. The approach validates computational methods to identify novel therapies in NASH in uncovering new pathways of disease development that can be rapidly translated into clinical trials.
Collapse
Affiliation(s)
- Dipankar Bhattacharya
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Ave. Room 1170, New York, NY, 10029, USA
| | - Christine Becker
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin Readhead
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Arizona State University-Banner Neurodegenerative Disease Research Center, Arizona, USA
| | - Nicolas Goossens
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Ave. Room 1170, New York, NY, 10029, USA
- Division of Gastroenterology, Geneva University Hospital, Geneva, Switzerland
| | - Jacqueline Novik
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Maria Isabel Fiel
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leslie P Cousens
- Emerging Innovations, Discovery Sciences, R&D, AstraZeneca, Boston, MA, USA
| | - Björn Magnusson
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Anna Backmark
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Ryan Hicks
- BioPharmaceuticals R&D Cell Therapy, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Joel T Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, Box 1123, 1425 Madison Ave. Room 1170, New York, NY, 10029, USA.
| |
Collapse
|
103
|
Das A, Agarwal P, Jain GK, Aggarwal G, Lather V, Pandita D. Repurposing drugs as novel triple negative breast cancer therapeutics. Anticancer Agents Med Chem 2021; 22:515-550. [PMID: 34674627 DOI: 10.2174/1871520621666211021143255] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 05/23/2021] [Accepted: 06/29/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Among all the types of breast cancer (BC), triple negative breast cancer (TNBC) is the most aggressive form having high metastasis and recurrence rate with limited treatment options. Conventional treatments such as chemotherapy and radiotherapy have lots of toxic side effects and also no FDA approved therapies are available till now. Repurposing of old clinically approved drugs towards various targets of TNBC is the new approach with lesser side effects and also leads to successful inexpensive drug development with less time consuming. Medicinal plants containg various phytoconstituents (flavonoids, alkaloids, phenols, essential oils, tanins, glycosides, lactones) plays very crucial role in combating various types of diseases and used in drug development process because of having lesser side effects. OBJECTIVE The present review focuses in summarization of various categories of repurposed drugs against multitarget of TNBC and also summarizes the phytochemical categories that targets TNBC singly or in combination with synthetic old drugs. METHODS Literature information was collected from various databases such as Pubmed, Web of Science, Scopus and Medline to understand and clarify the role and mechanism of repurposed synthetic drugs and phytoconstituents aginst TNBC by using keywords like "breast cancer", "repurposed drugs", "TNBC" and "phytoconstituents". RESULTS Various repurposed drugs and phytochemicals targeting different signaling pathways that exerts their cytotoxic activities on TNBC cells ultimately leads to apoptosis of cells and also lowers the recurrence rate and stops the metastasis process. CONCLUSION Inhibitory effects seen in different levels, which provides information and evidences to researchers towards drug developments process and thus further more investigations and researches need to be taken to get the better therapeutic treatment options against TNBC.
Collapse
Affiliation(s)
- Amiya Das
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Pallavi Agarwal
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| | - Geeta Aggarwal
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector-125, Noida, 201313. India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Institute of Pharmaceutical Sciences & Research, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, Govt. of NCT of Delhi, New Delhi, 110017. India
| |
Collapse
|
104
|
Puzari U, Fernandes PA, Mukherjee AK. Advances in the Therapeutic Application of Small-Molecule Inhibitors and Repurposed Drugs against Snakebite. J Med Chem 2021; 64:13938-13979. [PMID: 34565143 DOI: 10.1021/acs.jmedchem.1c00266] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The World Health Organization has declared snakebite as a neglected tropical disease. Antivenom administration is the sole therapy against venomous snakebite; however, several limitations of this therapy reinforce the dire need for an alternative and/or additional treatment against envenomation. Inhibitors against snake venoms have been explored from natural resources and are synthesized in the laboratory; however, repurposing of small-molecule therapeutics (SMTs) against the principal toxins of snake venoms to inhibit their lethality and/or obnoxious effect of envenomation has been garnering greater attention owing to their established pharmacokinetic properties, low-risk attributes, cost-effectiveness, ease of administration, and storage stability. Nevertheless, SMTs are yet to be approved and commercialized for snakebite treatment. Therefore, we have systematically reviewed and critically analyzed the scenario of small synthetic inhibitors and repurposed drugs against snake envenomation from 2005 to date and proposed novel approaches and commercialization strategies for the development of efficacious therapies against snake envenomation.
Collapse
Affiliation(s)
- Upasana Puzari
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India
| | - Pedro Alexandrino Fernandes
- LAQV@REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua Do Campo Alegre S/N, 4169-007 Porto, Portugal
| | - Ashis K Mukherjee
- Department of Molecular Biology and Biotechnology, School of Sciences, Tezpur University, Tezpur-784028, Assam, India.,Institute of Advanced Study in Science and Technology, Vigyan Path Garchuk, Paschim Boragaon, Guwahati-781035, Assam, India
| |
Collapse
|
105
|
Lin W, Sun J, Sadahira T, Xu N, Wada K, Liu C, Araki M, Xu A, Watanabe M, Nasu Y, Huang P. Discovery and Validation of Nitroxoline as a Novel STAT3 Inhibitor in Drug-resistant Urothelial Bladder Cancer. Int J Biol Sci 2021; 17:3255-3267. [PMID: 34421363 PMCID: PMC8375225 DOI: 10.7150/ijbs.63125] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/06/2021] [Indexed: 01/11/2023] Open
Abstract
Repeated cycles of first-line chemotherapy drugs such as doxorubicin (DOX) and cisplatin (CIS) trigger frequent chemoresistance in recurrent urothelial bladder cancer (UBC). Nitroxoline (NTX), an antibiotic to treat urinary tract infections, has been recently repurposed for cancer treatment. Here we aimed to investigate whether NTX suppresses drug-resistant UBC and its molecular mechanism. The drug-resistant cell lines T24/DOX and T24/CIS were established by continual exposure of parental cell line T24 to DOX and CIS, respectively. T24/DOX and T24/CIS cells were resistant to DOX and CIS, respectively, but they were sensitive to NTX time- and dose-dependently. Overexpressions of STAT3 and P-glycoprotein (P-gp) were identified in T24/DOX and T24/CIS, which could be reversed by NTX. Western blot revealed that NTX downregulated p-STAT3, c-Myc, Cyclin D1, CDK4, CDK6, Bcl-xL, Mcl-1, and Survivin, which were further confirmed by Stattic, a selective STAT3 inhibitor. In vivo, NTX exhibited the significant anti-tumor effect in T24/DOX and T24/CIS tumor-bearing mice. These results suggested that NTX-induced P-gp reversal, G0/G1 arrest, and apoptosis in drug-resistant UBC were mediated by inhibition of STAT3 signaling. Our findings repurpose NTX as a novel STAT3 inhibitor to induce P-gp reversal, G0/G1 arrest, and apoptosis in drug-resistant UBC.
Collapse
Affiliation(s)
- Wenfeng Lin
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jingkai Sun
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Takuya Sadahira
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Naijin Xu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Koichiro Wada
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Peng Huang
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.,Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.,Okayama Medical Innovation Center, Okayama University, Okayama, Japan
| |
Collapse
|
106
|
Drug Repurposing to Identify a Synergistic High-Order Drug Combination to Treat Sunitinib-Resistant Renal Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13163978. [PMID: 34439134 PMCID: PMC8391235 DOI: 10.3390/cancers13163978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary In this study, drug combination screening was used to design a multidrug combination consisting of repurposed drugs to treat sunitinib-resistant clear cell renal cell carcinoma. In the frame of this project, the multidrug combination has been optimized and validated and an insight into the mechanism of action is given. The multidrug combinations significantly altered the transcription of genes related to apoptosis and metabolic pathways. Further analysis of the metabolism revealed strong upregulation of the presence of sphingolipids after multidrug combination treatment. Final evaluation for translation of the multidrug combination in ex vivo organoid-like cultures demonstrated significant anti-cancer efficacy. Abstract Repurposed drugs have been evaluated for the management of clear cell renal cell carcinoma (ccRCC), but only a few have influenced the overall survival of patients with advanced disease. To combine repurposed non-oncology with oncological drugs, we applied our validated phenotypic method, which consisted of a reduced experimental part and data modeling. A synergistic optimized multidrug combination (ODC) was identified to significantly reduce the energy levels in cancer remaining inactive in non-cancerous cells. The ODC consisted of Rapta-C, erlotinib, metformin and parthenolide and low doses. Molecular and functional analysis of ODC revealed a loss of adhesiveness and induction of apoptosis. Gene-expression network analysis displayed significant alterations in the cellular metabolism, confirmed by LC-MS based metabolomic analysis, highlighting significant changes in the lipid classes. We used heterotypic in vitro 3D co-cultures and ex vivo organoids to validate the activity of the ODC, maintaining an efficacy of over 70%. Our results show that repurposed drugs can be combined to target cancer cells selectively with prominent activity. The strong impact on cell adherence and metabolism indicates a favorable mechanism of action of the ODC to treat ccRCC.
Collapse
|
107
|
McCarthy C, Fedele S, Ottensmeier C, Shaw RJ. Early-Phase Interventional Trials in Oral Cancer Prevention. Cancers (Basel) 2021; 13:cancers13153845. [PMID: 34359746 PMCID: PMC8345124 DOI: 10.3390/cancers13153845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/24/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Oral cancer is a devastating disease with increasing incidence worldwide. Oral epithelial dysplasia (OED) is a potentially malignant disorder and patients with OED are at increased risk of developing oral cancer. Current strategies for management of OED include surgery or close observation and both fail to address the underlying pathogenesis of the disease. There is an urgent need for evidence-based medical treatments for OED to prevent oral cancer development in this cohort. Chemoprevention trials to date have not delivered therapeutic agents for routine clinical practice. Historically, there has been significant heterogeneity in the design of oral cancer chemoprevention trials, with most failing to selectively recruit patients with biopsy-proven OED, which limits the usefulness of the findings in the OED population. The present paper aims to review the current evidence and the methodology of early-phase trials in oral cancer chemoprevention. Novel strategies in oral cancer chemoprevention will also be discussed. Abstract The increasing breadth of molecular targets, promise of immune-targeted therapies and repurposed agents have heightened interest in cancer prevention. While, to date, testing of oral cancer chemoprevention strategies has failed to deliver therapeutic agents for routine clinical practice, there remains an urgent need for further clinical research to overcome this hurdle. Patients at the greatest risk of disease stand to benefit the most from inclusion in clinical trials; therefore, there is a need to carefully define this population using validated clinical and molecular markers. Safety, tolerability and the efficacy of interventions is assessed through carefully selected endpoints. These endpoints may include pharmacodynamic, clinical, histological and on-target molecular modifications as an individual or as a composite endpoint. Early-phase trials provide an area of opportunity to explore novel and repurposed agents in the setting of oral cancer chemoprevention, eventually leading to phase III trials with clinical endpoints such as transformation and clinical outcome; these studies are large, lengthy and expensive and should be reserved for the most promising of agents. This paper will explore current evidence in oral cancer chemoprevention, drug repurposing, selection of appropriate endpoints for early-phase trials and novel therapeutic angles in oral cancer chemoprevention.
Collapse
Affiliation(s)
- Caroline McCarthy
- Liverpool Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L3 9TA, UK; (C.O.); (R.J.S.)
