1
|
Al Khzem AH, Gomaa MS, Alturki MS, Tawfeeq N, Sarafroz M, Alonaizi SM, Al Faran A, Alrumaihi LA, Alansari FA, Alghamdi AA. Drug Repurposing for Cancer Treatment: A Comprehensive Review. Int J Mol Sci 2024; 25:12441. [PMID: 39596504 PMCID: PMC11595001 DOI: 10.3390/ijms252212441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer ranks among the primary contributors to global mortality. In 2022, the global incidence of new cancer cases reached about 20 million, while the number of cancer-related fatalities reached 9.7 million. In Saudi Arabia, there were 13,399 deaths caused by cancer and 28,113 newly diagnosed cases of cancer. Drug repurposing is a drug discovery strategy that has gained special attention and implementation to enhance the process of drug development due to its time- and money-saving effect. It involves repositioning existing medications to new clinical applications. Cancer treatment is a therapeutic area where drug repurposing has shown the most prominent impact. This review presents a compilation of medications that have been repurposed for the treatment of various types of cancers. It describes the initial therapeutic and pharmacological classes of the repurposed drugs and their new applications and mechanisms of action in cancer treatment. The review reports on drugs from various pharmacological classes that have been successfully repurposed for cancer treatment, including approved ones and those in clinical trials and preclinical development. It stratifies drugs based on their anticancer repurpose as multi-type, type-specific, and mechanism-directed, and according to their pharmacological classes. The review also reflects on the future potential that drug repurposing has in the clinical development of novel anticancer therapies.
Collapse
Affiliation(s)
- Abdulaziz H. Al Khzem
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohamed S. Gomaa
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mansour S. Alturki
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Nada Tawfeeq
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Mohammad Sarafroz
- Department of Pharmaceutical Chemistry, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (M.S.A.); (N.T.); (M.S.)
| | - Shareefa M. Alonaizi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Alhassan Al Faran
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Laela Ahmed Alrumaihi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Fatimah Ahmed Alansari
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| | - Abdullah Abbas Alghamdi
- College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Eastern Province, Saudi Arabia; (S.M.A.); (A.A.F.); (L.A.A.); (F.A.A.); (A.A.A.)
| |
Collapse
|
2
|
Anton DB, de Lima JC, Dahmer BR, Camini AM, Goettert MI, Timmers LFSM. Taming the storm: potential anti-inflammatory compounds targeting SARS-CoV-2 MPro. Inflammopharmacology 2024:10.1007/s10787-024-01525-9. [PMID: 39048773 DOI: 10.1007/s10787-024-01525-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/05/2024] [Indexed: 07/27/2024]
Abstract
In severe COVID-19 cases, an exacerbated inflammatory response triggers a cytokine storm that can worsen the prognosis. Compounds with both antiviral and anti-inflammatory activities show promise as candidates for COVID-19 therapy, as they potentially act against the SARS-CoV-2 infection regardless of the disease stage. One of the most attractive drug targets among coronaviruses is the main protease (MPro). This enzyme is crucial for cleaving polyproteins into non-structural proteins required for viral replication. The aim of this review was to identify SARS-CoV-2 MPro inhibitors with both antiviral and anti-inflammatory properties. The interactions of the compounds within the SARS-CoV-2 MPro binding site were analyzed through molecular docking when data from crystallographic structures were unavailable. 18 compounds were selected and classified into five different superclasses. Five of them exhibit high potency against MPro: GC-376, baicalein, naringenin, heparin, and carmofur, with IC50 values below 0.2 μM. The MPro inhibitors selected have the potential to alleviate lung edema and decrease cytokine release. These molecules mainly target three critical inflammatory pathways: NF-κB, JAK/STAT, and MAPK, all previously associated with COVID-19 pathogenesis. The structures of the compounds occupy the S1/S2 substrate binding subsite of the MPro. They interact with residues from the catalytic dyad (His41 and Cys145) and/or with the oxyanion hole (Gly143, Ser144, and Cys145), which are pivotal for substrate recognition. The MPro SARS-CoV-2 inhibitors with potential anti-inflammatory activities present here could be optimized for maximum efficacy and safety and be explored as potential treatment of both mild and severe COVID-19.
Collapse
Affiliation(s)
- Débora Bublitz Anton
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Jeferson Camargo de Lima
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Bruno Rampanelli Dahmer
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Ana Micaela Camini
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil
| | - Marcia Inês Goettert
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmaceutical Sciences, Eberhard Karls Universität Tübingen, 72076, Tübingen, Germany
| | - Luis Fernando Saraiva Macedo Timmers
- Biotechnology Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil.
