1
|
Zheng J, Chen C, Huang Y, Fang S, Guo P, Liu S, Ouyang G. A fast solid-phase microextraction scheme for in vivo monitoring of bio-accumulation and bio-transformation of arbidol in living plants. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 956:177175. [PMID: 39461518 DOI: 10.1016/j.scitotenv.2024.177175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Abstract
Large quantity of the antiviral drug arbidol is used for resisting virus infection like the Corona Virus Disease 2019 and influenza, resulting in unanticipated environmental pollution. Herein, to investigate the environmental risks of the unanticipated arbidol contamination, a novel in vivo sampling probe was developed based on a bromo-substituted porous organic polymer (Br-POP) and then adopted for tracking the bio-accumulation and bio-transformation of arbidol in living plants by coupling with a nano-electrospray ionization fourier-transform ion cyclotron resonance mass spectrometry (Nano-ESI-FT-ICR-MS) method. The established method showed good extraction performance towards arbidol with limit of detection (LOD) of 0.48 ng g-1, and relative standard deviation (RSD) of single-and multiple- probe of 2.2 and 14 %. Owing to the interactions between the Br-POP and the target analytes, as well as the fast analysis process of Nano-ESI-FT-ICR-MS, <6 min was cost for total sampling and analysis duration, achieving hourly tracking of arbidol and its metabolites in this work. During 21-d in vivo tracking, the concentration of arbidol in living plant stems increased rapidly within 6 h and peaked at 413.93 ± 47.09 ng g-1. Meanwhile, it was found that dissolved organic matters (DOM) had significant effect on arbidol behaviors in living plants, resulting in a decrease of the maximum concentration of arbidol in plant stems (152.70 ± 42.44 ng g-1) and the change of dominant metabolite of arbidol that the S-oxidation rather than N-demethylation product of arbidol was dominant with DOM participation. Additionally, the plant root secretion was found to be significantly altered by arbidol exposure. To summarized, the combination of in vivo SPME and the FT-ICR-MS analysis provide new and important information regarding arbidol contamination and related alternation of plant root metabolism.
Collapse
Affiliation(s)
- Jiating Zheng
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China
| | - Chao Chen
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center, Guangzhou), Guangzhou 510070, China
| | - Yiquan Huang
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center, Guangzhou), Guangzhou 510070, China
| | - Shuting Fang
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China; Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center, Guangzhou), Guangzhou 510070, China
| | - Pengran Guo
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center, Guangzhou), Guangzhou 510070, China
| | - Shuqin Liu
- College of Environment and Climate, Guangdong Provincial Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, China.
| | - Gangfeng Ouyang
- School of Chemical Engineering and Technology, Sun Yat-sen University, Zhuhai 519082, China; Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Institute of Analysis, Guangdong Academy of Sciences (China National Analytical Center, Guangzhou), Guangzhou 510070, China
| |
Collapse
|
2
|
Chakraborty S, Chauhan A. Fighting the flu: a brief review on anti-influenza agents. Biotechnol Genet Eng Rev 2024; 40:858-909. [PMID: 36946567 DOI: 10.1080/02648725.2023.2191081] [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: 10/26/2022] [Accepted: 03/06/2023] [Indexed: 03/23/2023]
Abstract
The influenza virus causes one of the most prevalent and lethal infectious viral diseases of the respiratory system; the disease progression varies from acute self-limiting mild fever to disease chronicity and death. Although both the preventive and treatment measures have been vital in protecting humans against seasonal epidemics or sporadic pandemics, there are several challenges to curb the influenza virus such as limited or poor cross-protection against circulating virus strains, moderate protection in immune-compromised patients, and rapid emergence of resistance. Currently, there are four US-FDA-approved anti-influenza drugs to treat flu infection, viz. Rapivab, Relenza, Tamiflu, and Xofluza. These drugs are classified based on their mode of action against the viral replication cycle with the first three being Neuraminidase inhibitors, and the fourth one targeting the viral polymerase. The emergence of the drug-resistant strains of influenza, however, underscores the need for continuous innovation towards development and discovery of new anti-influenza agents with enhanced antiviral effects, greater safety, and improved tolerability. Here in this review, we highlighted commercially available antiviral agents besides those that are at different stages of development including under clinical trials, with a brief account of their antiviral mechanisms.