- Department of Oral Medicine, Liverpool University Dental Hospital, Liverpool L3 9TA, UK
- Correspondence: ; Tel.: +44-7904-363-109
| | - Stefano Fedele
- Eastman Dental Institute, University College London, 21 University Street, London WC1E 6DE, UK;
- National Institute for Health Research, University College London Hospitals Biomedical Research Centre, Maple House Suite A 1st floor, 149 Tottenham Court Road, London W1T 7DN, UK
| | - Christian Ottensmeier
- Liverpool Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L3 9TA, UK; (C.O.); (R.J.S.)
| | - Richard J. Shaw
- Liverpool Head and Neck Centre, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L3 9TA, UK; (C.O.); (R.J.S.)
| |
Collapse
|
108
|
Cheon YH, Lee CH, Kim S, Park GD, Kwak SC, Cho HJ, Kim JY, Lee MS. Pitavastatin prevents ovariectomy-induced osteoporosis by regulating osteoclastic resorption and osteoblastic formation. Biomed Pharmacother 2021; 139:111697. [PMID: 34243614 DOI: 10.1016/j.biopha.2021.111697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 10/21/2022] Open
Abstract
Excessive osteoclast activity, along with relatively weak osteoblast function, is strongly associated with bone disease. Therefore, studies to identify novel anti-osteoporosis candidates with dual actions of inhibiting osteoclastogenesis and increasing osteoblastogenesis may provide an ideal approach for treating osteoporosis. Pitavastatin, an inhibitor of 3-hydroxy-3 methyl-glutaryl coenzyme A reductase, has demonstrated various pharmacological activities, including anti-inflammation, bone anabolic effects, vasodilation, and inhibition of revascularization; however, the precise effects and mechanisms of pitavastatin on the regulation of osteoblast and osteoclast activity need to be comprehensively elucidated. Herein, we demonstrated that pitavastatin is a potential candidate for treating osteoporosis by enhancing osteoblast differentiation and bone growth and inhibiting osteoclast differentiation and bone resorption. Pitavastatin exerted dose-dependent inhibitory effects on receptor activator of nuclear factor kappa-B ligand-induced osteoclast formation, bone resorption, and osteoclast-specific marker gene expression. These inhibitory effects were achieved by inhibiting the Akt, NF-κB, and mitogen-activated protein kinase (p38, ERK, and JNK) signaling pathways, resulting in the downregulation of major transcription factors c-Fos and NFATc1. Furthermore, pitavastatin potentially stimulated osteoblast differentiation by activating alkaline phosphatase (ALP), enhancing mineralization by Alizarin Red S, and increasing the expression of osteoblastogenic marker genes such as runt-related transcription factor 2, ALP, osteocalcin, and collagen type 1 alpha. Furthermore, we evaluated the therapeutic potential of pitavastatin in ovariectomy-induced systematic bone loss based on micro-computed tomography and histological analysis of femurs. Our findings demonstrated a new function and mechanism for pitavastatin in bone remodeling, indicating its potential as a therapeutic candidate in treating osteoporosis by inhibiting osteoclastic resorption and promoting osteoblastic formation.
Collapse
Affiliation(s)
- Yoon-Hee Cheon
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Chang Hoon Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Soojin Kim
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Gyeong Do Park
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Sung Chul Kwak
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea; Department of Anatomy, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea
| | - Hae Joong Cho
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea; Department of Obstetrics and Gynecology, Wonkwang University Hospital, Iksan 54538, Republic of Korea
| | - Ju-Young Kim
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea.
| | - Myeung Su Lee
- Musculoskeletal and Immune Disease Research Institute, School of Medicine, Wonkwang University, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea; Division of Rheumatology, Department of Internal Medicine, Wonkwang University Hospital, 460 Iksandae-ro, Iksan, Jeonbuk 54538, Republic of Korea.
| |
Collapse
|
109
|
Abo Elmaaty A, Hamed MIA, Ismail MI, B. Elkaeed E, S. Abulkhair H, Khattab M, Al-Karmalawy AA. Computational Insights on the Potential of Some NSAIDs for Treating COVID-19: Priority Set and Lead Optimization. Molecules 2021; 26:3772. [PMID: 34205704 PMCID: PMC8234583 DOI: 10.3390/molecules26123772] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 02/05/2023] Open
Abstract
The discovery of drugs capable of inhibiting SARS-CoV-2 is a priority for human beings due to the severity of the global health pandemic caused by COVID-19. To this end, repurposing of FDA-approved drugs such as NSAIDs against COVID-19 can provide therapeutic alternatives that could be utilized as an effective safe treatment for COVID-19. The anti-inflammatory activity of NSAIDs is also advantageous in the treatment of COVID-19, as it was found that SARS-CoV-2 is responsible for provoking inflammatory cytokine storms resulting in lung damage. In this study, 40 FDA-approved NSAIDs were evaluated through molecular docking against the main protease of SARS-CoV-2. Among the tested compounds, sulfinpyrazone 2, indomethacin 3, and auranofin 4 were proposed as potential antagonists of COVID-19 main protease. Molecular dynamics simulations were also carried out for the most promising members of the screened NSAID candidates (2, 3, and 4) to unravel the dynamic properties of NSAIDs at the target receptor. The conducted quantum mechanical study revealed that the hybrid functional B3PW91 provides a good description of the spatial parameters of auranofin 4. Interestingly, a promising structure-activity relationship (SAR) was concluded from our study that could help in the future design of potential SARS-CoV-2 main protease inhibitors with expected anti-inflammatory effects as well. NSAIDs may be used by medicinal chemists as lead compounds for the development of potent SARS-CoV-2 (Mpro) inhibitors. In addition, some NSAIDs can be selectively designated for treatment of inflammation resulting from COVID-19.
Collapse
Affiliation(s)
- Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt;
| | - Mohammed I. A. Hamed
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, Fayoum University, Fayoum 63514, Egypt;
| | - Muhammad I. Ismail
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, Cairo-Suez Desert Road, Cairo 11837, Egypt;
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Ad Diriyah, Riyadh 13713, Saudi Arabia;
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt;
| | - Hamada S. Abulkhair
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11884, Egypt;
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| | - Muhammad Khattab
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre, Cairo 12622, Egypt;
| | - Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt
| |
Collapse
|
110
|
Nehme R, Hallal R, El Dor M, Kobeissy F, Gouilleux F, Mazurier F, Zibara K. Repurposing of Acriflavine to Target Chronic Myeloid Leukemia Treatment. Curr Med Chem 2021; 28:2218-2233. [PMID: 32900342 DOI: 10.2174/0929867327666200908114411] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 11/22/2022]
Abstract
Drug repurposing has lately received increasing interest in several diseases especially in cancers, due to its advantages in facilitating the development of new therapeutic strategies, by adopting a cost-friendly approach and avoiding the strict Food and Drug Administration (FDA) regulations. Acriflavine (ACF) is an FDA approved molecule that has been extensively studied since 1912 with antiseptic, trypanocidal, anti-viral, anti-bacterial and anti-cancer effects. ACF has been shown to block the growth of solid and hematopoietic tumor cells. Indeed, ACF acts as an inhibitor of various proteins, including DNA-dependent protein kinases C (DNA-PKcs), topoisomerase I and II, hypoxia-inducible factor 1α (HIF-1α), in addition to its recent discovery as an inhibitor of the signal transducer and activator of transcription (STAT). Chronic myeloid leukemia (CML) is a clonal myeloproliferative disorder characterized by the expression of the constitutively active tyrosine kinase BCR-ABL. This protein allows the activation of several signaling pathways known for their role in cell proliferation and survival, such as the JAK/STAT pathway. CML therapy, based on tyrosine kinase inhibitors (TKIs), such as imatinib (IM), is highly effective. However, 15% of patients are refractory to IM, where in some cases, 20-30% of patients become resistant. Thus, we suggest the repurposing of ACF in CML after IM failure or in combination with IM to improve the anti-tumor effects of IM. In this review, we present the different pharmacological properties of ACF along with its anti-leukemic effects in the hope of its repurposing in CML therapy.
Collapse
Affiliation(s)
- Rawan Nehme
- Universite de Tours, EA7501 GICC, Tours, France
| | | | - Maya El Dor
- Universite de Tours, EA7501 GICC, Tours, France
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | | | | |
Collapse
|
111
|
Yacouba A, Olowo-Okere A, Yunusa I. Repurposing of antibiotics for clinical management of COVID-19: a narrative review. Ann Clin Microbiol Antimicrob 2021; 20:37. [PMID: 34020659 PMCID: PMC8139224 DOI: 10.1186/s12941-021-00444-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 05/11/2021] [Indexed: 12/22/2022] Open
Abstract
Background Drug repurposing otherwise known as drug repositioning or drug re-profiling is a time-tested approach in drug discovery through which new medical uses are being established for already known drugs. Antibiotics are among the pharmacological agents being investigated for potential anti-SARS-COV-2 activities. The antibiotics are used either to resolve bacterial infections co-existing with COVID-19 infections or exploitation of their potential antiviral activities. Herein, we aimed to review the various antibiotics that have been repositioned for the management of COVID-19. Methods This literature review was conducted from a methodical search on PubMed and Web of Science regarding antibiotics used in patients with COVID-19 up to July 5, 2020. Results Macrolide and specifically azithromycin is the most common antibiotic used in the clinical management of COVID-19. The other antibiotics used in COVID-19 includes teicoplanin, clarithromycin, doxycycline, tetracyclines, levofloxacin, moxifloxacin, ciprofloxacin, and cefuroxime. In patients with COVID-19, antibiotics are used for their immune-modulating, anti-inflammatory, and antiviral properties. The precise antiviral mechanism of most of these antibiotics has not been determined. Moreover, the use of some of these antibiotics against SARS-CoV-2 infection remains highly controversial and not widely accepted. Conclusion The heavy use of antibiotics during the COVID-19 pandemic would likely worsen antibiotic resistance crisis. Consequently, antibiotic stewardship should be strengthened in order to prevent the impacts of COVID-19 on the antibiotic resistance crisis.
Collapse
Affiliation(s)
- Abdourahamane Yacouba
- Faculté des Sciences de la Santé, Université Abdou Moumouni, P.M.B. 10896, Niamey, Niger.
| | - Ahmed Olowo-Okere
- Faculty of Pharmaceutical Sciences, Usmanu Danfodiyo University, P.M.B. 2346, Sokoto, Nigeria
| | - Ismaeel Yunusa
- Department of Clinical Pharmacy and Outcomes Sciences, University of South Carolina College of Pharmacy, Columbia, SC, USA
| |
Collapse
|
112
|
Sun YJ, Velez G, Parsons DE, Li K, Ortiz ME, Sharma S, McCray PB, Bassuk AG, Mahajan VB. Structure-based phylogeny identifies avoralstat as a TMPRSS2 inhibitor that prevents SARS-CoV-2 infection in mice. J Clin Invest 2021; 131:147973. [PMID: 33844653 PMCID: PMC8121520 DOI: 10.1172/jci147973] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/07/2021] [Indexed: 12/19/2022] Open
Abstract
Drugs targeting host proteins can act prophylactically to reduce viral burden early in disease and limit morbidity, even with antivirals and vaccination. Transmembrane serine protease 2 (TMPRSS2) is a human protease required for SARS coronavirus 2 (SARS-CoV-2) viral entry and may represent such a target. We hypothesized that drugs selected from proteins related by their tertiary structure, rather than their primary structure, were likely to interact with TMPRSS2. We created a structure-based phylogenetic computational tool named 3DPhyloFold to systematically identify structurally similar serine proteases with known therapeutic inhibitors and demonstrated effective inhibition of SARS-CoV-2 infection in vitro and in vivo. Several candidate compounds, avoralstat, PCI-27483, antipain, and soybean trypsin inhibitor, inhibited TMPRSS2 in biochemical and cell infection assays. Avoralstat, a clinically tested kallikrein-related B1 inhibitor, inhibited SARS-CoV-2 entry and replication in human airway epithelial cells. In an in vivo proof of principle, avoralstat significantly reduced lung tissue titers and mitigated weight loss when administered prophylactically to mice susceptible to SARS-CoV-2, indicating its potential to be repositioned for coronavirus disease 2019 (COVID-19) prophylaxis in humans.