- Medical Science Graduate Program, Universidade do Vale do Taquari (Univates), Lajeado, CEP 95914-014, Brazil.
| |
Collapse
|
3
|
Oudah KH, Najm MAA, Barghash RF, Kutkat O, GabAllah M, Albohy A, Abouzid KAM. Drug repurposing of pyrazolotriazine derivatives as potential anti-SARS-CoV-2 agents: in vitro and in silico studies. BMC Chem 2024; 18:132. [PMID: 39014447 PMCID: PMC11253567 DOI: 10.1186/s13065-024-01233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
The search for new molecules targeting SARS-CoV-2 has been a priority since 2020. The continuous evolution of new mutants increases the need for more research in the area. One way to find new leads is to repurpose existing drugs and molecules against the required target. Here, we present the in vitro and in silico screening of ten previously synthesized and reported compounds as anti-COVID 19 agents. The compounds were screened in vitro against VERO-E6 cells to find their Cytotoxic Concentration (CC50) and their Inhibitory Concentration (IC50). Compounds 1, 2, and 5 revealed a promising anti-SARS-CoV-2 of (IC50 = 2.4, 11.2 and 2.8 µM), respectively while compounds 3 and 7 showed moderate activity of (IC50 = 17.8 and 26.1 µM) compared to Chloroquine which showed an IC50 of 24.9 µM. Among tested compounds, 1 showed the highest selectivity (CC50/IC50) of 192.8. Docking, molecular dynamics and ADME studies were done to investigate potential interactions between compounds and SARS-CoV-2 targets as well as to study the possibility of using them as lead compounds.
Collapse
Affiliation(s)
- Khulood H Oudah
- Department of Pharmacy, Mazaya University Collage, Nasiriyah, Thi-Qar, Iraq
| | - Mazin A A Najm
- Department of Pharmacy, Mazaya University Collage, Nasiriyah, Thi-Qar, Iraq
| | - Reham F Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Giza, 12622, Egypt
- Faculty of Biotechnology, October University for Modern Science and Arts (MSA University), Giza, Egypt
| | - Omnia Kutkat
- Center of Scientifc Excellence for Infuenza Viruses, National Research Centre, Dokki, Giza, 12622, Egypt
- Department of microbiology, Faculty of pharmacy, Ahram Canadian University, 6 th of October, Giza, 12566, Egypt
| | - Mohamed GabAllah
- Center of Scientifc Excellence for Infuenza Viruses, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Amgad Albohy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, 11837, Cairo, Egypt.
| | - Khaled A M Abouzid
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Abbassia, 11566, Cairo, Egypt.
| |
Collapse
|
4
|
Hussain SS, Libby EF, Lever JEP, Tipper JL, Phillips SE, Mazur M, Li Q, Campos-Gómez J, Harrod KS, Rowe SM. ACE-2 Blockade & TMPRSS2 Inhibition Mitigate SARS-CoV-2 Severity Following Cigarette Smoke Exposure in Airway Epithelial Cells In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.23.600238. [PMID: 38979208 PMCID: PMC11230175 DOI: 10.1101/2024.06.23.600238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cigarette smoking is associated with COVID-19 prevalence and severity, but the mechanistic basis for how smoking alters SARS-CoV-2 pathogenesis is unknown. A potential explanation is that smoking alters the expression of the SARS-CoV-2 cellular receptor and point of entry, angiotensin converting enzyme-2 (ACE-2), and its cofactors including transmembrane protease serine 2 (TMPRSS2). We investigated the impact of cigarette smoking on the expression of ACE-2, TMPRSS2, and other known cofactors of SARS-CoV-2 infection and the resultant effects on infection severity in vitro. Cigarette smoke extract (CSE) exposure increased ACE-2 and TMPRSS2 mRNA expression compared to air control in ferret airway cells, Calu-3 cells, and primary human bronchial epithelial (HBE) cells derived from normal and COPD donors. CSE-exposed ferret airway cells inoculated with SARS-CoV-2 had a significantly higher intracellular viral load versus vehicle-exposed cells. Likewise, CSE-exposure increased both SARS-CoV-2 intracellular viral load and viral replication in both normal and COPD HBE cells over vehicle control. Apoptosis was increased in CSE-exposed, SARS-CoV-2-infected HBE cells. Knockdown of ACE-2 via an antisense oligonucleotide (ASO) reduced SARS-CoV-2 viral load and infection in CSE-exposed ferret airway cells that was augmented by co-administration of camostat mesylate to block TMPRSS2 activity. Smoking increases SARS-CoV-2 infection via upregulation of ACE2 and TMPRSS2.
Collapse
|
5
|
Fernandes Q, Inchakalody VP, Bedhiafi T, Mestiri S, Taib N, Uddin S, Merhi M, Dermime S. Chronic inflammation and cancer; the two sides of a coin. Life Sci 2024; 338:122390. [PMID: 38160787 DOI: 10.1016/j.lfs.2023.122390] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
The correlation between chronic inflammation and cancer was initially identified in the 19th century. Biomolecules like interleukins, chemokines, tumor necrosis factors, growth factors, and adhesion molecules, which regulate inflammation, are recognized contributors to neoplastic transformation through various mechanisms, including oncogenic mutations, resistance to apoptosis, and adaptive responses like angiogenesis. This review aims to establish connections between the intricate and complex mechanisms of chronic inflammation and cancer. We illuminate implicit signaling mechanisms that drive the association between chronic inflammation and the initiation/progression of cancer, exploring potential impacts on other diseases. Additionally, we discuss the modalities of currently available therapeutic options for chronic inflammation and cancer, emphasizing the dual nature of such therapies. A thorough understanding of the molecular basis of chronic inflammation is crucial for developing novel approaches in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Varghese Philipose Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Takwa Bedhiafi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
6
|
D’Amato A, Mariconda A, Iacopetta D, Ceramella J, Catalano A, Sinicropi MS, Longo P. Complexes of Ruthenium(II) as Promising Dual-Active Agents against Cancer and Viral Infections. Pharmaceuticals (Basel) 2023; 16:1729. [PMID: 38139855 PMCID: PMC10747139 DOI: 10.3390/ph16121729] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Poor responses to medical care and the failure of pharmacological treatment for many high-frequency diseases, such as cancer and viral infections, have been widely documented. In this context, numerous metal-based substances, including cisplatin, auranofin, various gold metallodrugs, and ruthenium complexes, are under study as possible anticancer and antiviral agents. The two Ru(III) and Ru(II) complexes, namely, BOLD-100 and RAPTA-C, are presently being studied in a clinical trial and preclinical studies evaluation, respectively, as anticancer agents. Interestingly, BOLD-100 has also recently demonstrated antiviral activity against SARS-CoV-2, which is the virus responsible for the COVID-19 pandemic. Over the last years, much effort has been dedicated to discovering new dual anticancer-antiviral agents. Ru-based complexes could be very suitable in this respect. Thus, this review focuses on the most recent studies regarding newly synthesized Ru(II) complexes for use as anticancer and/or antiviral agents.