Collapse
Affiliation(s)
| | - Ashwini Chauhan
- Department of Microbiology, Tripura University, Agartala, India
| |
Collapse
|
3
|
Israr J, Alam S, Kumar A. Drug repurposing for respiratory infections. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:207-230. [PMID: 38942538 DOI: 10.1016/bs.pmbts.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Respiratory infections such as Coronavirus disease 2019 are a substantial worldwide health challenge, frequently resulting in severe sickness and death, especially in susceptible groups. Conventional drug development for respiratory infections faces obstacles such as extended timescales, substantial expenses, and the rise of resistance to current treatments. Drug repurposing is a potential method that has evolved to quickly find and reuse existing medications for treating respiratory infections. Drug repurposing utilizes medications previously approved for different purposes, providing a cost-effective and time-efficient method to tackle pressing medical needs. This chapter summarizes current progress and obstacles in repurposing medications for respiratory infections, focusing on notable examples of repurposed pharmaceuticals and their probable modes of action. The text also explores the significance of computational approaches, high-throughput screening, and preclinical investigations in identifying potential candidates for repurposing. The text delves into the significance of regulatory factors, clinical trial structure, and actual data in confirming the effectiveness and safety of repurposed medications for respiratory infections. Drug repurposing is a valuable technique for quickly increasing the range of treatments for respiratory infections, leading to better patient outcomes and decreasing the worldwide disease burden.
Collapse
Affiliation(s)
- Juveriya Israr
- Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India; Department of Biotechnology, Era University, Lucknow, Uttar Pradesh, India
| | - Shabroz Alam
- Department of Biotechnology, Era University, Lucknow, Uttar Pradesh, India
| | - Ajay Kumar
- Department of Biotechnology, Faculty of Engineering and Technology, Rama University, Mandhana, Kanpur, Uttar Pradesh, India.
| |
Collapse
|
4
|
Iyer K, Yan Z, Ross SR. Entry inhibitors as arenavirus antivirals. Front Microbiol 2024; 15:1382953. [PMID: 38650890 PMCID: PMC11033450 DOI: 10.3389/fmicb.2024.1382953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Arenaviruses belonging to the Arenaviridae family, genus mammarenavirus, are enveloped, single-stranded RNA viruses primarily found in rodent species, that cause severe hemorrhagic fever in humans. With high mortality rates and limited treatment options, the search for effective antivirals is imperative. Current treatments, notably ribavirin and other nucleoside inhibitors, are only partially effective and have significant side effects. The high lethality and lack of treatment, coupled with the absence of vaccines for all but Junín virus, has led to the classification of these viruses as Category A pathogens by the Centers for Disease Control (CDC). This review focuses on entry inhibitors as potential therapeutics against mammarenaviruses, which include both New World and Old World arenaviruses. Various entry inhibition strategies, including small molecule inhibitors and neutralizing antibodies, have been explored through high throughput screening, genome-wide studies, and drug repurposing. Notable progress has been made in identifying molecules that target receptor binding, internalization, or fusion steps. Despite promising preclinical results, the translation of entry inhibitors to approved human therapeutics has faced challenges. Many have only been tested in in vitro or animal models, and a number of candidates showed efficacy only against specific arenaviruses, limiting their broader applicability. The widespread existence of arenaviruses in various rodent species and their potential for their zoonotic transmission also underscores the need for rapid development and deployment of successful pan-arenavirus therapeutics. The diverse pool of candidate molecules in the pipeline provides hope for the eventual discovery of a broadly effective arenavirus antiviral.
Collapse
Affiliation(s)
| | | | - Susan R. Ross
- Department of Microbiology and Immunology, University of Illinois, College of Medicine, Chicago, IL, United States
| |
Collapse
|
5
|
Xu S, Esmaeili S, Cardozo-Ojeda EF, Goyal A, White JM, Polyak SJ, Schiffer JT. Two-way pharmacodynamic modeling of drug combinations and its application to pairs of repurposed Ebola and SARS-CoV-2 agents. Antimicrob Agents Chemother 2024; 68:e0101523. [PMID: 38470112 PMCID: PMC10989026 DOI: 10.1128/aac.01015-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 02/20/2024] [Indexed: 03/13/2024] Open
Abstract
Existing pharmacodynamic (PD) mathematical models for drug combinations discriminate antagonistic, additive, multiplicative, and synergistic effects, but fail to consider how concentration-dependent drug interaction effects may vary across an entire dose-response matrix. We developed a two-way pharmacodynamic (TWPD) model to capture the PD of two-drug combinations. TWPD captures interactions between upstream and downstream drugs that act on different stages of viral replication, by quantifying upstream drug efficacy and concentration-dependent effects on downstream drug pharmacodynamic parameters. We applied TWPD to previously published in vitro drug matrixes for repurposed potential anti-Ebola and anti-SARS-CoV-2 drug pairs. Depending on the drug pairing, the model recapitulated combined efficacies as or more accurately than existing models and can be used to infer efficacy at untested drug concentrations. TWPD fits the data slightly better in one direction for all drug pairs, meaning that we can tentatively infer the upstream drug. Based on its high accuracy, TWPD could be used in concert with PK models to estimate the therapeutic effects of drug pairs in vivo.