Collapse
Affiliation(s)
- Young Joo Sun
- Molecular Surgery Lab, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA
| | - Gabriel Velez
- Molecular Surgery Lab, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA
- Medical Scientist Training Program, University of Iowa, Iowa City, Iowa, USA
| | - Dylan E. Parsons
- Molecular Surgery Lab, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA
- Stanford ChEM-H Medicinal Chemistry Knowledge Center, Stanford University, Palo Alto, California, USA
| | | | | | | | - Paul B. McCray
- Department of Pediatrics
- Department of Microbiology and Immunology
| | - Alexander G. Bassuk
- Department of Pediatrics
- Department of Neurology, and
- Iowa Neuroscience Institute, University of Iowa, Iowa City, Iowa, USA
| | - Vinit B. Mahajan
- Molecular Surgery Lab, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
113
|
Verbaanderd C, Rooman I, Huys I. Exploring new uses for existing drugs: innovative mechanisms to fund independent clinical research. Trials 2021; 22:322. [PMID: 33947441 PMCID: PMC8093905 DOI: 10.1186/s13063-021-05273-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Finding new therapeutic uses for existing medicines could lead to safe, affordable and timely new treatment options for patients with high medical needs. However, due to a lack of economic incentives, pharmaceutical developers are rarely interested to invest in research with approved medicines, especially when they are out of basic patent or regulatory protection. Consequently, potential new uses for these medicines are mainly studied in independent clinical trials initiated and led by researchers from academia, research institutes, or collaborative groups. Yet, additional financial support is needed to conduct expensive phase III clinical trials to confirm the results from exploratory research. METHODS In this study, scientific and grey literature was searched to identify and evaluate new mechanisms for funding clinical trials with repurposed medicines. Semi-structured interviews were conducted with 16 European stakeholders with expertise in clinical research, funding mechanisms and/or drug repurposing between November 2018 and February 2019 to consider the future perspectives of applying new funding mechanisms. RESULTS Traditional grant funding awarded by government and philanthropic organisations or companies is well known and widely implemented in all research fields. In contrast, only little research has focused on the application potential of newer mechanisms to fund independent clinical research, such as social impact bonds, crowdfunding or public-private partnerships. Interviewees stated that there is a substantial need for additional financial support in health research, especially in areas where there is limited commercial interest. However, the implementation of new funding mechanisms is facing several practical and financial challenges, such as a lack of expertise and guidelines, high transaction costs and difficulties to measure health outcomes. Furthermore, interviewees highlighted the need for increased collaboration and centralisation at a European and international level to make clinical research more efficient and reduce the need for additional funding. CONCLUSIONS New funding mechanisms to support clinical research may become more important in the future but the unresolved issues identified in the current study warrant further exploration.
Collapse
Affiliation(s)
- Ciska Verbaanderd
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
- Anticancer Fund, Strombeek-Bever, Belgium.
| | - Ilse Rooman
- Anticancer Fund, Strombeek-Bever, Belgium
- Oncology Research Centre, Vrije Universiteit Brussel, Brussels, Belgium
| | - Isabelle Huys
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| |
Collapse
|
114
|
Al-Karmalawy AA, Dahab MA, Metwaly AM, Elhady SS, Elkaeed EB, Eissa IH, Darwish KM. Molecular Docking and Dynamics Simulation Revealed the Potential Inhibitory Activity of ACEIs Against SARS-CoV-2 Targeting the hACE2 Receptor. Front Chem 2021; 9:661230. [PMID: 34017819 PMCID: PMC8129187 DOI: 10.3389/fchem.2021.661230] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
The rapid and global spread of a new human coronavirus, Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) has produced an immediate urgency to discover promising targets for the treatment of COVID-19. Here, we consider drug repurposing as an attractive approach that can facilitate the drug discovery process by repurposing existing pharmaceuticals to treat illnesses other than their primary indications. We review current information concerning the global health issue of COVID-19 including promising approved drugs, e.g., human angiotensin-converting enzyme inhibitors (hACEIs). Besides, we describe computational approaches to be used in drug repurposing and highlight examples of in-silico studies of drug development efforts against SARS-CoV-2. Alacepril and lisinopril were found to interact with human angiotensin-converting enzyme 2 (hACE2), the host entranceway for SARS-CoV-2 spike protein, through exhibiting the most acceptable rmsd_refine values and the best binding affinity through forming a strong hydrogen bond with Asn90, which is assumed to be essential for the activity, as well as significant extra interactions with other receptor-binding residues. Furthermore, molecular dynamics (MD) simulations followed by calculation of the binding free energy were also carried out for the most promising two ligand-pocket complexes from docking studies (alacepril and lisinopril) to clarify some information on their thermodynamic and dynamic properties and confirm the docking results as well. These results we obtained probably provided an excellent lead candidate for the development of therapeutic drugs against COVID-19. Eventually, animal experiments and accurate clinical trials are needed to confirm the potential preventive and treatment effect of these compounds.
Collapse
Affiliation(s)
- Ahmed A. Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt, Damietta, Egypt
| | - Mohammed A. Dahab
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed M. Metwaly
- Pharmacognosy Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Sameh S. Elhady
- Department of Natural Products, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Eslam B. Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, Ad Diriyah, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Ibrahim H. Eissa
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, Egypt
| | - Khaled M. Darwish
- Department of Medicinal Chemistry, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
115
|
Odiase E, Zhang X, Chang Y, Nelson M, Balaji U, Gu J, Zhang Q, Pan Z, Jon Spechler S, Souza RF. In Esophageal Squamous Cells From Eosinophilic Esophagitis Patients, Th2 Cytokines Increase Eotaxin-3 Secretion Through Effects on Intracellular Calcium and a Non-Gastric Proton Pump. Gastroenterology 2021; 160:2072-2088.e6. [PMID: 33581123 PMCID: PMC9013281 DOI: 10.1053/j.gastro.2021.02.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS In upper airway cells, T helper 2 cytokines that signal through interleukin-4 (IL-4) receptor-α have been shown to stimulate eotaxin-3 secretion via a nongastric proton pump (ngH+,K+ATPase). To seek novel targets for eosinophilic esophagitis (EoE) treatments, we evaluated ngH+,K+ATPase expression in EoE squamous cells, and explored molecular pathways involved in eotaxin-3 secretion by IL-4 receptor-α signaling. METHODS ngH+,K+ATPase expression in EoE cells was evaluated by quantitative real-time polymerase chain reaction and Western blotting. IL-4-stimulated eotaxin-3 secretion was measured by enzyme-linked immunosorbent assay after treatment with omeprazole, SCH 28080 (potassium-competitive acid blocker), ethylene glycol-bis(β-aminoethyl)-N,N,N',N'-tetraacetoxymethyl ester (calcium chelator), 2-aminoethoxydiphenyl borate (inhibitor of endoplasmic reticulum calcium release), verapamil, and diltiazem (L-type calcium channel inhibitors). Intracellular calcium transients were measured by Fluo-4 fluorescence. Key experiments were confirmed in EoE primary cells and in RNA sequencing datasets from mucosal biopsies of patients with EoE and controls. RESULTS EoE cells expressed ngH+,K+ATPase messenger RNA and protein. Omeprazole and SCH 28080 decreased IL-4-stimulated eotaxin-3 secretion. IL-4 increased intracellular calcium transients, and IL-4-stimulated eotaxin-3 secretion was blocked by ethylene glycol-bis(β-aminoethyl)-N,N,N',N'-tetraacetoxymethyl ester, 2-aminoethoxydiphenyl borate, verapamil, and diltiazem. The combination of omeprazole and verapamil suppressed IL-4-stimulated eotaxin-3 secretion more than either agent alone. EoE biopsies expressed higher ngH+,K+ATPase and exhibited more calcium signaling than controls. CONCLUSIONS EoE cells express a nongastric proton pump that mediates T helper 2 cytokine-stimulated eotaxin-3 secretion. IL-4 induces calcium release from the endoplasmic reticulum and calcium entry via L-type calcium channels, increasing intracellular calcium that contributes to eotaxin-3 secretion by EoE cells. L-type calcium channel inhibitors block T helper 2 cytokine-stimulated eotaxin-3 secretion, suggesting a potential role for these agents in EoE treatment.
Collapse
Affiliation(s)
- Eunice Odiase
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas,Department of Pediatrics, Children’s Hospital of Colorado, Aurora, Colorado
| | - Xi Zhang
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Yan Chang
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Melissa Nelson
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Uthra Balaji
- Biostatistics Core, Baylor Scott & White Research Institute, Dallas, Texas
| | - Jinghua Gu
- Biostatistics Core, Baylor Scott & White Research Institute, Dallas, Texas
| | - Qiuyang Zhang
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Stuart Jon Spechler
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| | - Rhonda F. Souza
- Department of Medicine, Center for Esophageal Diseases, Baylor University Medical Center and Center for Esophageal Research, Baylor Scott & White Research Institute, Dallas, Texas
| |
Collapse
|
116
|
Pessanha de Carvalho L, Kreidenweiss A, Held J. Drug Repurposing: A Review of Old and New Antibiotics for the Treatment of Malaria: Identifying Antibiotics with a Fast Onset of Antiplasmodial Action. Molecules 2021; 26:2304. [PMID: 33921170 PMCID: PMC8071546 DOI: 10.3390/molecules26082304] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 11/24/2022] Open
Abstract
Malaria is one of the most life-threatening infectious diseases and constitutes a major health problem, especially in Africa. Although artemisinin combination therapies remain efficacious to treat malaria, the emergence of resistant parasites emphasizes the urgent need of new alternative chemotherapies. One strategy is the repurposing of existing drugs. Herein, we reviewed the antimalarial effects of marketed antibiotics, and described in detail the fast-acting antibiotics that showed activity in nanomolar concentrations. Antibiotics have been used for prophylaxis and treatment of malaria for many years and are of particular interest because they might exert a different mode of action than current antimalarials, and can be used simultaneously to treat concomitant bacterial infections.
Collapse
Affiliation(s)
- Lais Pessanha de Carvalho
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
| | - Andrea Kreidenweiss
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
- Centre de Recherches Medicales de Lambaréné (CERMEL), Lambaréné BP 242, Gabon
| | - Jana Held
- Institute of Tropical Medicine, University of Tuebingen, 72074 Tuebingen, Germany; (L.P.d.C.); (A.K.)
- Centre de Recherches Medicales de Lambaréné (CERMEL), Lambaréné BP 242, Gabon
| |
Collapse
|
117
|
Dotolo S, Marabotti A, Facchiano A, Tagliaferri R. A review on drug repurposing applicable to COVID-19. Brief Bioinform 2021; 22:726-741. [PMID: 33147623 PMCID: PMC7665348 DOI: 10.1093/bib/bbaa288] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 09/16/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Drug repurposing involves the identification of new applications for existing drugs at a lower cost and in a shorter time. There are different computational drug-repurposing strategies and some of these approaches have been applied to the coronavirus disease 2019 (COVID-19) pandemic. Computational drug-repositioning approaches applied to COVID-19 can be broadly categorized into (i) network-based models, (ii) structure-based approaches and (iii) artificial intelligence (AI) approaches. Network-based approaches are divided into two categories: network-based clustering approaches and network-based propagation approaches. Both of them allowed to annotate some important patterns, to identify proteins that are functionally associated with COVID-19 and to discover novel drug–disease or drug–target relationships useful for new therapies. Structure-based approaches allowed to identify small chemical compounds able to bind macromolecular targets to evaluate how a chemical compound can interact with the biological counterpart, trying to find new applications for existing drugs. AI-based networks appear, at the moment, less relevant since they need more data for their application.
Collapse
Affiliation(s)
| | | | | | - Roberto Tagliaferri
- Artificial Intelligence, Statistical Pattern Recognition, Clustering, Biomedical imaging and Bioinformatics
| |
Collapse
|
118
|
Repositioning metformin and propranolol for colorectal and triple negative breast cancers treatment. Sci Rep 2021; 11:8091. [PMID: 33854147 PMCID: PMC8047046 DOI: 10.1038/s41598-021-87525-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 03/22/2021] [Indexed: 12/27/2022] Open
Abstract
Drug repositioning refers to new uses for existing drugs outside the scope of the original medical indications. This approach fastens the process of drug development allowing finding effective drugs with reduced side effects and lower costs. Colorectal cancer (CRC) is often diagnosed at advanced stages, when the probability of chemotherapy resistance is higher. Triple negative breast cancer (TNBC) is the most aggressive type of breast cancer, highly metastatic and difficult to treat. For both tumor types, available treatments are generally associated to severe side effects. In our work, we explored the effect of combining metformin and propranolol, two repositioned drugs, in both tumor types. We demonstrate that treatment affects viability, epithelial-mesenchymal transition and migratory potential of CRC cells as we described before for TNBC. We show that combined treatment affects different steps leading to metastasis in TNBC. Moreover, combined treatment is also effective preventing the development of 5-FU resistant CRC. Our data suggest that combination of metformin and propranolol could be useful as a putative adjuvant treatment for both TNBC and CRC and an alternative for chemo-resistant CRC, providing a low-cost alternative therapy without associated toxicity.