Collapse
Affiliation(s)
- Assunta D’Amato
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| | | | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (D.I.); (J.C.); (M.S.S.)
| | - Pasquale Longo
- Department of Chemistry and Biology, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, Italy; (A.D.); (P.L.)
| |
Collapse
|
7
|
Laxmikeshav K, Rahman Z, Mahale A, Gurukkala Valapil D, Sharma P, George J, Phanindranath R, Dandekar MP, Kulkarni OP, Nagesh N, Shankaraiah N. Benzimidazole derivatives as tubulin polymerization inhibitors: Design, synthesis and in vitro cytotoxicity studies. Bioorg Med Chem Lett 2023; 96:129494. [PMID: 37797804 DOI: 10.1016/j.bmcl.2023.129494] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/28/2023] [Accepted: 10/01/2023] [Indexed: 10/07/2023]
Abstract
A new class of benzimidazole derivatives as tubulin polymerization inhibitors has been designed and synthesized in this study. The in vitro anticancer profile of the developed molecules was reconnoitred on selected human cancer cells. The highest cytotoxicity was illustrated by compounds 7n and 7u with IC50 values ranging from 2.55 to 17.89 µM with specificity toward SK-Mel-28 cells. They displayed 5-fold less cytotoxicity towards normal rat kidney epithelial NRK52E cells, which implies that they are not harmful to normal, healthy cells. The cellular staining procedures like AO/EB, DCFDA, and DAPI were applied to comprehend the inherent mechanism of apoptosis which displayed nuclear and morphological alterations. The Annexin V binding and JC-1 studies were executed to evaluate the extent of apoptosis and the decline in mitochondrial transmembrane potential in SK-Mel-28 cell lines. Compound 7n dose-dependently arrested the G2/M phase of the cell cycle and the target-based outcomes proposed tubulin polymerization inhibition by 7n (IC50 of 5.05±0.13 μM). Computational studies were also conducted on the tubulin protein (PDB ID: 3E22) to investigate the stabilized binding interactions of compounds 7n and 7u with tubulin, respectively.
Collapse
Affiliation(s)
- Kritika Laxmikeshav
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ziaur Rahman
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ashutosh Mahale
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Durgesh Gurukkala Valapil
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Pravesh Sharma
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Joel George
- CSIR-Centre for Cellular and Molecular Biology, Medical Biotechnology Complex, ANNEXE II, Uppal Road, Hyderabad 500007, India
| | - Regur Phanindranath
- CSIR-Centre for Cellular and Molecular Biology, Medical Biotechnology Complex, ANNEXE II, Uppal Road, Hyderabad 500007, India
| | - Manoj P Dandekar
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| | - Onkar P Kulkarni
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Hyderabad Campus, Hyderabad 500078, India
| | - Narayana Nagesh
- CSIR-Centre for Cellular and Molecular Biology, Medical Biotechnology Complex, ANNEXE II, Uppal Road, Hyderabad 500007, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India.
| |
Collapse
|
8
|
Li V, Lee Y, Lee C, Kim H. Repurposing existing drugs for monkeypox: applications of virtual screening methods. Genes Genomics 2023; 45:1347-1355. [PMID: 37713070 PMCID: PMC10587275 DOI: 10.1007/s13258-023-01449-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Monkeypox is endemic to African region and has become of Global concern recently due to its outbreaks in non-endemic countries. Although, the disease was first recorded in 1970, no monkeypox specific drug or vaccine exists as of now. METHODS We applied drug repositioning method, testing effectiveness of currently approved drugs against emerging disease, as one of the most affordable approaches for discovering novel treatment measures. Techniques such as virtual ligand-based and structure-based screening were applied to identify potential drug candidates against monkeypox. RESULTS We narrowed down our results to 6 antiviral and 20 anti-tumor drugs that exhibit theoretically higher potency than tecovirimat, the currently approved drug for monkeypox disease. CONCLUSIONS Our results indicated that selected drug compounds displayed strong binding affinity for p37 receptor of monkeypox virus and therefore can potentially be used in future studies to confirm their effectiveness against the disease.