Collapse
Affiliation(s)
- Shuang Xu
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shadisadat Esmaeili
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - E. Fabian Cardozo-Ojeda
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Ashish Goyal
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Judith M. White
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Joshua T. Schiffer
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| |
Collapse
|
6
|
Brady DK, Gurijala AR, Huang L, Hussain AA, Lingan AL, Pembridge OG, Ratangee BA, Sealy TT, Vallone KT, Clements TP. A guide to COVID-19 antiviral therapeutics: a summary and perspective of the antiviral weapons against SARS-CoV-2 infection. FEBS J 2024; 291:1632-1662. [PMID: 36266238 PMCID: PMC9874604 DOI: 10.1111/febs.16662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/11/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022]
Abstract
Antiviral therapies are integral in the fight against SARS-CoV-2 (i.e. severe acute respiratory syndrome coronavirus 2), the causative agent of COVID-19. Antiviral therapeutics can be divided into categories based on how they combat the virus, including viral entry into the host cell, viral replication, protein trafficking, post-translational processing, and immune response regulation. Drugs that target how the virus enters the cell include: Evusheld, REGEN-COV, bamlanivimab and etesevimab, bebtelovimab, sotrovimab, Arbidol, nitazoxanide, and chloroquine. Drugs that prevent the virus from replicating include: Paxlovid, remdesivir, molnupiravir, favipiravir, ribavirin, and Kaletra. Drugs that interfere with protein trafficking and post-translational processing include nitazoxanide and ivermectin. Lastly, drugs that target immune response regulation include interferons and the use of anti-inflammatory drugs such as dexamethasone. Antiviral therapies offer an alternative solution for those unable or unwilling to be vaccinated and are a vital weapon in the battle against the global pandemic. Learning more about these therapies helps raise awareness in the general population about the options available to them with respect to aiding in the reduction of the severity of COVID-19 infection. In this 'A Guide To' article, we provide an in-depth insight into the development of antiviral therapeutics against SARS-CoV-2 and their ability to help fight COVID-19.
Collapse
Affiliation(s)
- Drugan K. Brady
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Aashi R. Gurijala
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Liyu Huang
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Ali A. Hussain
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Audrey L. Lingan
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | | | - Brina A. Ratangee
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Tristan T. Sealy
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | - Kyle T. Vallone
- Department of Biological SciencesVanderbilt UniversityNashvilleTNUSA
| | | |
Collapse
|
7
|
Zeng W, Han C, Mohammed S, Li S, Song Y, Sun F, Du Y. Indole-containing pharmaceuticals: targets, pharmacological activities, and SAR studies. RSC Med Chem 2024; 15:788-808. [PMID: 38516587 PMCID: PMC10953485 DOI: 10.1039/d3md00677h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/29/2024] [Indexed: 03/23/2024] Open
Abstract
Indole is a prestigious heterocyclic skeleton widely found in both naturally-occurring and biologically-active compounds. Pharmaceutical agents containing an indole skeleton in their framework possess a wide range of pharmacological properties, including antiviral, antitumor, analgesic, and other therapeutic activities, and many indole-containing drugs have been proven to have excellent pharmacokinetic and pharmacological effects. Over the past few decades, the FDA has approved over 40 indole-containing drugs for the treatment of various clinical conditions, and the development of indole-related drugs has attracted significant attention from medicinal chemists. This review aims to provide an overview of all the approved drugs that contain an indole nucleus, focusing on their targets, pharmacological activities, and SAR studies.
Collapse
Affiliation(s)
- Wei Zeng
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Chi Han
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Sarah Mohammed
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Shanshan Li
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| | - Yixuan Song
- Department of Chemical Engineering & Biotechnology, University of Cambridge CB2 3RA Cambridge UK
| | - Fengxia Sun
- Research Center for Chemical Safety & Security and Verification Technology & College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology Shijiazhuang 050018 China
| | - Yunfei Du
- School of Pharmaceutical Science and Technology, Tianjin University Tianjin 300072 China
| |
Collapse
|
8
|
Khramchikhin AV, Skryl’nikova MA, Gureev MA, Zarubaev VV, Esaulkova IL, Ilyina PA, Mammeri OA, Spiridonova DV, Porozov YB, Ostrovskii VA. Novel 1,2,4-Triazole- and Tetrazole-Containing 4 H-Thiopyrano[2,3- b]quinolines: Synthesis Based on the Thio-Michael/aza-Morita-Baylis-Hillman Tandem Reaction and Investigation of Antiviral Activity. Molecules 2023; 28:7427. [PMID: 37959845 PMCID: PMC10650458 DOI: 10.3390/molecules28217427] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/22/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
A novel method for synthesizing 1,2,4-triazole- and tetrazole-containing 4H-thiopyrano[2,3-b]quinolines using a new combination of the thio-Michael and aza-Morita-Baylis-Hillman reactions was developed. Target compounds were evaluated for their cytotoxicities and antiviral activities against influenza A/Puerto Rico/8/34 virus in MDCK cells. The compounds showed low toxicity and some exhibited moderate antiviral activity. Molecular docking identified the M2 channel and polymerase basic protein 2 as potential targets. We observed that the antiviral activity of thiopyrano[2,3-b]quinolines is notably affected by both the nature and position of the substituent within the tetrazole ring, as well as the substituent within the benzene moiety of quinoline. These findings contribute to the further search for new antiviral agents against influenza A viruses among derivatives of thiopyrano[2,3-b]quinoline.