Collapse
|
119
|
Abstract
INTRODUCTION Knowledge graphs have proven to be promising systems of information storage and retrieval. Due to the recent explosion of heterogeneous multimodal data sources generated in the biomedical domain, and an industry shift toward a systems biology approach, knowledge graphs have emerged as attractive methods of data storage and hypothesis generation. AREAS COVERED In this review, the author summarizes the applications of knowledge graphs in drug discovery. They evaluate their utility; differentiating between academic exercises in graph theory, and useful tools to derive novel insights, highlighting target identification and drug repurposing as two areas showing particular promise. They provide a case study on COVID-19, summarizing the research that used knowledge graphs to identify repurposable drug candidates. They describe the dangers of degree and literature bias, and discuss mitigation strategies. EXPERT OPINION Whilst knowledge graphs and graph-based machine learning have certainly shown promise, they remain relatively immature technologies. Many popular link prediction algorithms fail to address strong biases in biomedical data, and only highlight biological associations, failing to model causal relationships in complex dynamic biological systems. These problems need to be addressed before knowledge graphs reach their true potential in drug discovery.
Collapse
Affiliation(s)
- Finlay MacLean
- Target Identification., BenevolentAI, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
120
|
Functional Fine-Tuning of Metabolic Pathways by the Endocannabinoid System-Implications for Health and Disease. Int J Mol Sci 2021; 22:ijms22073661. [PMID: 33915889 PMCID: PMC8036872 DOI: 10.3390/ijms22073661] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
The endocannabinoid system (ECS) employs a huge network of molecules (receptors, ligands, and enzymatic machinery molecules) whose interactions with other cellular networks have still not been fully elucidated. Endogenous cannabinoids are molecules with the primary function of control of multiple metabolic pathways. Maintenance of tissue and cellular homeostasis by functional fine-tuning of essential metabolic pathways is one of the key characteristics of the ECS. It is implicated in a variety of physiological and pathological states and an attractive pharmacological target yet to reach its full potential. This review will focus on the involvement of ECS in glucose and lipid metabolism, food intake regulation, immune homeostasis, respiratory health, inflammation, cancer and other physiological and pathological states will be substantiated using freely available data from open-access databases, experimental data and literature review. Future directions should envision capturing its diversity and exploiting pharmacological options beyond the classical ECS suspects (exogenous cannabinoids and cannabinoid receptor monomers) as signaling through cannabinoid receptor heteromers offers new possibilities for different biochemical outcomes in the cell.
Collapse
|
121
|
Mishra CB, Pandey P, Sharma RD, Malik MZ, Mongre RK, Lynn AM, Prasad R, Jeon R, Prakash A. Identifying the natural polyphenol catechin as a multi-targeted agent against SARS-CoV-2 for the plausible therapy of COVID-19: an integrated computational approach. Brief Bioinform 2021; 22:1346-1360. [PMID: 33386025 PMCID: PMC7799228 DOI: 10.1093/bib/bbaa378] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 11/03/2020] [Accepted: 11/26/2020] [Indexed: 01/18/2023] Open
Abstract
The global pandemic crisis, coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has claimed the lives of millions of people across the world. Development and testing of anti-SARS-CoV-2 drugs or vaccines have not turned to be realistic within the timeframe needed to combat this pandemic. Here, we report a comprehensive computational approach to identify the multi-targeted drug molecules against the SARS-CoV-2 proteins, whichare crucially involved in the viral-host interaction, replication of the virus inside the host, disease progression and transmission of coronavirus infection. Virtual screening of 75 FDA-approved potential antiviral drugs against the target proteins, spike (S) glycoprotein, human angiotensin-converting enzyme 2 (hACE2), 3-chymotrypsin-like cysteine protease (3CLpro), cathepsin L (CTSL), nucleocapsid protein, RNA-dependent RNA polymerase (RdRp) and non-structural protein 6 (NSP6), resulted in the selection of seven drugs which preferentially bind to the target proteins. Further, the molecular interactions determined by molecular dynamics simulation revealed that among the 75 drug molecules, catechin can effectively bind to 3CLpro, CTSL, RBD of S protein, NSP6 and nucleocapsid protein. It is more conveniently involved in key molecular interactions, showing binding free energy (ΔGbind) in the range of -5.09 kcal/mol (CTSL) to -26.09 kcal/mol (NSP6). At the binding pocket, catechin is majorly stabilized by the hydrophobic interactions, displays ΔEvdW values: -7.59 to -37.39 kcal/mol. Thus, the structural insights of better binding affinity and favorable molecular interaction of catechin toward multiple target proteins signify that catechin can be potentially explored as a multi-targeted agent against COVID-19.
Collapse
Affiliation(s)
| | - Preeti Pandey
- Department of Chemistry & Biochemistry, University of Oklahoma, OK, USA
| | | | - Md Zubbair Malik
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Raj Kumar Mongre
- College of Pharmacy, Sookmyung Women’s University, Seoul, South Korea
| | - Andrew M Lynn
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology and is the dean of Faculty of Science Engineering and Technology, Amity University Haryana, Haryana 122413, India
| | - Raok Jeon
- College of Pharmacy, Sookmyung Women’s University, Seoul, South Korea
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity Institute of Integrative Sciences and Health, Amity University, Haryana
| |
Collapse
|
122
|
Billamboz M, Fatima Z, Hameed S, Jawhara S. Promising Drug Candidates and New Strategies for Fighting against the Emerging Superbug Candida auris. Microorganisms 2021; 9:microorganisms9030634. [PMID: 33803604 PMCID: PMC8003017 DOI: 10.3390/microorganisms9030634] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/04/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Invasive fungal infections represent an expanding threat to public health. During the past decade, a paradigm shift of candidiasis from Candida albicans to non-albicans Candida species has fundamentally increased with the advent of Candida auris. C. auris was identified in 2009 and is now recognized as an emerging species of concern and underscores the urgent need for novel drug development strategies. In this review, we discuss the genomic epidemiology and the main virulence factors of C. auris. We also focus on the different new strategies and results obtained during the past decade in the field of antifungal design against this emerging C. auris pathogen yeast, based on a medicinal chemist point of view. Critical analyses of chemical features and physicochemical descriptors will be carried out along with the description of reported strategies.
Collapse
Affiliation(s)
- Muriel Billamboz
- Inserm, CHU Lille, Institut Pasteur Lille, Université Lille, U1167—RID-AGE—Facteurs de Risque et Déterminants Moléculaires des Maladies liées au Vieillissement, F-59000 Lille, France
- Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, F-59000 Lille, France
- Correspondence: (M.B.); (S.J.)
| | - Zeeshan Fatima
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram 122413, India; (Z.F.); (S.H.)
| | - Saif Hameed
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Gurugram 122413, India; (Z.F.); (S.H.)
| | - Samir Jawhara
- UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Centre National de la Recherche Scientifique, INSERM U1285, University of Lille, F-59000 Lille, France
- Correspondence: (M.B.); (S.J.)
| |
Collapse
|
123
|
Elmaaty AA, Alnajjar R, Hamed MIA, Khattab M, Khalifa MM, Al-Karmalawy AA. Revisiting activity of some glucocorticoids as a potential inhibitor of SARS-CoV-2 main protease: theoretical study. RSC Adv 2021; 11:10027-10042. [PMID: 35423530 PMCID: PMC8695394 DOI: 10.1039/d0ra10674g] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Accepted: 02/17/2021] [Indexed: 02/05/2023] Open
Abstract
The global breakout of COVID-19 and raised death toll has prompted scientists to develop novel drugs capable of inhibiting SARS-CoV-2. Conducting studies on repurposing some FDA-approved glucocorticoids can be a promising prospective for finding a treatment for COVID-19. In addition, the use of anti-inflammatory drugs, such as glucocorticoids, is a pivotal step in the treatment of critical cases of COVID-19, as they can provoke an inflammatory cytokine storm, damaging lungs. In this study, 22 FDA-approved glucocorticoids were identified through in silico (molecular docking) studies as the potential inhibitors of COVID-19's main protease. From tested compounds, ciclesonide 11, dexamethasone 2, betamethasone 1, hydrocortisone 4, fludrocortisone 3, and triamcinolone 8 are suggested as the most potent glucocorticoids active against COVID-19's main protease. Moreover, molecular dynamics simulations followed by the calculations of the binding free energy using MM-GBSA were carried out for the aforementioned promising candidate-screened glucocorticoids. In addition, quantum chemical calculations revealed two electron-rich sites on ciclesonide where binding interactions with the main protease and cleavage of the prodrug to the active metabolite take place. Our results have ramifications for conducting preclinical and clinical studies on promising glucocorticoids to hasten the development of effective therapeutics against COVID-19. Another advantage is that some glucocorticoids can be prioritized over others for the treatment of inflammation accompanying COVID-19.
Collapse
Affiliation(s)
- Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University Port Said 42526 Egypt
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi Benghazi Libya
- Department of Chemistry, University of Cape Town Rondebosch 7701 South Africa
| | - Mohammed I A Hamed
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, Fayoum University Fayoum 63514 Egypt
| | - Muhammad Khattab
- Department of Chemistry of Natural and Microbial Products, Division of Pharmaceutical and Drug Industries, National Research Centre Cairo 12622 Egypt
| | - Mohamed M Khalifa
- Department of Pharmaceutical Medicinal Chemistry& Drug Design, Faculty of Pharmacy (Boys), Al-Azhar University Cairo 11884 Egypt
| | - Ahmed A Al-Karmalawy
- Department of Pharmaceutical Medicinal Chemistry, Faculty of Pharmacy, Horus University-Egypt New Damietta 34518 Egypt
| |
Collapse
|
124
|
Dos Santos MC, Scaini JLR, Lopes MVC, Rodrigues BG, Silva NO, Borges CRL, Dos Santos SC, Dos Santos Machado K, Werhli AV, da Silva PEA, Lourenço MCS, da Silva ET, de Souza MVN, de Lima VR, Gonçalves RSB. Mefloquine synergism with anti-tuberculosis drugs and correlation to membrane effects: Biologic, spectroscopic and molecular dynamics simulations studies. Bioorg Chem 2021; 110:104786. [PMID: 33740676 DOI: 10.1016/j.bioorg.2021.104786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/07/2020] [Accepted: 02/26/2021] [Indexed: 12/26/2022]
Abstract
Studies displaying the combination of mefloquine (MFL) with anti-tuberculosis (TB) substances are limited in the literature. In this work, the effect of MFL-association with two first-line anti-TB drugs and six fluoroquinolones was evaluated against Mycobacterium tuberculosis drug resistant strains. MFL showed synergistic interaction with isoniazid, pyrazinamide, and several fluoroquinolones, reaching fractional inhibitory concentration indexes (FICIs) ranging from 0.03 to 0.5. In order to better understand the observed results, two approaches have been explored: (i) spectroscopic responses attributed to the effect of MFL on physicochemical properties related to a liposomal membrane model composed by soybean asolectin; (ii) molecular dynamics (MD) simulation data regarding MFL interaction with a membrane model based on PIM2, a lipid constituent of the mycobacterial cell wall. FTIR and NMR data showed that MFL affects expressively the region between the phosphate and the first methylene groups of soybean asolectin membranes, disordering these regions. MD simulations results detected high MFL density in the glycolipid interface and showed that the drug increases the membrane lateral diffusion, enhancing its permeability. The obtained results suggest that synergistic activities related to MFL are attributed to its effect of lipid disorder and membrane permeability enhancement.