Collapse
Affiliation(s)
- Vladimir Li
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Youngho Lee
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Chul Lee
- Laboratory of Neurogenetics of Language, The Rockefeller University, New York, NY, USA
| | - Heebal Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea.
- Department of Agricultural Biotechnology, Research Institute for Agriculture and Life Sciences, Seoul National University, Gwanak-gu 1, Gwanak-ro, Seoul, 08826, Republic of Korea.
- eGnome, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Jamir E, Sarma H, Priyadarsinee L, Kiewhuo K, Nagamani S, Sastry GN. Polypharmacology guided drug repositioning approach for SARS-CoV2. PLoS One 2023; 18:e0289890. [PMID: 37556478 PMCID: PMC10411734 DOI: 10.1371/journal.pone.0289890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
Drug repurposing has emerged as an important strategy and it has a great potential in identifying therapeutic applications for COVID-19. An extensive virtual screening of 4193 FDA approved drugs has been carried out against 24 proteins of SARS-CoV2 (NSP1-10 and NSP12-16, envelope, membrane, nucleoprotein, spike, ORF3a, ORF6, ORF7a, ORF8, and ORF9b). The drugs were classified into top 10 and bottom 10 drugs based on the docking scores followed by the distribution of their therapeutic indications. As a result, the top 10 drugs were found to have therapeutic indications for cancer, pain, neurological disorders, and viral and bacterial diseases. As drug resistance is one of the major challenges in antiviral drug discovery, polypharmacology and network pharmacology approaches were employed in the study to identify drugs interacting with multiple targets and drugs such as dihydroergotamine, ergotamine, bisdequalinium chloride, midostaurin, temoporfin, tirilazad, and venetoclax were identified among the multi-targeting drugs. Further, a pathway analysis of the genes related to the multi-targeting drugs was carried which provides insight into the mechanism of drugs and identifying targetable genes and biological pathways involved in SARS-CoV2.
Collapse
Affiliation(s)
- Esther Jamir
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
| | - Lipsa Priyadarsinee
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kikrusenuo Kiewhuo
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Selvaraman Nagamani
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - G. Narahari Sastry
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
10
|
Zhang L. Biomedical equipments, vaccine and drug in the prevention, diagnosis and treatment of COVID-19. Heliyon 2023; 9:e18089. [PMID: 37483808 PMCID: PMC10362228 DOI: 10.1016/j.heliyon.2023.e18089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/05/2023] [Accepted: 07/06/2023] [Indexed: 07/25/2023] Open
Abstract
SARS-CoV-2 virus caused an infectious disease, named COVID-19. Biomedical equipments, vaccine and drug have played a crucial role in the prevention, diagnosis and treatment. Nevertheless, up to now, there still has been no literature summarizing the diagnosis, prevention and treatment of this infectious disease from the perspective of biomedical equipments. Thus, this review wants to give an overview on the biomedical equipments, vaccine and drug in the prevention, diagnosis and treatment of this disease, and avoids the overlap with previous research, more emphasis on biomedical equipments, and less emphasis on biomaterials. The existing problems in the current research and application were summarized, and the future research direction was proposed, so as to provide reference to deal with similar viral infections in the future.
Collapse
|
11
|
Hakem A, Desmarets L, Sahli R, Malek RB, Camuzet C, François N, Lefèvre G, Samaillie J, Moureu S, Sahpaz S, Belouzard S, Ksouri R, Séron K, Rivière C. Luteolin Isolated from Juncus acutus L., a Potential Remedy for Human Coronavirus 229E. Molecules 2023; 28:molecules28114263. [PMID: 37298740 DOI: 10.3390/molecules28114263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 05/17/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, addressed the lack of specific antiviral drugs against coronaviruses. In this study, bioguided fractionation performed on both ethyl acetate and aqueous sub-extracts of Juncus acutus stems led to identifying luteolin as a highly active antiviral molecule against human coronavirus HCoV-229E. The apolar sub-extract (CH2Cl2) containing phenanthrene derivatives did not show antiviral activity against this coronavirus. Infection tests on Huh-7 cells, expressing or not the cellular protease TMPRSS2, using luciferase reporter virus HCoV-229E-Luc showed that luteolin exhibited a dose-dependent inhibition of infection. Respective IC50 values of 1.77 µM and 1.95 µM were determined. Under its glycosylated form (luteolin-7-O-glucoside), luteolin was inactive against HCoV-229E. Time of addition assay showed that utmost anti-HCoV-229E activity of luteolin was achieved when added at the post-inoculation step, indicating that luteolin acts as an inhibitor of the replication step of HCoV-229E. Unfortunately, no obvious antiviral activity for luteolin was found against SARS-CoV-2 and MERS-CoV in this study. In conclusion, luteolin isolated from Juncus acutus is a new inhibitor of alphacoronavirus HCoV-229E.