Collapse
Affiliation(s)
- Andrey V. Khramchikhin
- Department of Chemistry and Technology of Organic Nitrogen Compounds, Saint Petersburg State Institute of Technology (Technical University), 26 Moskovsky Avenue, 190013 St. Petersburg, Russia; (A.V.K.); (M.A.S.)
| | - Mariya A. Skryl’nikova
- Department of Chemistry and Technology of Organic Nitrogen Compounds, Saint Petersburg State Institute of Technology (Technical University), 26 Moskovsky Avenue, 190013 St. Petersburg, Russia; (A.V.K.); (M.A.S.)
- Saint Petersburg Federal Research Center of the Russian Academy of Sciences (SPC RAS), 39, 14th Line, 199178 St. Petersburg, Russia
| | - Maxim A. Gureev
- Center of Bio- and Chemoinformatics, I. M. Sechenov First Moscow State Medical University, 8, Trubetskaya Street, Bld. 2, 119991 Moscow, Russia; (M.A.G.); (Y.B.P.)
- St. Petersburg School of Physics, Mathematics and Computer Science, HSE University, 16, Soyuza Pechatnikov Str., 190008 St. Petersburg , Russia
| | - Vladimir V. Zarubaev
- Saint Petersburg Pasteur Research Institute of Epidemiology and Microbiology, 14 Mira Street, 197101 St. Petersburg, Russia; (V.V.Z.); (I.L.E.); (P.A.I.)
| | - Iana L. Esaulkova
- Saint Petersburg Pasteur Research Institute of Epidemiology and Microbiology, 14 Mira Street, 197101 St. Petersburg, Russia; (V.V.Z.); (I.L.E.); (P.A.I.)
| | - Polina A. Ilyina
- Saint Petersburg Pasteur Research Institute of Epidemiology and Microbiology, 14 Mira Street, 197101 St. Petersburg, Russia; (V.V.Z.); (I.L.E.); (P.A.I.)
| | - Oussama Abdelhamid Mammeri
- Chemical Analysis and Materials Research Center, St. Petersburg State University, 26, Universitetskii Prospect, Petergof, 198504 St. Petersburg, Russia;
| | - Dar’ya V. Spiridonova
- Research Park, St. Petersburg State University, 26, Universitetskaya Emb. 7/9, 199034 St. Petersburg, Russia;
| | - Yuri B. Porozov
- Center of Bio- and Chemoinformatics, I. M. Sechenov First Moscow State Medical University, 8, Trubetskaya Street, Bld. 2, 119991 Moscow, Russia; (M.A.G.); (Y.B.P.)
- St. Petersburg School of Physics, Mathematics and Computer Science, HSE University, 16, Soyuza Pechatnikov Str., 190008 St. Petersburg , Russia
| | - Vladimir A. Ostrovskii
- Department of Chemistry and Technology of Organic Nitrogen Compounds, Saint Petersburg State Institute of Technology (Technical University), 26 Moskovsky Avenue, 190013 St. Petersburg, Russia; (A.V.K.); (M.A.S.)