Collapse
Affiliation(s)
- Marinalva Cardoso Dos Santos
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - João Luís Rheingantz Scaini
- COMBI-Lab, Grupo de Biologia Computacional, Centro de Ciências Computacionais, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil; Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Márcio Vinicius Costa Lopes
- Instituto de Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos, 149 - Cidade Universitária, Rio de Janeiro, RJ 21941-909, Brazil
| | - Beatriz Gonçalves Rodrigues
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Nichole Osti Silva
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Carla Roberta Lopes Borges
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Sandra Cruz Dos Santos
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Karina Dos Santos Machado
- COMBI-Lab, Grupo de Biologia Computacional, Centro de Ciências Computacionais, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Adriano Velasque Werhli
- COMBI-Lab, Grupo de Biologia Computacional, Centro de Ciências Computacionais, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Pedro Eduardo Almeida da Silva
- Faculdade de Medicina, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil
| | - Maria C S Lourenço
- Instituto de Pesquisas Clínica Evandro Chagas-IPEC, Av. Brasil, 4365 Manguinhos, Rio de Janeiro, Brazil
| | - Emerson T da Silva
- FioCruz-Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos-Far-Manguinhos, Rua Sizenando Nabuco, 100, Manguinhos, 21041-250 Rio de Janeiro, RJ, Brazil
| | - Marcus V N de Souza
- FioCruz-Fundação Oswaldo Cruz, Instituto de Tecnologia em Fármacos-Far-Manguinhos, Rua Sizenando Nabuco, 100, Manguinhos, 21041-250 Rio de Janeiro, RJ, Brazil
| | - Vânia Rodrigues de Lima
- Grupo de Investigação de Interações Moleculares em Membranas, Escola de Química e Alimentos, Programa de Pós-Graduação em Química Tecnológica e Ambiental, Universidade Federal do Rio Grande - FURG, Av. Itália, km 8, Campus Carreiros, 96203-900, Rio Grande, RS, Brazil.
| | - Raoni Schroeder B Gonçalves
- Instituto de Química, Universidade Federal do Rio de Janeiro, Av. Athos da Silveira Ramos, 149 - Cidade Universitária, Rio de Janeiro, RJ 21941-909, Brazil.
| |
Collapse
|
125
|
Vitner EB, Achdout H, Avraham R, Politi B, Cherry L, Tamir H, Yahalom-Ronen Y, Paran N, Melamed S, Erez N, Israely T. Glucosylceramide synthase inhibitors prevent replication of SARS-CoV-2 and influenza virus. J Biol Chem 2021; 296:100470. [PMID: 33639165 PMCID: PMC7904475 DOI: 10.1016/j.jbc.2021.100470] [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: 12/18/2020] [Revised: 02/20/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The ongoing COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a major threat to global health. Vaccines are ideal solutions to prevent infection, but treatments are also needed for those who have contracted the virus to limit negative outcomes, when vaccines are not applicable. Viruses must cross host cell membranes during their life cycle, creating a dependency on processes involving membrane dynamics. Thus, in this study, we examined whether the synthetic machinery for glycosphingolipids, biologically active components of cell membranes, can serve as a therapeutic target to combat SARS-CoV-2. We examined the antiviral effect of two specific inhibitors of glucosylceramide synthase (GCS): (i) Genz-123346, an analogue of the United States Food and Drug Administration-approved drug Cerdelga and (ii) GENZ-667161, an analogue of venglustat, which is currently under phase III clinical trials. We found that both GCS inhibitors inhibit replication of SARS-CoV-2. Moreover, these inhibitors also disrupt replication of influenza virus A/PR/8/34 (H1N1). Our data imply that synthesis of glycosphingolipids is necessary to support viral life cycles and suggest that GCS inhibitors should be further explored as antiviral therapies.
Collapse
Affiliation(s)
- Einat B Vitner
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel.
| | - Hagit Achdout
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Roy Avraham
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Boaz Politi
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Lilach Cherry
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Hadas Tamir
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Yfat Yahalom-Ronen
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Nir Paran
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Sharon Melamed
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Noam Erez
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Tomer Israely
- Departments of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| |
Collapse
|
126
|
Veale CGL. Into the Fray! A Beginner's Guide to Medicinal Chemistry. ChemMedChem 2021; 16:1199-1225. [PMID: 33591595 DOI: 10.1002/cmdc.202000929] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Indexed: 12/31/2022]
Abstract
Modern medicinal chemistry is a complex, multidimensional discipline that operates at the interface of the chemical and biological sciences. The medicinal chemistry contribution to drug discovery is typically described in the context of the well-recited linear progression of the drug discovery pipeline. However, compound optimization is idiosyncratic to each project, and clear definitions of hit and lead molecules and the subsequent progress along the pipeline becomes easily blurred. In addition, this description lacks insight into the entangled relationship between chemical and pharmacological properties, and thus provides limited guidance on how innovative medicinal chemistry strategies can be applied to solve optimization problems, regardless of the stage in the pipeline. Through discussion and illustrative examples, this article seeks to provide insights into the finesse of medicinal chemistry and the subtlety of balancing chemical properties pharmacology. In so doing, it aims to serve as an accessible and simple-to-digest guide for anyone who wishes to learn about the underlying principles of medicinal chemistry, in a context that has been decoupled from the pipeline description.
Collapse
Affiliation(s)
- Clinton G L Veale
- School of Chemistry and Physics, Pietermaritzburg Campus, University of KwaZulu-Natal, Private Bag X01, Pietermaritzburg, Scottsville, 3209, South Africa
| |
Collapse
|
127
|
Roy S, Dhaneshwar S, Bhasin B. Drug Repurposing: An Emerging Tool for Drug Reuse, Recycling and Discovery. Curr Drug Res Rev 2021; 13:101-119. [PMID: 33573567 DOI: 10.2174/2589977513666210211163711] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 09/07/2020] [Accepted: 10/26/2020] [Indexed: 11/22/2022]
Abstract
Drug repositioning or repurposing is a revolutionary breakthrough in drug development that focuses on rediscovering new uses for old therapeutic agents. Drug repositioning can be defined more precisely as the process of exploring new indications for an already approved drug while drug repurposing includes overall re-development approaches grounded in the identical chemical structure of the active drug moiety as in the original product. The repositioning approach accelerates the drug development process, curtails the cost and risk inherent to drug development. The strategy focuses on the polypharmacology of drugs to unlocks novel opportunities for logically designing more efficient therapeutic agents for unmet medical disorders. Drug repositioning also expresses certain regulatory challenges that hamper its further utilization. The review outlines the eminent role of drug repositioning in new drug discovery, methods to predict the molecular targets of a drug molecule, advantages that the strategy offers to the pharmaceutical industries, explaining how the industrial collaborations with academics can assist in the discovering more repositioning opportunities. The focus of the review is to highlight the latest applications of drug repositioning in various disorders. The review also includes a comparison of old and new therapeutic uses of repurposed drugs, assessing their novel mechanisms of action and pharmacological effects in the management of various disorders. Various restrictions and challenges that repurposed drugs come across during their development and regulatory phases are also highlighted.
Collapse
Affiliation(s)
- Supriya Roy
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Suneela Dhaneshwar
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, India
| | - Bhavya Bhasin
- Poona College of Pharmacy, Bharati Vidyapeeth University, Pune, India
| |
Collapse
|
128
|
Lem FF, Opook F, Lee DJH, Chee FT, Lawson FP, Chin SN. Molecular Mechanism of Action of Repurposed Drugs and Traditional Chinese Medicine Used for the Treatment of Patients Infected With COVID-19: A Systematic Scoping Review. Front Pharmacol 2021; 11:585331. [PMID: 33746739 PMCID: PMC7970521 DOI: 10.3389/fphar.2020.585331] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 12/21/2020] [Indexed: 12/15/2022] Open
Abstract
Background: The emergence of COVID-19 as a pandemic has resulted in the need for urgent development of vaccines and drugs and the conduction of clinical trials to fight the outbreak. Because of the time constraints associated with the development of vaccines and effective drugs, drug repurposing and other alternative treatment methods have been used to treat patients that have been infected by the SARS-CoV-2 virus and have acquired COVID-19. Objective: The objective of this systematic scoping review is to provide an overview of the molecular mechanism of action of repurposed drugs or alternative treatment medicines used to attenuate COVID-19 disease. Method: The research articles or gray literature, including theses, government reports, and official news online, were identified from four databases and one search engine. The full content of a total of 160 articles that fulfilled our inclusion criteria was analyzed and information about six drugs (ritonavir, lopinavir, oseltamivir, remdesivir, favipiravir, and chloroquine) and four Traditional Chinese Medicines (Shuang Huang Lian Kou Fu Ye, TCM combination of Bu Huan Jin Zheng Qi San and Da Yuan Yin, Xue Bi Jing Injection, and Qing Fei Pai Du Tang) was extracted. Results: All of the repurposed drugs and complementary medicine that have been used for the treatment of COVID-19 depend on the ability of the drug to inhibit the proliferation of the SARS-CoV-2 virus by binding to enzyme active sites, viral chain termination, or triggering of the molecular pathway, whereas Traditional Chinese Medicine plays a pivotal role in triggering the inflammation pathway, such as the neuraminidase blocker, to fight the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Fui Fui Lem
- Clinical Research Centre, Hospital Queen Elizabeth, Ministry of Health Malaysia, Kota Kinabalu, Malaysia
| | - Fernandes Opook
- Wildlife Health, Genetic and Forensic Laboratory, Kota Kinabalu, Malaysia
| | | | - Fong Tyng Chee
- Faculty of Sustainable Agriculture, Universiti Malaysia Sabah, Sandakan, Malaysia
| | - Fahcina P. Lawson
- School of Medicine, The Johns Hopkins University, Baltimore, MD, United States
| | - Su Na Chin
- Faculty of Science and Natural Resources, Universiti Malaysia Sabah, Kota Kinabalu, Malaysia
| |
Collapse
|
129
|
Cortés H, Reyes-Hernández OD, Alcalá-Alcalá S, Bernal-Chávez SA, Caballero-Florán IH, González-Torres M, Sharifi-Rad J, González-Del Carmen M, Figueroa-González G, Leyva-Gómez G. Repurposing of Drug Candidates for Treatment of Skin Cancer. Front Oncol 2021; 10:605714. [PMID: 33489912 PMCID: PMC7821387 DOI: 10.3389/fonc.2020.605714] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 11/27/2020] [Indexed: 12/24/2022] Open
Abstract
Skin cancers are highly prevalent malignancies that affect millions of people worldwide. These include melanomas and nonmelanoma skin cancers. Melanomas are among the most dangerous cancers, while nonmelanoma skin cancers generally exhibit a more benign clinical pattern; however, they may sometimes be aggressive and metastatic. Melanomas typically appear in body regions exposed to the sun, although they may also appear in areas that do not usually get sun exposure. Thus, their development is multifactorial, comprising endogenous and exogenous risk factors. The management of skin cancer depends on the type; it is usually based on surgery, chemotherapy, immunotherapy, and targeted therapy. In this respect, oncological treatments have demonstrated some progress in the last years; however, current therapies still present various disadvantages such as little cell specificity, recurrent relapses, high toxicity, and increased costs. Furthermore, the pursuit of novel medications is expensive, and the authorization for their clinical utilization may take 10-15 years. Thus, repositioning of drugs previously approved and utilized for other diseases has emerged as an excellent alternative. In this mini-review, we aimed to provide an updated overview of drugs' repurposing to treat skin cancer and discuss future perspectives.