Collapse
Affiliation(s)
- Asma Hakem
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
- Laboratory of Aromatic and Medicinal Plants, Biotechnology Centre of Borj-Cedria (CBBC), Hammam-Lif 2050, Tunisia
| | - Lowiese Desmarets
- Center for Infection and Immunity of Lille (CIIL), Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, 59000 Lille, France
| | - Ramla Sahli
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
- Laboratory of Aromatic and Medicinal Plants, Biotechnology Centre of Borj-Cedria (CBBC), Hammam-Lif 2050, Tunisia
| | - Rawen Ben Malek
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| | - Charline Camuzet
- Center for Infection and Immunity of Lille (CIIL), Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, 59000 Lille, France
| | - Nathan François
- Center for Infection and Immunity of Lille (CIIL), Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, 59000 Lille, France
| | - Gabriel Lefèvre
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| | - Jennifer Samaillie
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| | - Sophie Moureu
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| | - Sevser Sahpaz
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| | - Sandrine Belouzard
- Center for Infection and Immunity of Lille (CIIL), Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, 59000 Lille, France
| | - Riadh Ksouri
- Laboratory of Aromatic and Medicinal Plants, Biotechnology Centre of Borj-Cedria (CBBC), Hammam-Lif 2050, Tunisia
| | - Karin Séron
- Center for Infection and Immunity of Lille (CIIL), Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, U1019-UMR 9017, 59000 Lille, France
| | - Céline Rivière
- Joint Research Unit 1158, BioEcoAgro, Univ. Lille, INRAE, Univ. Liège, UPJV, JUNIA, Univ. Artois, Univ. Littoral Côte d'Opale, ICV-Institut Charles Viollette, 59650 Villeneuve-d'Ascq, France
| |
Collapse
|
12
|
Bukhari SNA, Abdelgawad MA, Ahmed N, Amjad MW, Hussain MA, Elsherif MA, Ejaz H, Alotaibi NH, Filipović I, Janković N. Synthesis, Characterization, and Biological Evaluation of Meldrum's Acid Derivatives: Dual Activity and Molecular Docking Study. Pharmaceuticals (Basel) 2023; 16:281. [PMID: 37259425 PMCID: PMC9968196 DOI: 10.3390/ph16020281] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 09/28/2023] Open
Abstract
In the presented study, eight novel Meldrum's acid derivatives containing various vanillic groups were synthesized. Vanillidene Meldrum's acid compounds were tested against different cancer cell lines and microbes. Out of nine, three showed very good biological activity against E. coli, and HeLa and A549 cell lines. It is shown that the O-alkyl substituted derivatives possessed better antimicrobial and anticancer activities in comparison with the O-acyl ones. The decyl substituted molecule (3i) has the highest activity against E. coli (MIC = 12.4 μM) and cancer cell lines (HeLa, A549, and LS174 = 15.7, 21.8, and 30.5 μM, respectively). The selectivity index of 3i is 4.8 (HeLa). The molecular docking study indicates that compound 3i showed good binding affinity to DNA, E. coli Gyrase B, and topoisomerase II beta. The covalent docking showed that 3i was a Michael acceptor for the nucleophiles Lys and Ser. The best Eb was noted for the topoisomerase II beta-LYS482-3i cluster.
Collapse
Affiliation(s)
- Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka 72388, Al Jouf, Saudi Arabia
| | | | - Naveed Ahmed
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72388, Al Jouf, Saudi Arabia
| | - Muhammad Wahab Amjad
- Center for Ultrasound Molecular Imaging and Therapeutics, Pittsburgh Heart, Lung, Blood and Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Muhammad Ajaz Hussain
- Centre for Organic Chemistry, School of Chemistry, University of the Punjab, Lahore 54590, Pakistan
| | - Mervat A. Elsherif
- Chemistry Department, College of Science, Jouf University, Sakaka 72388, Al Jouf, Saudi Arabia
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Al Jouf, Saudi Arabia
| | - Nasser H. Alotaibi
- Department of Clinical Pharmacy, College of Pharmacy, Jouf University, Sakaka 72388, Al Jouf, Saudi Arabia
| | - Ignjat Filipović
- University of Kragujevac, Faculty of Science, Radoja Domanovića 12, 34000 Kragujevac, Serbia
| | - Nenad Janković
- University of Kragujevac, Department of Science, Institute for Information Technologies Kragujevac, Jovana Cvijića bb, 34000 Kragujevac, Serbia
| |
Collapse
|
13
|
Duhalde Vega M, Olivera D, Gastão Davanzo G, Bertullo M, Noya V, Fabiano de Souza G, Primon Muraro S, Castro I, Arévalo AP, Crispo M, Galliussi G, Russo S, Charbonnier D, Rammauro F, Jeldres M, Alamón C, Varela V, Batthyany C, Bollati-Fogolín M, Oppezzo P, Pritsch O, Proença-Módena JL, Nakaya HI, Trias E, Barbeito L, Anegon I, Cuturi MC, Moraes-Vieira P, Segovia M, Hill M. PD-1/PD-L1 blockade abrogates a dysfunctional innate-adaptive immune axis in critical β-coronavirus disease. SCIENCE ADVANCES 2022; 8:eabn6545. [PMID: 36129987 PMCID: PMC9491709 DOI: 10.1126/sciadv.abn6545] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Severe COVID-19 is associated with hyperinflammation and weak T cell responses against SARS-CoV-2. However, the links between those processes remain partially characterized. Moreover, whether and how therapeutically manipulating T cells may benefit patients are unknown. Our genetic and pharmacological evidence demonstrates that the ion channel TMEM176B inhibited inflammasome activation triggered by SARS-CoV-2 and SARS-CoV-2-related murine β-coronavirus. Tmem176b-/- mice infected with murine β-coronavirus developed inflammasome-dependent T cell dysfunction and critical disease, which was controlled by modulating dysfunctional T cells with PD-1 blockers. In critical COVID-19, inflammasome activation correlated with dysfunctional T cells and low monocytic TMEM176B expression, whereas PD-L1 blockade rescued T cell functionality. Here, we mechanistically link T cell dysfunction and inflammation, supporting a cancer immunotherapy to reinforce T cell immunity in critical β-coronavirus disease.