- Saint Petersburg Federal Research Center of the Russian Academy of Sciences (SPC RAS), 39, 14th Line, 199178 St. Petersburg, Russia
| |
Collapse
|
9
|
Bai X, Xi S, Chen G, Fan X, Wang K, Li Y, Zhao Y, Wang W, Tian Y. Multicenter, randomized controlled, open label evaluation of the efficacy and safety of arbidol hydrochloride tablets in the treatment of influenza-like cases. BMC Infect Dis 2023; 23:585. [PMID: 37674112 PMCID: PMC10483848 DOI: 10.1186/s12879-023-08570-9] [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: 02/20/2023] [Accepted: 08/28/2023] [Indexed: 09/08/2023] Open
Abstract
OBJECTIVE To study the efficacy and safety of arbidol hydrochloride tablets as a treatment for influenza-like diseases. METHODS In this multicenter, randomized, controlled, open label study, a total of 412 influenza-like cases were collected from 14 hospitals in seven regions of Hebei Province from September 2021 to March 2022. Patients were randomly divided into two groups. The control group (n = 207) were administered oseltamivir phosphate capsules for five days and the experimental group (n = 205) were administered arbidol hydrochloride tablets for five days. The primary endpoint was the time to normal body temperature, and the secondary endpoints included the time to remission of influenza symptoms, incidence of influenza-like complications, and incidence of adverse reactions. RESULTS Before treatment, there was no significant difference between the two groups in general conditions, blood routine, body temperature, or symptom severity. After treatment, there was no significant difference between the groups in the mean time to fever remission (59.24 h ± 25.21 vs. 61.05 h ± 29.47) or the mean time to remission of influenza symptoms (57.31 h ± 30.19 vs. 62.02 h ± 32.08). Survival analyses using Log-rank and Wilcoxon bilateral tests showed that there was no significant difference in fever relief time or influenza symptom relief time between the two groups. Regarding the incidence of complications and adverse events, there was only one case of tracheitis, one case of nausea, one case of vomiting, and one case of dizziness in the control group. In the experimental group, there was one case of nausea, one case of vomiting, and one case of drowsiness. In addition, one patient in the control group was hospitalized for urinary calculi. CONCLUSION There was no significant difference between the patients with influenza-like cases treated with arbidol hydrochloride tablets and those treated with oseltamivir phosphate capsules. Further, the patients treated with arbidol hydrochloride tablets had fewer adverse reactions, and thus, the tablets were safe to use.
Collapse
Affiliation(s)
- Xinfeng Bai
- Emergency Department, Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, 050051, Hebei, China
| | - Suya Xi
- Hebei Chest Hospital, Shijiazhuang, Hebei, China
| | - Guiyan Chen
- Chengde Central Hospital, Chengde, Hebei, China
| | | | - Kaiwei Wang
- The Sixth People's Hospital of Hengshui, Hengshui, Hebei, China
| | - Yong Li
- Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yang Zhao
- Qinhuangdao Traditional Chinese Medicine Hospital, Qinhuangdao, Hebei, China
| | - Weizhan Wang
- Harrison International Peace Hospital, Hengshui, Hebei, China
| | - Yingping Tian
- Emergency Department, Second Hospital of Hebei Medical University, No. 215 Heping West Road, Xinhua District, Shijiazhuang, 050051, Hebei, China.
| |
Collapse
|
10
|
Bostanghadiri N, Ziaeefar P, Mofrad MG, Yousefzadeh P, Hashemi A, Darban-Sarokhalil D. COVID-19: An Overview of SARS-CoV-2 Variants-The Current Vaccines and Drug Development. BIOMED RESEARCH INTERNATIONAL 2023; 2023:1879554. [PMID: 37674935 PMCID: PMC10480030 DOI: 10.1155/2023/1879554] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/08/2023]
Abstract
The world is presently in crisis facing an outbreak of a health-threatening microorganism known as COVID-19, responsible for causing uncommon viral pneumonia in humans. The virus was first reported in Wuhan, China, in early December 2019, and it quickly became a global concern due to the pandemic. Challenges in this regard have been compounded by the emergence of several variants such as B.1.1.7, B.1.351, P1, and B.1.617, which show an increase in transmission power and resistance to therapies and vaccines. Ongoing researches are focused on developing and manufacturing standard treatment strategies and effective vaccines to control the pandemic. Despite developing several vaccines such as Pfizer/BioNTech and Moderna approved by the U.S. Food and Drug Administration (FDA) and other vaccines in phase 4 clinical trials, preventive measures are mandatory to control the COVID-19 pandemic. In this review, based on the latest findings, we will discuss different types of drugs as therapeutic options and confirmed or developing vaccine candidates against SARS-CoV-2. We also discuss in detail the challenges posed by the variants and their effect on therapeutic and preventive interventions.