Collapse
Affiliation(s)
- Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Octavio D. Reyes-Hernández
- Laboratorio de Biología Molecular del Cáncer, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Sergio Alcalá-Alcalá
- Facultad de Farmacia, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Sergio A. Bernal-Chávez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Isaac H. Caballero-Florán
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Maykel González-Torres
- CONACyT-Laboratorio de Biotecnología, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de México, Mexico
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Facultad de Medicina, Universidad del Azuay, Cuenca, Ecuador
| | | | - Gabriela Figueroa-González
- Laboratorio de Farmacogenética, UMIEZ, Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
130
|
Su Q, Li T, He PF, Lu XC, Yu Q, Gao QC, Wang ZJ, Wu MN, Yang D, Qi JS. Trichostatin A ameliorates Alzheimer's disease-related pathology and cognitive deficits by increasing albumin expression and Aβ clearance in APP/PS1 mice. Alzheimers Res Ther 2021; 13:7. [PMID: 33397436 PMCID: PMC7784383 DOI: 10.1186/s13195-020-00746-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/08/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is an intractable neurodegenerative disorder in the elderly population, currently lacking a cure. Trichostatin A (TSA), a histone deacetylase inhibitor, showed some neuroprotective roles, but its pathology-improvement effects in AD are still uncertain, and the underlying mechanisms remain to be elucidated. The present study aims to examine the anti-AD effects of TSA, particularly investigating its underlying cellular and molecular mechanisms. METHODS Novel object recognition and Morris water maze tests were used to evaluate the memory-ameliorating effects of TSA in APP/PS1 transgenic mice. Immunofluorescence, Western blotting, Simoa assay, and transmission electron microscopy were utilized to examine the pathology-improvement effects of TSA. Microglial activity was assessed by Western blotting and transwell migration assay. Protein-protein interactions were analyzed by co-immunoprecipitation and LC-MS/MS. RESULTS TSA treatment not only reduced amyloid β (Aβ) plaques and soluble Aβ oligomers in the brain, but also effectively improved learning and memory behaviors of APP/PS1 mice. In vitro study suggested that the improvement of Aβ pathology by TSA was attributed to the enhancement of Aβ clearance, mainly by the phagocytosis of microglia, and the endocytosis and transport of microvascular endothelial cells. Notably, a meaningful discovery in the study was that TSA dramatically upregulated the expression level of albumin in cell culture, by which TSA inhibited Aβ aggregation and promoted the phagocytosis of Aβ oligomers. CONCLUSIONS These findings provide a new insight into the pathogenesis of AD and suggest TSA as a novel promising candidate for the AD treatment.
Collapse
Affiliation(s)
- Qiang Su
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Tian Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Pei-Feng He
- Institute of Medical Data Sciences and School of Management, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Xue-Chun Lu
- Department of Hematology, the Second Medical Center & National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qi Yu
- Institute of Medical Data Sciences and School of Management, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qi-Chao Gao
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Dan Yang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
131
|
Zheng W, D’Aiuto L, Demers MJ, Muralidaran V, Wood JA, Wesesky M, Chattopadhyay A, Nimgaonkar VL. Insights into bioinformatic approaches for repurposing compounds as anti-viral drugs. Antivir Chem Chemother 2021; 29:20402066211036822. [PMID: 34463534 PMCID: PMC8411619 DOI: 10.1177/20402066211036822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Drug repurposing is a cost-effective strategy to identify drugs with novel effects. We searched for drugs exhibiting inhibitory activity to Herpes Simplex virus 1 (HSV-1). Our strategy utilized gene expression data generated from HSV-1-infected cell cultures which was paired with drug effects on gene expression. Gene expression data from HSV-1 infected and uninfected neurons were analyzed using BaseSpace Correlation Engine (Illumina®). Based on the general Signature Reversing Principle (SRP), we hypothesized that the effects of candidate antiviral drugs on gene expression would be diametrically opposite (negatively correlated) to those effects induced by HSV-1 infection. RESULTS We initially identified compounds capable of inducing changes in gene expression opposite to those which were consequent to HSV-1 infection. The most promising negatively correlated drugs (Valproic acid, Vorinostat) did not significantly inhibit HSV-1 infection further in African green monkey kidney epithelial cells (Vero cells). Next, we tested Sulforaphane and Menadione which showed effects similar to those caused by viral infections (positively correlated). Intriguingly, Sulforaphane caused a modest but significant inhibition of HSV-1 infection in Vero cells (IC50 = 180.4 µM, p = 0.008), but exhibited toxicity when further explored in human neuronal progenitor cells (NPCs) derived from induced pluripotent stem cells. CONCLUSIONS These results reveal the limits of the commonly used SRP strategy when applied to the identification of novel antiviral drugs and highlight the necessity to refine the SRP strategy to increase its utility.
Collapse
Affiliation(s)
- Wenxiao Zheng
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Third Xiangya Hospital, Xiangya School of Medicine, Central
South University, Changsha, China
- Department of Psychiatry, and National Clinical Research Center
for Mental Disorders, The Second Xiangya Hospital, Central South University,
Changsha, China
| | - Leonardo D’Aiuto
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Matthew J Demers
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Vaishali Muralidaran
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Joel A Wood
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Maribeth Wesesky
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Ansuman Chattopadhyay
- Molecular Biology Information Service, Health Sciences Library
System, University of
Pittsburgh, University of Pittsburgh, Pittsburgh, USA
| | - Vishwajit L Nimgaonkar
- Department of Psychiatry, Western Psychiatric Institute and
Clinic, University of Pittsburgh School of Medicine, Pittsburgh, USA
- Department of Human Genetics, Graduate School of Public Health,
University of Pittsburgh, Pittsburgh, USA
- Behavioral Health Service Line, Veterans Administration
Pittsburgh Healthcare System, Pittsburgh, USA
| |
Collapse
|
132
|
Bellera CL, Llanos M, Gantner ME, Rodriguez S, Gavernet L, Comini M, Talevi A. Can drug repurposing strategies be the solution to the COVID-19 crisis? Expert Opin Drug Discov 2020; 16:605-612. [PMID: 33345645 DOI: 10.1080/17460441.2021.1863943] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction: The COVID-19 pandemic resulted in disastrous human and economic costs, mainly due to the initial lack of specific treatments. Complementary to immunotherapies, drug repurposing is possibly the best option to arrive at COVID-19 treatments in the short term.Areas covered: Repurposing prospects undergoing clinical trials or with some level of evidence emerging from clinical studies are overviewed. The authors discuss some possible intellectual property and commercial barriers to drug repurposing, and strategies to facilitate equitable access to incoming therapeutic solutions, highlighting the importance of collaborative drug discovery models. Based on a critical analysis of the available literature about in silico screens against SARS-CoV-2 main protease, the authors illustrate how frequently overconfident conclusions are being drawn in COVID-19-related literature.Expert opinion: Most of the current clinical trials on potential COVID-19 treatments are, in fact, drug repurposing examples. In October 2020, the FDA approved a repurposed antiviral, remdesivir, as the first treatment for COVID-19. Considering the high expectations invested in approaching therapeutic solutions, the scientific community must be careful not to raise unrealistic expectations. Today more than ever, the conclusions drawn in scientific reports have to be fully supported by the level of evidence, avoiding any sort of unfounded speculation.
Collapse
Affiliation(s)
- Carolina L Bellera
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina.,Argentinean National Council of Scientific and Technical Research (CONICET), Argentina
| | - Manuel Llanos
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina.,Argentinean National Council of Scientific and Technical Research (CONICET), Argentina
| | - Melisa E Gantner
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina
| | - Santiago Rodriguez
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina.,Argentinean National Council of Scientific and Technical Research (CONICET), Argentina
| | - Luciana Gavernet
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina.,Argentinean National Council of Scientific and Technical Research (CONICET), Argentina
| | - Marcelo Comini
- Group Redox Biology of Trypanosomes, Institut Pasteur De Montevideo, Montevideo, Uruguay
| | - Alan Talevi
- Laboratory of Bioactive Research and Development (Lideb), Department of Biological Sciences, Faculty of Exact Sciences, Universidad Nacional De La Plata (UNLP), Buenos Aires, Argentina.,Argentinean National Council of Scientific and Technical Research (CONICET), Argentina
| |
Collapse
|
133
|
Pratama MRF, Poerwono H, Siswodihardjo S. Molecular Docking of Novel 5-O-benzoylpinostrobin Derivatives as SARS-CoV-2 Main Protease Inhibitors. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2020.57] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Mohammad Rizki Fadhil Pratama
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Universitas Airlangga, Jl Dr Ir H Soekarno Mulyorejo, Surabaya, East Java, Indonesia
| | - Hadi Poerwono
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Airlangga, Jl Dr Ir H Soekarno Mulyorejo, Surabaya, East Java, Indonesia
| | - Siswandono Siswodihardjo
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Universitas Airlangga, Jl Dr Ir H Soekarno Mulyorejo, Surabaya, East Java, Indonesia
| |
Collapse
|
134
|
Juárez-López D, Schcolnik-Cabrera A. Drug Repurposing: Considerations to Surpass While Re-directing Old Compounds for New Treatments. Arch Med Res 2020; 52:243-251. [PMID: 33190955 DOI: 10.1016/j.arcmed.2020.10.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 10/21/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
Drug repurposing has increased in recent years as an attractive option for treating a number of diseases. Compared to those brought forward via traditional chemical development, drugs intended for repurposing can enter the market faster and with lower investment from pharmaceutical companies. However, a common trend is to focus on diseases that yield higher returns to the industry, such as cancer and common metabolic and inflammatory conditions, resulting in orphan illnesses and neglected tropical diseases having fewer repurposing options for affected patients. In addition, certain legal concerns, including limited patent coverage for the repurposed drugs and pharmacological challenges in performing clinical trials, reduce the likelihood of success. In this review, we discuss the most important concerns that affect the pathway of drug repurposing, with special emphasis on the economic revenues, government-industry associations, and legal considerations that together impact the pharmaceutical industry's decision-making on which compounds may be eligible for repurposing.
Collapse
Affiliation(s)
- Daniel Juárez-López
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alejandro Schcolnik-Cabrera
- Département de Biochimie et Médecine Moléculaire, Université de Montréal, Succursale Centre-Ville, Montréal, QC, Canada; Maisonneuve-Rosemont Hospital Research Centre, Montréal, QC, Canada.
| |
Collapse
|
135
|
Kumar N, Gahlawat A, Kumar RN, Singh YP, Modi G, Garg P. Drug repurposing for Alzheimer’s disease: in silico and in vitro investigation of FDA-approved drugs as acetylcholinesterase inhibitors. J Biomol Struct Dyn 2020; 40:2878-2892. [DOI: 10.1080/07391102.2020.1844054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Navneet Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, India
| | - Anuj Gahlawat
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, India
| | - Rajaram Naresh Kumar
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, India
| | - Yash Pal Singh
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Gyan Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, India
| | - Prabha Garg
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, India
| |
Collapse
|
136
|
Babayeva M, Loewy Z. Repurposing Drugs for COVID-19: Pharmacokinetics and Pharmacogenomics of Chloroquine and Hydroxychloroquine. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2020; 13:531-542. [PMID: 33122936 PMCID: PMC7591012 DOI: 10.2147/pgpm.s275964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022]
Abstract
Background A new coronavirus SARS-CoV-2 has been identified as the etiological agent of the severe acute respiratory syndrome, COVID-19, the source and cause of the 2019–20 coronavirus pandemic. Hydroxychloroquine and chloroquine have gathered extraordinary attention as therapeutic candidates against SARS-CoV-2 infections. While there is growing scientific data on the therapeutic effect, there is also concern for toxicity of the medications. The therapy of COVID-19 by hydroxychloroquine and chloroquine is off-label. Studies to analyze the personalized effect and safety are lacking. Methods A review of the literature was performed using Medline/PubMed/Embase database. A variety of keywords were employed in keyword/title/abstract searches. The electronic search was followed by extensive hand searching using reference lists from the identified articles. Results A total of 126 results were obtained after screening all sources. Mechanisms underlying variability in drug concentrations and therapeutic response with chloroquine and hydroxychloroquine in mediating beneficial and adverse effects of chloroquine and hydroxychloroquine were reviewed and analyzed. Pharmacogenomic studies from various disease states were evaluated to elucidate the role of genetic variation in drug response and toxicity. Conclusion Knowledge of the pharmacokinetics and pharmacogenomics of chloroquine and hydroxychloroquine is necessary for effective and safe dosing and to avoid treatment failure and severe complications.