Collapse
Affiliation(s)
- Maite Duhalde Vega
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Daniela Olivera
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
| | - Gustavo Gastão Davanzo
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | | | - Verónica Noya
- Laboratory of Molecular Biology, Sanatorio Americano, 11600 Montevideo, Uruguay
| | - Gabriela Fabiano de Souza
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | - Stéfanie Primon Muraro
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | | | - Ana Paula Arévalo
- Laboratory Animals Biotechnology, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Martina Crispo
- Laboratory Animals Biotechnology, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Germán Galliussi
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Sofía Russo
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
| | - David Charbonnier
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Florencia Rammauro
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
- Laboratory of Immunovirology, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Mathías Jeldres
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
| | - Catalina Alamón
- Laboratorio de Neurodegeneración, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Valentina Varela
- Laboratorio de Neurodegeneración, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Carlos Batthyany
- Laboratory of Vascular Biology and Drug Development, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | | | - Pablo Oppezzo
- Research Laboratory on Chronic Lymphocytic Leukemia, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Otto Pritsch
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
- Laboratory of Immunovirology, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - José Luiz Proença-Módena
- Laboratory of Emerging Viruses, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | | | - Emiliano Trias
- Laboratorio de Neurodegeneración, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Luis Barbeito
- Laboratorio de Neurodegeneración, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
| | - Ignacio Anegon
- INSERM UMR 1064, Center for Research in Transplantation and Immunology; Université de Nantes; CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), 44093 Nantes, France
| | - María Cristina Cuturi
- INSERM UMR 1064, Center for Research in Transplantation and Immunology; Université de Nantes; CHU Nantes, Institut de Transplantation Urologie Néphrologie (ITUN), 44093 Nantes, France
| | - Pedro Moraes-Vieira
- Laboratory of Immunometabolism, Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, SP, Brazil
| | - Mercedes Segovia
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
- Corresponding author. (M.S.); (M.H.)
| | - Marcelo Hill
- Laboratory of Immunoregulation and Inflammation, Institut Pasteur de Montevideo, 11400 Montevideo, Uruguay
- Immunobiology Department, Faculty of Medicine, University of the Republic, 11800 Montevideo, Uruguay
- Corresponding author. (M.S.); (M.H.)
| |
Collapse
|
14
|
Fawazy NG, Panda SS, Mostafa A, Kariuki BM, Bekheit MS, Moatasim Y, Kutkat O, Fayad W, El-Manawaty MA, Soliman AAF, El-Shiekh RA, Srour AM, Barghash RF, Girgis AS. Development of spiro-3-indolin-2-one containing compounds of antiproliferative and anti-SARS-CoV-2 properties. Sci Rep 2022; 12:13880. [PMID: 35974029 PMCID: PMC9380671 DOI: 10.1038/s41598-022-17883-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 08/02/2022] [Indexed: 11/09/2022] Open
Abstract
A series of 1″-(alkylsulfonyl)-dispiro[indoline-3,2'-pyrrolidine-3',3″-piperidine]-2,4″-diones 6a‒o has been synthesized through regioselective multi-component azomethine dipolar cycloaddition reaction of 1-(alkylsulfonyl)-3,5-bis(ylidene)-piperidin-4-ones 3a‒h. X-ray diffraction studies (6b‒d,h) confirmed the structures. The majority of the synthesized analogs reveal promising antiproliferation properties against a variety of human cancer cell lines (MCF7, HCT116, A431 and PaCa2) with good selectivity index towards normal cell (RPE1). Some of the synthesized agents exhibit potent inhibitory properties against the tested cell lines with higher efficacies than the standard references (sunitinib and 5-fluorouracil). Compound 6m is the most potent. Multi-targeted inhibitory properties against EGFR and VEGFR-2 have been observed for the synthesized agents. Flow cytometry supports the antiproliferation properties and shows the tested agents as apoptosis and necrosis forming. Vero cell viral infection model demonstrates the anti-SARS-CoV-2 properties of the synthesized agents. Compound 6f is the most promising (about 3.3 and 4.8 times the potency of the standard references, chloroquine and hydroxychloroquine). QSAR models explain and support the observed biological properties.