Collapse
Affiliation(s)
- Narjess Bostanghadiri
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Pardis Ziaeefar
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morvarid Golrokh Mofrad
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Parsa Yousefzadeh
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Hashemi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Davood Darban-Sarokhalil
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Kang Y, Shi Y, Xu S. Arbidol: The current demand, strategies, and antiviral mechanisms. Immun Inflamm Dis 2023; 11:e984. [PMID: 37647451 PMCID: PMC10461429 DOI: 10.1002/iid3.984] [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: 08/31/2022] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 09/01/2023] Open
Abstract
BACKGROUND High morbidity and mortality of influenza virus infection have made it become one of the most lethal diseases threatening public health; the lack of drugs with strong antiviral activity against virus strains exacerbates the problem. METHODS Two independent researchers searched relevant studies using Embase, PubMed, Web of Science, Google Scholar, and MEDLINE databases from its inception to December 2022. RESULTS Based on the different antiviral mechanisms, current antiviral strategies can be mainly classified into virus-targeting approaches such as neuraminidase inhibitors, matrix protein 2 ion channel inhibitors, polymerase acidic protein inhibitors and other host-targeting antivirals. However, highly viral gene mutation has underscored the necessity of novel antiviral drug development. Arbidol (ARB) is a Russian-made indole-derivative small molecule licensed in Russia and China for the prevention and treatment of influenza and other respiratory viral infections. ARB also has inhibitory effects on many other viruses such as severe acute respiratory syndrome coronavirus 2, Coxsackie virus, respiratory syncytial virus, Hantaan virus, herpes simplex virus, and hepatitis B and C viruses. ARB is a promising drug which can not only exert activity against virus at different steps of virus replication cycle, but also directly target on hosts before infection to prevent virus invasion. CONCLUSION ARB is a broad-spectrum antiviral drug that inhibits several viruses in vivo and in vitro, with high safety profile and low resistance; the antiviral mechanisms of ARB deserve to be further explored and more high-quality clinical studies are required to establish the efficacy and safety of ARB.
Collapse
Affiliation(s)
- Yue Kang
- Jiangsu Key Laboratory of NeurodegenerationSchool of Pharmacy, Nanjing University of Chinese MedicineNanjingJiangsuChina
| | - Yin Shi
- Department of PharmacyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| | - Silu Xu
- Department of PharmacyJiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingJiangsuChina
| |
Collapse
|
12
|
Novel [1,2,4]triazolo[3,4- b][1,3,4]thiadiazine and [1,2,4]triazolo[3,4- b][1,3,4]thiadiazepine Derivatives: Synthesis, Anti-Viral In Vitro Study and Target Validation Activity. Molecules 2022; 27:molecules27227940. [PMID: 36432042 PMCID: PMC9694146 DOI: 10.3390/molecules27227940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
This study of the interaction system of binucleophilic 3-substituted 4-amino-4H-1,2,4-triazole-5-thiols and 3-phenyl-2-propynal made it possible to develop a new approach to synthesis of such isomeric classes as 7-benzylidene-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazine and 8-phenyl-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazepine. Among the 20 compounds studied in vitro against influenza A/Puerto Rico/8/34 (H1N1) virus, half of them demonstrated selectivity index (SI) of 10 or higher and one of them (4-((3-phenylprop-2-yn-1-yl)amino)-4H-1,2,4-triazole-3-thiol) possessed the highest (SI > 300). Docking results and values showed that the preferred interactant for our ligands was M2 proton channel of the influenza A virus. Protein-ligand interactions modeling showed that the aliphatic moiety of ligands could negatively regulate target activity level.
Collapse
Key Words
- [1,2,4]triazolo[3,4-b][1,3,4]thiadiazepines
- [1,2,4]triazolo[3,4-b][1,3,4]thiadiazines
- annelated heterocycles
- anti-viral activity
- drug design
- influenza virus
Collapse
|
13
|
Abstract
Lassa Fever (LF) is a viral hemorrhagic fever endemic in West Africa. LF begins with flu-like symptoms that are difficult to distinguish from other common endemic diseases such as malaria, dengue, and yellow fever making it hard to diagnose clinically. Availability of a rapid diagnostic test and other serological and molecular assays facilitates accurate diagnosis of LF. Lassa virus therapeutics are currently in different stages of preclinical development. Arevirumab, a cocktail of monoclonal antibodies, demonstrates a great safety and efficacy profile in non-human primates. Major efforts have been made in the development of a Lassa virus vaccine. Two vaccine candidates, MeV-NP and pLASV-GPC are undergoing evaluation in phase I clinical trials.
Collapse
Affiliation(s)
- Lilia I Melnik
- Department of Microbiology and Immunology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70118, USA.
| |
Collapse
|
14
|
Li Z, Li T, Liu M, Ivanovic T. Hemagglutinin Stability Determines Influenza A Virus Susceptibility to a Broad-Spectrum Fusion Inhibitor Arbidol. ACS Infect Dis 2022; 8:1543-1552. [PMID: 35819162 PMCID: PMC9810120 DOI: 10.1021/acsinfecdis.2c00178] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Understanding mechanisms of resistance to antiviral inhibitors can reveal nuanced features of targeted viral mechanisms and, in turn, lead to improved strategies for inhibitor design. Arbidol is a broad-spectrum antiviral that binds to and prevents the fusion-associated conformational changes in the trimeric influenza A virus (IAV) hemagglutinin (HA). The rate-limiting step during the HA-mediated membrane fusion is the release of the hydrophobic fusion peptides from a conserved pocket on HA. Here, we investigated how destabilizing or stabilizing mutations in or near the fusion peptide affect viral sensitivity to Arbidol. The degree of sensitivity was proportional to the extent of fusion-peptide stability on the prefusion HA: stabilized mutants were more sensitive, and destabilized ones were resistant to Arbidol. Single-virion membrane fusion experiments for representative wild-type (WT) and mutant viruses demonstrated that resistance is a direct consequence of fusion-peptide destabilization not requiring reduced Arbidol binding to HA. Our results support the model whereby the probability of individual HAs extending to engage the target membrane is determined by the composite of two critical forces: a "tug" on the fusion peptide by HA rearrangements near the Arbidol binding site and the key interactions stabilizing the fusion peptide in the prefusion pocket. Arbidol increases and destabilizing mutations decrease the free-energy cost for fusion-peptide release, accounting for the observed resistance. Our findings have broad implications for fusion inhibitor design, viral mechanisms of resistance, and our basic understanding of HA-mediated membrane fusion.