Collapse
Affiliation(s)
| | - Zvi Loewy
- Touro College of Pharmacy, New York, NY, USA.,New York Medical College, Valhalla, NY, USA
| |
Collapse
|
137
|
Parvathaneni V, Gupta V. Utilizing drug repurposing against COVID-19 - Efficacy, limitations, and challenges. Life Sci 2020; 259:118275. [PMID: 32818545 PMCID: PMC7430345 DOI: 10.1016/j.lfs.2020.118275] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/10/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023]
Abstract
The recent outbreak of Coronavirus disease (COVID-19), first in Eastern Asia and then essentially across the world has been declared a pandemic by the WHO. COVID-19 is caused by a novel virus SARS-CoV2 (2019-nCoV), against which there is currently no vaccine available; and current antiviral therapies have failed, causing a very high mortality rate. Drug repurposing i.e. utilizing an approved drug for different indication, offers a time- and cost-efficient alternative for making new therapies available to patients. Although there are several reports presenting novel approaches to treat COVID-19, still an attentive review of previous scientific literature is essential to overcome their failure to exhibit efficacy. There is an urgent need to provide a comprehensive outlook toward utilizing drug repurposing as a tool for discovery of new therapies against COVID-19. In this article, we aim to provide a to-the-point review of current literature regarding efficacy of repurposed drugs against COVID-19 and other respiratory infections caused by coronaviruses. We have briefly discussed COVID-19 epidemiology, and then have discussed drug repurposing approaches and examples, specific to respiratory viruses. Limitations of utilization of repurposed drug molecules such as dosage regimen and associated challenges such as localized delivery in respiratory tract have also been discussed in detail.
Collapse
Affiliation(s)
- Vineela Parvathaneni
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Vivek Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
138
|
Abstract
The pandemic of coronavirus infection 2019 (COVID-19) due to the serious respiratory condition created by the coronavirus 2 (SARS-CoV-2) presents a challenge to recognize effective strategies for management and treatment. In general, COVID-19 is an acute disease that can also be fatal, with an ongoing 10.2% case morbidity rate. Extreme illness may bring about death because of enormous alveolar damage and hemorrhage along with progressive respiratory failure. The rapidly expanding information with respect to SARS-CoV-2 research suggests a substantial number of potential drug targets. The most encouraging treatment to date is suggested to be with the help of remdesivir, hydroxychloroquine, and many such repurposed drugs. Remdesivir has a strong in vitro activity for SARS-CoV-2, yet it is not the drug of choice as affirmed by the US Food and Drug Administration and presently is being tried in progressing randomized preliminaries. The COVID-19 pandemic has been the worst worldwide general health emergency of this age and, possibly, since the pandemic influenza outbreak of 1918. The speed and volume of clinical preliminaries propelled to examine potential treatments for COVID-19 feature both the need and capacity to create abundant evidence even in the center of a pandemic. No treatments have been demonstrated as accurate and dependable to date. This review presents a concise precise of the targets and broad treatment strategies for the benefit of researchers.
Collapse
|
139
|
Usha T, Middha SK, Kukanur AA, Shravani RV, Anupama MN, Harshitha N, Rahamath A, Kukanuri SA, Goyal AK. Drug Repurposing Approaches: Existing Leads For Novel Threats And Drug Targets. Curr Protein Pept Sci 2020; 22:CPPS-EPUB-110124. [PMID: 32957901 DOI: 10.2174/1389203721666200921152853] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 07/29/2020] [Accepted: 08/07/2020] [Indexed: 11/22/2022]
Abstract
Drug Repurposing (DR) is an alternative to the traditional drug discovery process. It is cost and time effective, with high returns and low risk process that can tackle the increasing need for interventions for varied diseases and new outbreaks. Repurposing of old drugs for other diseases has gained a wider attention, as there have been several old drugs approved by FDA for new diseases. In the global emergency of COVID19 pandemic, this is one of the strategies implemented in repurposing of old anti-infective, anti-rheumatic and anti-thrombotic drugs. The goal of the current review is to elaborate the process of DR, its advantages, repurposed drugs for a plethora of disorders, and the evolution of related academic publications. Further, detailed are the computational approaches: literature mining and semantic inference, network-based drug repositioning, signature matching, retrospective clinical analysis, molecular docking and experimental phenotypic screening. We discuss the legal and economical potential barriers in DR, existent collaborative models and recommendations for overcoming these hurdles and leveraging the complete potential of DR in finding new indications.
Collapse
Affiliation(s)
- Talambedu Usha
- Department of Biochemistry, Bangalore University, Bengaluru, Karnataka. India
| | - Sushil K Middha
- DBT-BIF Centre, Department of Biotechnology, Maharani Lakshmi Ammanni College for Women(mLAC), Bengaluru, Karnataka. India
| | | | | | | | | | - Ameena Rahamath
- Department of Biochemistry, mLAC, Bengaluru, Karnataka. India
| | | | - Arvind K Goyal
- Department of Biotechnology, Bodoland University, Kokrajhar783370, BTAD, Assam. India
| |
Collapse
|
140
|
Pandey A, Nikam AN, Shreya AB, Mutalik SP, Gopalan D, Kulkarni S, Padya BS, Fernandes G, Mutalik S, Prassl R. Potential therapeutic targets for combating SARS-CoV-2: Drug repurposing, clinical trials and recent advancements. Life Sci 2020; 256:117883. [PMID: 32497632 PMCID: PMC7263255 DOI: 10.1016/j.lfs.2020.117883] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/31/2022]
Abstract
The present pandemic of SARS-CoV-2 has been a tough task for the whole world to deal with. With the absence of specific drugs or vaccines against SARS-CoV-2, the situation is very difficult to control. Apart from the absence of specific therapies, the lack of knowledge about potential therapeutic targets and individual perception is adding to the complications. The present review describes the novel SARS-CoV-2 structure, surface proteins, asymptomatic and symptomatic transmission in addition to the genotype and phenotype of SARS-CoV-2 along with genetic strains and similarity between SARS, MERS and SARS-CoV-2. Therapeutic strategies such as inhibition of the endocytic pathway and suppressing RNA polymerase activity by metal ions, which could be quite beneficial for controlling COVID-19, are outlined. The drug repurposing for SARS-CoV-2 is discussed in detail along with therapeutic classes such as antivirals, antibiotics, and amino quinolones and their probable role in suppressing SARS-CoV-2 with reference to case studies. The ongoing clinical trials both with respect to drug repurposing and vaccines are summarized along with a brief description. The recent advancements and future perspective of ongoing research for therapy and detection of SARS-CoV-2 are provided. The review, in brief, summarizes epidemiology, therapy and the current scenario for combating SARS-CoV-2.
Collapse
Affiliation(s)
- Abhjieet Pandey
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India; Gottfried Schatz Research Centre for Cell Signalling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Ajinkya Nitin Nikam
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Ajjappla Basavaraj Shreya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Sadhana P Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Divya Gopalan
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Sanjay Kulkarni
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Bharath Singh Padya
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Gasper Fernandes
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576106, Karnataka State, India.
| | - Ruth Prassl
- Gottfried Schatz Research Centre for Cell Signalling, Metabolism and Aging, Medical University of Graz, Graz, Austria..
| |
Collapse
|
141
|
Cardoso WB, Mendanha SA. Molecular dynamics simulation of docking structures of SARS-CoV-2 main protease and HIV protease inhibitors. J Mol Struct 2020; 1225:129143. [PMID: 32863430 PMCID: PMC7443253 DOI: 10.1016/j.molstruc.2020.129143] [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: 07/29/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022]
Abstract
We consider possible repurposed-drugs candidates against SARS-CoV-2. 10 different HIV protease inhibitors were investigated. In silico simulations were used to study protease inhibitors for SARS-CoV-2.
In this paper we investigate 10 different HIV protease inhibitors (HPIs) as possible repurposed-drugs candidates against SARS-CoV-2. To this end, we execute molecular docking and molecular dynamics simulations. The in silico data demonstrated that, despite their molecular differences, all HPIs presented a similar behavior for the parameters analyzed, with the exception of Nelfinavir that showed better results for most of the molecular dynamics parameters in comparison with the N3 inhibitor.
Collapse
Affiliation(s)
- Wesley B Cardoso
- Instituto de Física, Universidade Federal de Goiás, 74.690-900, Goiânia, Goiás, Brazil
| | - Sebastião A Mendanha
- Instituto de Física, Universidade Federal de Goiás, 74.690-900, Goiânia, Goiás, Brazil
| |
Collapse
|
142
|
Screening Repurposing Libraries for Identification of Drugs with Novel Antifungal Activity. Antimicrob Agents Chemother 2020; 64:AAC.00924-20. [PMID: 32660991 DOI: 10.1128/aac.00924-20] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Fungal organisms are ubiquitous in nature, and progress of modern medicine is creating an expanding number of severely compromised patients susceptible to a variety of opportunistic fungal infections. These infections are difficult to diagnose and treat, leading to high mortality rates. The limited antifungal arsenal, the toxicity of current antifungal drugs, the development of resistance, and the emergence of new multidrug-resistant fungi, all highlight the urgent need for new antifungal agents. Unfortunately, the development of a novel antifungal is a rather long and expensive proposition, and no new classes of antifungal agents have reached the market in the last 2 decades. Drug repurposing, or finding new indications for old drugs, represents a promising alternative pathway to drug development that is particularly appealing within the academic environment. In the last few years, there has been a growing interest in repurposing approaches in the antifungal arena, with multiple groups of investigators having performed screenings of different repurposing libraries against different pathogenic fungi in search for drugs with previously unrecognized antifungal effects. Overall, these repurposing efforts may lead to the fast deployment of drugs with novel antifungal activity, which can rapidly bring benefits to patients, while at the same time reducing health care costs.
Collapse
|
143
|
Pérez-Plasencia C, López-Urrutia E, García-Castillo V, Trujano-Camacho S, López-Camarillo C, Campos-Parra AD. Interplay Between Autophagy and Wnt/β-Catenin Signaling in Cancer: Therapeutic Potential Through Drug Repositioning. Front Oncol 2020; 10:1037. [PMID: 33014767 PMCID: PMC7461967 DOI: 10.3389/fonc.2020.01037] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 05/26/2020] [Indexed: 12/20/2022] Open
Abstract
The widespread dysregulation that characterizes cancer cells has been dissected and many regulation pathways common to multiple cancer types have been described in depth. Wnt/β-catenin signaling and autophagy are among these principal pathways, which contribute to tumor growth and resistance to anticancer therapies. Currently, several therapeutic strategies that target either Wnt/β-catenin signaling or autophagy are in various stages of development. Targeted therapies that block specific elements that participate in both pathways; are subject to in vitro studies as well as pre-clinical and early clinical trials. Strikingly, drugs designed for other diseases also impact these pathways, which is relevant since they are already FDA-approved and sometimes even routinely used in the clinic. The main focus of this mini-review is to highlight the importance of drug repositioning to inhibit the Wnt/β-catenin and autophagy pathways, with an emphasis on the interplay between them. The data we found strongly suggested that this field is worth further examination.
Collapse
Affiliation(s)
- Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Eduardo López-Urrutia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Mexico
| | - Verónica García-Castillo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla, Mexico
| | - Samuel Trujano-Camacho
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Mexico City, Mexico
| | - Alma D. Campos-Parra
- Laboratorio de Genómica, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| |
Collapse
|
144
|
Computational Drug Repositioning: Current Progress and Challenges. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10155076] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Novel drug discovery is time-consuming, costly, and a high-investment process due to the high attrition rate. Therefore, many trials are conducted to reuse existing drugs to treat pressing conditions and diseases, since their safety profiles and pharmacokinetics are already available. Drug repositioning is a strategy to identify a new indication of existing or already approved drugs, beyond the scope of their original use. Various computational and experimental approaches to incorporate available resources have been suggested for gaining a better understanding of disease mechanisms and the identification of repurposed drug candidates for personalized pharmacotherapy. In this review, we introduce publicly available databases for drug repositioning and summarize the approaches taken for drug repositioning. We also highlight and compare their characteristics and challenges, which should be addressed for the future realization of drug repositioning.