Collapse
Affiliation(s)
- Nehmedo G Fawazy
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Siva S Panda
- Department of Chemistry and Physics, Augusta University, Augusta, GA, 30912, USA
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Benson M Kariuki
- School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, UK
| | - Mohamed S Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Omnia Kutkat
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza, 12622, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - May A El-Manawaty
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Ahmed A F Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| | - Aladdin M Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Reham F Barghash
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt
| | - Adel S Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza, 12622, Egypt.
| |
Collapse
|
15
|
Pal D, Song IH, Dashrath Warkad S, Song KS, Seong Yeom G, Saha S, Shinde PB, Balasaheb Nimse S. Indazole-based microtubule-targeting agents as potential candidates for anticancer drugs discovery. Bioorg Chem 2022; 122:105735. [PMID: 35298962 DOI: 10.1016/j.bioorg.2022.105735] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/12/2022] [Accepted: 03/07/2022] [Indexed: 12/13/2022]
Abstract
Tremendous research is focused on developing novel drug candidates targeting microtubules to inhibit their function in several cellular processes, including cell division. In this regard, several indazole derivatives were sought to target the colchicine binding site on the β-tubulin, a crucial protein required to form microtubules, to develop microtubule targeting agents. Even though there are several reviews on the indazole-based compounds, none of them focused on using indazole scaffold to develop microtubule targeting agents. Therefore, this review aims to present the advances in research on compounds containing indazole scaffolds as microtubule targeting agents based on the articles published in the last two decades. Among the articles reviewed, we found that compounds 6 and 7 showed the lowest IC50 values of 0.6 ∼ 0.9 nM in the cell line studies, making them the strongest indazole derivatives that target microtubules. The compounds 30, 31, 37 (IC50 = ∼ 1 nM) and compounds 8, 38 (IC50 = ∼ 2 nM) have proved to be potent microtubule inhibitors. The compounds 18, 31, 44, 45 also showed strong anticancer activity (IC50 = ∼ 8 nM). It is important to notice that except for compounds 9, 12, 13, 15, and SRF, the top activity compounds including 6, 7, 8, 10, 11, 30, 31, 37, 44, and 45 contain 3,4,5‑trimethoxyphenyl substitution similar to that of colchicine. Therefore, it appears that the 3,4,5‑trimethoxyphenyl substituent on the indazole scaffold is crucial for targeting CBS.
Collapse
Affiliation(s)
- Dilipkumar Pal
- Department of Pharmaceutical Sciences, Guru Ghasidas Viswavidyalaya (A Central University), Bilaspur, Chhattisgarh 495009, India
| | - In-Ho Song
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 200702, South Korea
| | | | - Keum-Soo Song
- Biometrix Technology, Inc., 2-2 Bio Venture Plaza 56, Chuncheon 24232, South Korea
| | - Gyu Seong Yeom
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 200702, South Korea
| | - Supriyo Saha
- Sardar Bhagwan Singh Post Graduate Institute of Biomedical Science and Research, India
| | - Pramod B Shinde
- Natural Products & Green Chemistry Division, CSIR-Central Salt and Marine Chemicals Research Institute (CSIR-CSMCRI), Council of Scientific and Industrial Research (CSIR), Bhavnagar 364002, Gujarat, India
| | - Satish Balasaheb Nimse
- Institute of Applied Chemistry and Department of Chemistry, Hallym University, Chuncheon 200702, South Korea.
| |
Collapse
|
16
|
Fenton C, Keam SJ. Emerging small molecule antivirals may fit neatly into COVID-19 treatment. DRUGS & THERAPY PERSPECTIVES 2022; 38:112-126. [PMID: 35250258 PMCID: PMC8882464 DOI: 10.1007/s40267-022-00897-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 12/15/2022]
Abstract
Numerous treatments exist for COVID-19, the illness caused by SARS-CoV-2 virus, although most are not well established; among these are several small molecule antiviral agents. Intravenous remdesivir is an established treatment worldwide for inpatients and in some countries is also available for use in non-hospitalised high risk patients to prevent progression to severe disease and hospitalization. Oral molnupiravir and oral nirmatrelvir-ritonavir are also available in several countries to prevent progression to severe disease and hospitalization for high-risk outpatients. Many other antiviral small molecules that may have therapeutic potential are under investigation in clinical trials. This article provides a summary of key molecular targets, pharmacology and preliminary data on the efficacy and safety of small molecule antiviral agents being investigated for the treatment of COVID-19.
Collapse
Affiliation(s)
- Caroline Fenton
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754 New Zealand
| | - Susan J. Keam
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754 New Zealand
| |
Collapse
|
17
|
Hu T, Pan M, Yin Y, Wang C, Cui Y, Wang Q. The Regulatory Network of Cyclic GMP-AMP Synthase-Stimulator of Interferon Genes Pathway in Viral Evasion. Front Microbiol 2021; 12:790714. [PMID: 34966372 PMCID: PMC8711784 DOI: 10.3389/fmicb.2021.790714] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/04/2021] [Indexed: 01/06/2023] Open
Abstract
Virus infection has been consistently threatening public health. The cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway is a critical defender to sense various pathogens and trigger innate immunity of mammalian cells. cGAS recognizes the pathogenic DNA in the cytosol and then synthesizes 2'3'-cyclic GMP-AMP (2'3'cGAMP). As the second messenger, cGAMP activates STING and induces the following cascade to produce type I interferon (IFN-I) to protect against infections. However, viruses have evolved numerous strategies to hinder the cGAS-STING signal transduction, promoting their immune evasion. Here we outline the current status of the viral evasion mechanism underlying the regulation of the cGAS-STING pathway, focusing on how post-transcriptional modifications, viral proteins, and non-coding RNAs involve innate immunity during viral infection, attempting to inspire new targets discovery and uncover potential clinical antiviral treatments.