Collapse
|
15
|
Drug Repurposing for COVID-19: A Review and a Novel Strategy to Identify New Targets and Potential Drug Candidates. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27092723. [PMID: 35566073 PMCID: PMC9099573 DOI: 10.3390/molecules27092723] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/14/2022] [Accepted: 04/21/2022] [Indexed: 02/01/2023]
Abstract
In December 2019, the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) was first identified in the province of Wuhan, China. Since then, there have been over 400 million confirmed cases and 5.8 million deaths by COVID-19 reported worldwide. The urgent need for therapies against SARS-CoV-2 led researchers to use drug repurposing approaches. This strategy allows the reduction in risks, time, and costs associated with drug development. In many cases, a repurposed drug can enter directly to preclinical testing and clinical trials, thus accelerating the whole drug discovery process. In this work, we will give a general overview of the main developments in COVID-19 treatment, focusing on the contribution of the drug repurposing paradigm to find effective drugs against this disease. Finally, we will present our findings using a new drug repurposing strategy that identified 11 compounds that may be potentially effective against COVID-19. To our knowledge, seven of these drugs have never been tested against SARS-CoV-2 and are potential candidates for in vitro and in vivo studies to evaluate their effectiveness in COVID-19 treatment.
Collapse
|
16
|
Broad-Spectrum Activity of Small Molecules Acting against Influenza a Virus: Biological and Computational Studies. Pharmaceuticals (Basel) 2022; 15:ph15030301. [PMID: 35337099 PMCID: PMC8952214 DOI: 10.3390/ph15030301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 01/25/2023] Open
Abstract
Influenza still represents a problematic disease, involving millions of people every year and causing hundreds of thousands of deaths. Only a few drugs are clinically available. The search for an effective weapon is still ongoing. In this scenario, we recently identified new drug-like compounds with antiviral activity toward two A/H1N1 Influenza virus strains, which were demonstrated to interfere with the processes mediated by hemagglutinin (HA). In the present work, the compound’s ability to act against the A/H3N2 viral strain has been evaluated in hemagglutination inhibition (HI) assays. Two of the five tested compounds were also active toward the A/H3N2 Influenza virus. To validate the scaffold activity, analogue compounds of two broad-spectrum molecules were selected and purchased for HI testing on both A/H1N1 and A/H3N2 Influenza viruses. Forty-three compounds were tested, and four proved to be active toward all three viral strains. A computational study has been carried out to depict the HA binding process of the most interesting compounds.
Collapse
|
17
|
Beheshtirouy S, Khani E, Khiali S, Entezari-Maleki T. Investigational antiviral drugs for the treatment of COVID-19 patients. Arch Virol 2022; 167:751-805. [PMID: 35138438 DOI: 10.1007/s00705-022-05368-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/15/2021] [Indexed: 12/27/2022]
Abstract
In the current pandemic of coronavirus disease 2019 (COVID-19), antiviral drugs are at the center of attention because of their critical role against severe acute respiratory disease syndrome coronavirus 2 (SARS-CoV-2). In addition to designing new antivirals against SARS-COV-2, a drug repurposing strategy is a practical approach for treating COVID-19. A brief insight about antivirals would help clinicians to choose the best medication for the treatment of COVID-19. In this review, we discuss both novel and repurposed investigational antivirals, focusing on in vitro, in vivo, and clinical trial studies.