Collapse
|
145
|
Ahmad J, Ikram S, Ahmad F, Rehman IU, Mushtaq M. SARS-CoV-2 RNA Dependent RNA polymerase (RdRp) - A drug repurposing study. Heliyon 2020; 6:e04502. [PMID: 32754651 PMCID: PMC7377705 DOI: 10.1016/j.heliyon.2020.e04502] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/18/2020] [Accepted: 07/15/2020] [Indexed: 12/19/2022] Open
Abstract
The outbreak of SARS-CoV-2 in December 2019 in China subsequently lead to a pandemic. Lack of vaccine and specific anti-viral drugs started a global health disaster. For a sustained control and protection, development of potential anti-viral drugs is one of the targeted approach. Although, designing and developing a panel of new drugs molecules are always encouraged. However, in the current emergency, drug repurposing study is one of the most effective and fast track option. The crystal structure of a SARS-CoV-2 (Severe acute respiratory syndrome coronavirus 2) RNA Dependent RNA Polymerase (RdRp) has recently been deciphered through X-ray crystallography. The single-chain of core RNA Dependent RNA Polymerase relies on virus-encoded cofactors nsp7 and two units of nsp8 for its optimum function. This study explored the FDA approved database of 7922 molecules and screened against the core polymerase along with cofactors. Here we report a panel of FDA approved drugs that show substantial interactions with key amino acid residues of the active site. Interestingly, some of the identified drugs (Ornipressin, Lypressin, Examorelin, Polymyxin B1) bind strongly within the binding pockets of both forms of RdRp. Besides, we found strong candidates for the complex form as well which include Nacortocin, Cistinexine, Cisatracurium (among others). These drugs have the potential to be considered while contriving therapeutic options.
Collapse
Affiliation(s)
- Jamshaid Ahmad
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar, KP, Pakistan
| | - Saima Ikram
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar, KP, Pakistan
| | - Fawad Ahmad
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar, KP, Pakistan
| | - Irshad Ur Rehman
- Centre of Biotechnology and Microbiology, University of Peshawar, Peshawar, KP, Pakistan
| | | |
Collapse
|
146
|
Abstract
The current global pandemic COVID-19 caused by the SARS-CoV-2 virus has already inflicted insurmountable damage both to the human lives and global economy. There is an immediate need for identification of effective drugs to contain the disastrous virus outbreak. Global efforts are already underway at a war footing to identify the best drug combination to address the disease. In this review, an attempt has been made to understand the SARS-CoV-2 life cycle, and based on this information potential druggable targets against SARS-CoV-2 are summarized. Also, the strategies for ongoing and future drug discovery against the SARS-CoV-2 virus are outlined. Given the urgency to find a definitive cure, ongoing drug repurposing efforts being carried out by various organizations are also described. The unprecedented crisis requires extraordinary efforts from the scientific community to effectively address the issue and prevent further loss of human lives and health.
Collapse
Affiliation(s)
- Ambrish Saxena
- Indian Institute of Technology Tirupati, Tirupati, India
| |
Collapse
|
147
|
Gupta A, Kumar S, Kumar R, Choudhary AK, Kumari K, Singh P, Kumar V. COVID-19: Emergence of Infectious Diseases, Nanotechnology Aspects, Challenges, and Future Perspectives. ChemistrySelect 2020; 5:7521-7533. [PMID: 32835089 PMCID: PMC7361534 DOI: 10.1002/slct.202001709] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/22/2020] [Indexed: 01/08/2023]
Abstract
Wuhan, a city of China, is the epicenter for the pandemic outbreak of coronavirus disease-2019 (COVID-19). It has become a severe public health challenge to the world and established a public health emergency of international worry. This infectious disease has pulled down the economy of almost all top developed nations. The coronaviruses (CoVs) known for various epidemics caused time to time. Infectious diseases such as severe acute respiratory syndrome (SARS) and middle east respiratory syndrome (MERS), followed by COVID-19, are all coronaviruses led outbreaks that scourged the history of mankind. CoVs evolved themselves to more infectious, transmissible, and more pandemic with time. To prevent the spread of the SARS-CoV-2, many countries have ordered the complete lockdown to combat the outbreak. This paper briefly discussed the historical background of CoVs and the evolution of human coronaviruses (HCoVs), the case studies and the development of their antiviral medications. The viral infection encountered with present-day challenges and futuristic approaches with the help of nanotechnology to minimize the spread of infectious viruses. The antiviral drugs and their clinical advances, along with herbal medicines for viral inhibition and immunity boosters, are described. Elaboration of tables related to CoVs for the compilation of the literature has been adopted for the better understanding.
Collapse
Affiliation(s)
- Akanksha Gupta
- Department of ChemistrySri Venkateswara CollegeUniversity of DelhiIndia.
| | - Sanjay Kumar
- Department of ChemistryDeshbandhu CollegeUniversity of DelhiIndia.
| | - Ravinder Kumar
- Department of Chemistry, Gurukula Kangri VishwavidyalayaHaridwarIndia.
| | | | - Kamlesh Kumari
- Department of ZoologyDeen Dayal Upadhyaya CollegeDelhiIndia.
| | - Prashant Singh
- Department of ChemistryAtma Ram Sanatan Dharma CollegeDelhi UniversityNew DelhiIndia.
| | - Vinod Kumar
- Department of ChemistryKirori Mal CollegeUniversity of DelhiIndia
- Special Centre for Nano SciencesJawaharlal Nehru UniversityDelhiIndia
| |
Collapse
|
148
|
Abdulla A, Wang B, Qian F, Kee T, Blasiak A, Ong YH, Hooi L, Parekh F, Soriano R, Olinger GG, Keppo J, Hardesty CL, Chow EK, Ho D, Ding X. Project IDentif.AI: Harnessing Artificial Intelligence to Rapidly Optimize Combination Therapy Development for Infectious Disease Intervention. ADVANCED THERAPEUTICS 2020; 3:2000034. [PMID: 32838027 PMCID: PMC7235487 DOI: 10.1002/adtp.202000034] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Indexed: 12/24/2022]
Abstract
In 2019/2020, the emergence of coronavirus disease 2019 (COVID-19) resulted in rapid increases in infection rates as well as patient mortality. Treatment options addressing COVID-19 included drug repurposing, investigational therapies such as remdesivir, and vaccine development. Combination therapy based on drug repurposing is among the most widely pursued of these efforts. Multi-drug regimens are traditionally designed by selecting drugs based on their mechanism of action. This is followed by dose-finding to achieve drug synergy. This approach is widely-used for drug development and repurposing. Realizing synergistic combinations, however, is a substantially different outcome compared to globally optimizing combination therapy, which realizes the best possible treatment outcome by a set of candidate therapies and doses toward a disease indication. To address this challenge, the results of Project IDentif.AI (Identifying Infectious Disease Combination Therapy with Artificial Intelligence) are reported. An AI-based platform is used to interrogate a massive 12 drug/dose parameter space, rapidly identifying actionable combination therapies that optimally inhibit A549 lung cell infection by vesicular stomatitis virus within three days of project start. Importantly, a sevenfold difference in efficacy is observed between the top-ranked combination being optimally and sub-optimally dosed, demonstrating the critical importance of ideal drug and dose identification. This platform is disease indication and disease mechanism-agnostic, and potentially applicable to the systematic N-of-1 and population-wide design of highly efficacious and tolerable clinical regimens. This work also discusses key factors ranging from healthcare economics to global health policy that may serve to drive the broader deployment of this platform to address COVID-19 and future pandemics.
Collapse
Affiliation(s)
- Aynur Abdulla
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Boqian Wang
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Feng Qian
- Ministry of Education Key Laboratory of Contemporary AnthropologyHuman Phenome InstituteSchool of Life SciencesFudan UniversityShanghai200438China
| | - Theodore Kee
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
| | - Agata Blasiak
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
| | - Yoong Hun Ong
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
| | - Lissa Hooi
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore117599Singapore
| | | | | | - Gene G. Olinger
- Global Health Surveillance and Diagnostic DivisionMRIGlobalGaithersburgMD20878USA
- Boston University School of MedicineDivision of Infectious DiseasesBostonMA02118USA
| | - Jussi Keppo
- NUS Business School and Institute of Operations Research and AnalyticsNational University of SingaporeSingapore119245Singapore
| | - Chris L. Hardesty
- KPMG Global Health and Life Sciences Centre of ExcellenceSingapore048581Singapore
| | - Edward K. Chow
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- Cancer Science Institute of SingaporeNational University of SingaporeSingapore117599Singapore
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore117600Singapore
| | - Dean Ho
- The N.1 Institute for Health (N.1)National University of SingaporeSingapore117456Singapore
- The Institute for Digital Medicine (WisDM)Yong Loo Lin School of MedicineNational University of SingaporeSingapore11756Singapore
- Department of Biomedical EngineeringNUS EngineeringNational University of SingaporeSingapore117583Singapore
- Department of PharmacologyYong Loo Lin School of MedicineNational University of SingaporeSingapore117600Singapore
| | - Xianting Ding
- Institute for Personalized MedicineSchool of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| |
Collapse
|
149
|
Buonaguro L, Buonaguro FM. Knowledge-based repositioning of the anti-HCV direct antiviral agent Sofosbuvir as SARS-CoV-2 treatment. Infect Agent Cancer 2020; 15:32. [PMID: 32419838 PMCID: PMC7215134 DOI: 10.1186/s13027-020-00302-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 05/05/2020] [Indexed: 01/21/2023] Open
Abstract
The new human coronavirus named SARS-CoV-2 is a positive-sense RNA virus for which no specific drugs are currently available. A knowledge-based analysis strongly suggests a possible repositioning of the anti-HCV direct antiviral agent (DAA) Sofosbuvir as treatment for SARS-CoV-2. Indeed, the RNA-dependent RNA-polymerases (RdRp) of the two viruses show high sequence and structural homology, supporting the likelihood of binding the Sofosbuvir molecule with similar efficiency. Such a repositioning would allow the containment of the SARS-CoV-2 pandemic and limit the progression of disease to potentially deadly COVID19.
Collapse
Affiliation(s)
- Luigi Buonaguro
- Laboratory of Innovative Immunological Models, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola, 1, 80131 Naples, Italy
| | - Franco M. Buonaguro
- Laboratory of Molecular Biology and Viral Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori, “Fondazione Pascale” – IRCCS, Via Mariano Semmola 52, 80131 Naples, Italy
| |
Collapse
|
150
|
Petykó ZI, Inotai A, Holtorf AP, Brixner D, Kaló Z. Barriers and facilitators of exploiting the potential of value-added medicines. Expert Rev Pharmacoecon Outcomes Res 2020; 20:229-236. [PMID: 32321326 DOI: 10.1080/14737167.2020.1758558] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Pharmaceutical research and development (R&D) is costly and only a minority of patients can access innovative medicines due to affordability constraints. Value-added medicines (VAMs) can offer potential benefits at significantly lower R&D costs. AREAS COVERED VAMs may address different health care needs and problems, including off-label use of medicines, poor patient adherence, problems related to polypharmacy, need for home and/or personalized health care services. However, several barriers prevent societies from maximizing the benefits of incremental innovation related to VAMs. Generic manufacturers have limited budget and experience to demonstrate the value of new VAMs. Current market exclusivity options do not efficiently exclude freeridership and do not guarantee a return on investment for VAM innovators. Value propositions of VAMs are limitedly consistent with current HTA frameworks, consequently, incremental innovation is not acknowledged, nor rewarded with differential pricing by payers. Moreover, VAMs are often perceived solely as generic medicines by prescribers. EXPERT OPINION Current practices may need to be reconsidered to exploit the full societal benefit of VAMs, including more efficient policies to guarantee market exclusivity for incremental innovation, acknowledgment of a fair price premium based on a specific value framework and the acceptance of low-cost evidence generation methods.
Collapse
Affiliation(s)
- Zsuzsanna Ida Petykó
- Center for Health Technology Assessment, Semmelweis University , Budapest, Hungary.,Syreon Research Institute , Budapest, Hungary
| | - András Inotai
- Center for Health Technology Assessment, Semmelweis University , Budapest, Hungary.,Syreon Research Institute , Budapest, Hungary
| | | | - Diana Brixner
- Pharmacotherapy Outcomes Research Center, Department of Pharmacotherapy, University of Utah , Salt Lake City, UT, USA
| | - Zoltán Kaló
- Center for Health Technology Assessment, Semmelweis University , Budapest, Hungary.,Syreon Research Institute , Budapest, Hungary
| |
Collapse
|