Collapse
Affiliation(s)
- Tongyu Hu
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mingyu Pan
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yue Yin
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Ye Cui
- Division of Immunology, The Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, Department of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
18
|
Srour AM, Panda SS, Mostafa A, Fayad W, El-Manawaty MA, A. F. Soliman A, Moatasim Y, El Taweel A, Abdelhameed MF, Bekheit MS, Ali MA, Girgis AS. Synthesis of aspirin-curcumin mimic conjugates of potential antitumor and anti-SARS-CoV-2 properties. Bioorg Chem 2021; 117:105466. [PMID: 34775204 PMCID: PMC8566089 DOI: 10.1016/j.bioorg.2021.105466] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 10/31/2021] [Indexed: 12/16/2022]
Abstract
Series of piperidone-salicylate conjugates were synthesized through the reaction of 3E,5E-bis(arylidene)-4-piperidones with the appropriate acid chloride of acetylsalicylate in the presence of triethylamine. All the synthesized conjugates reveal antiproliferative properties against A431 (squamous skin) cancer cell line with potency higher than that of 5-fluorouracil. Many of the synthesized agents also exhibit promising antiproliferative properties against HCT116 (colon) cancer cell line, of which 5o and 5c are the most effective with 12.9, 9.8 folds potency compared with Sunitinib. Promising activity is also shown against MCF7 (breast) cancer cell line with 1.19, 1.12 folds relative to 5-fluorouracil. PI-flow cytometry of compound 5c supports the arrest of cell cycle at G1-phase. However, compound 5o and Sunitinib arrest the cell cycle at S-phase. The synthesized conjugates can be considered as multi-targeted tyrosine kinase inhibitors due to the promising properties against VEGFR-2 and EGFR in MCF7 and HCT116. CDOCKER studies support the EGFR inhibitory properties. Compounds 5p and 5i possessing thienylidene heterocycle are anti-SARS-CoV-2 with high therapeutic indices. Many of the synthesized agents show enhanced COX-1/2 properties than aspirin with better selectivity index towards COX-2 relative to COX-1. The possible applicability of the potent candidates discovered as antitumor and anti-SARS-CoV-2 is supported by the safe profile against normal (non-cancer, RPE1 and VERO-E6) cells.
Collapse
Affiliation(s)
- Aladdin M. Srour
- Department of Therapeutic Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Siva S. Panda
- Department of Chemistry & Physics, Augusta University, Augusta, GA 30912, US
| | - Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Walid Fayad
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - May A. El-Manawaty
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Ahmed A. F. Soliman
- Drug Bioassay-Cell Culture Laboratory, Pharmacognosy Department, National Research Centre, Dokki, Giza 12622, Egypt
| | - Yassmin Moatasim
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Ahmed El Taweel
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | | | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, National Research Centre, Giza 12622, Egypt
| | - Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt,Corresponding author
| |
Collapse
|
19
|
Thorat N, Pricl S, Parchur AK, Somvanshi SB, Li Q, Umrao S, Townley H. Safeguarding COVID-19 and cancer management: drug design and therapeutic approach. OPEN RESEARCH EUROPE 2021; 1:77. [PMID: 37645153 PMCID: PMC10445946 DOI: 10.12688/openreseurope.13841.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/24/2021] [Indexed: 08/31/2023]
Abstract
Recent clinical cohort studies have highlighted that there is a three-fold greater SARS-Cov-2 infection risk in cancer patients, and overall mortality in individuals with tumours is increased by 41% with respect to general COVID-19 patients. Thus, access to therapeutics and intensive care is compromised for people with both diseases (comorbidity) and there is risk of delayed access to diagnosis. This comorbidity has resulted in extensive burden on the treatment of patients and health care system across the globe; moreover, mortality of hospitalized patients with comorbidity is reported to be 30% higher than for individuals affected by either disease. In this data-driven review, we aim specifically to address drug discoveries and clinical data of cancer management during the COVID-19 pandemic. The review will extensively address the treatment of COVID-19/cancer comorbidity; treatment protocols and new drug discoveries, including the description of drugs currently available in clinical settings; demographic features; and COVID-19 outcomes in cancer patients worldwide.
Collapse
Affiliation(s)
- Nanasaheb Thorat
- Nuffield Department of Women’s & Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Sabrina Pricl
- MolBNL@UniTS-DEA, University of Trieste, Piazzale Europa 1, Trieste, 34127, Italy
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, 90-136, Poland
| | - Abdul K. Parchur
- Radiation Oncology, Froedtert Hospital & Medical College of Wisconsin, Medical College of Wisconsin, Wisconsin, USA
| | - Sandeep B. Somvanshi
- School of Materials Engineering, Purdue University, West Stadium Avenue, West Lafayette, USA
| | - Qifei Li
- Division of Newborn Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Sachin Umrao
- Department of Therapeutic Radiology, Yale School of Medicine, Yale University, New Haven, USA
| | - Helen Townley
- Nuffield Department of Women’s & Reproductive Health, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Department of Engineering Science, University of Oxford, Oxford, UK
| |
Collapse
|