Collapse
Affiliation(s)
- Samineh Beheshtirouy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elnaz Khani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sajad Khiali
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Taher Entezari-Maleki
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran. .,Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
18
|
Ul'yanovskii NV, Kosyakov DS, Sypalov SA, Varsegov IS, Shavrina IS, Lebedev AT. Antiviral drug Umifenovir (Arbidol) in municipal wastewater during the COVID-19 pandemic: Estimated levels and transformation. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 805:150380. [PMID: 34818770 PMCID: PMC8451976 DOI: 10.1016/j.scitotenv.2021.150380] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/12/2021] [Accepted: 09/12/2021] [Indexed: 05/04/2023]
Abstract
An indole derivative umifenovir (Arbidol) is one of the most widely used antiviral drugs for the prevention and treatment of COVID-19 and some other viral infections. The purpose of the present study was to shed light on the transformation processes of umifenovir in municipal wastewater, including disinfection with active chlorine, as well as to assess the levels of the antiviral drug and its metabolites entering and accumulating in natural reservoirs under conditions of the SARS-CoV-2 pandemic. The combination of high-performance liquid chromatography with electrospray ionization high-resolution mass-spectrometry and inductively coupled plasma mass spectrometry was used for tentative identification and quantification of umifenovir and its transformation products in model reaction mixtures and real samples of wastewater, river water, biological sludge and bottom sediments taken at the wastewater treatment plant in Arkhangelsk, a large cultural and industrial center at the Russian North. Laboratory experiments allowed identifying fifteen bromine-containing transformation products, forming at the initial stages of the chlorination and fourteen classic volatile and semi volatile disinfection by-products with bromoform as the dominant one. Chlorinated derivatives are only the minor disinfection by-products forming by substitution of alkylamine group in the aromatic ring. The schemes of umifenovir transformation in reactions with dissolved oxygen and sodium hypochlorite are proposed. Two established primary transformation products formed by oxidation of the thioether group to sulfoxide and elimination of thiophenol were detected in noticeable concentrations in the wastewater together with their precursor. The level of umifenovir reached 1.3 mg kg-1 in the sludge and municipal wastewater treat contained 1 μg L-1 of that drug, while its removal during biological wastewater treatment was about 40%. Pronounced accumulation of umifenovir and its transformation products in biological sludge and bottom sediments of natural reservoirs may be a source of the future secondary pollution of the environment.
Collapse
Affiliation(s)
- Nikolay V Ul'yanovskii
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation
| | - Dmitry S Kosyakov
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation.
| | - Sergey A Sypalov
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation
| | - Ilya S Varsegov
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation
| | - Irina S Shavrina
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation
| | - Albert T Lebedev
- Core Facility Center 'Arktika', Northern (Arctic) Federal University, Arkhangelsk 163002, Russian Federation; Department of Chemistry, M. V. Lomonosov Moscow State University, Leninskie Gory 1/3, Moscow 119991, Russian Federation
| |
Collapse
|
19
|
Yang J, Liu S. Influenza Virus Entry inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:123-135. [DOI: 10.1007/978-981-16-8702-0_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
20
|
Therapeutic options in coronavirus treatment. CORONAVIRUS DRUG DISCOVERY 2022. [PMCID: PMC9217689 DOI: 10.1016/b978-0-323-85156-5.00021-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This chapter details the various therapeutic options available for the treatment of the novel coronavirus, SARS-CoV-2, that has brought the world to a standstill. As at 3.53 CEST, June 28, 2020, WHO reported 9,843,073 confirmed cases of COVID-19, with a death toll of 495,760. The rate of the spread of this disease is alarming posing serious threat to the world healthcare system. Clinical investigations and research are on the way for the development of vaccines or antiviral drugs. Despite this effort, no medication has been found to be very effective for its treatment. In this chapter, emphasis was laid on the need for repurposing of antiviral drugs to combat COVID-19 along with other alternatives such as convalescent plasma therapy and exploitation of drugs from medicinal plants and other natural resources.
Collapse
|
21
|
Amani B, Amani B, Zareei S, Zareei M. Efficacy and safety of arbidol (umifenovir) in patients with COVID-19: A systematic review and meta-analysis. Immun Inflamm Dis 2021; 9:1197-1208. [PMID: 34347937 PMCID: PMC8426686 DOI: 10.1002/iid3.502] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 07/10/2021] [Accepted: 07/16/2021] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To provide the latest evidence for the efficacy and safety of arbidol (umifenovir) in COVID-19 treatment. METHODS A literature systematic search was carried out in PubMed, Cochrane Library, Embase, and medRxiv up to May 2021. The Cochrane risk of bias tool and Newcastle-Ottawa scale were used to assess the quality of included studies. Meta-analysis was performed using RevMan 5.3. RESULTS Sixteen studies were met the inclusion criteria. No significant difference was observed between arbidol and non-antiviral treatment groups neither for primary outcomes, including the negative rate of PCR (NR-PCR) on Day 7 (risk ratio [RR]: 0.94; 95% confidence interval (CI): 0.78-1.14) and Day 14 (RR: 1.10; 95% CI: 0.96-1.25), and PCR negative conversion time (PCR-NCT; mean difference [MD]: 0.74; 95% CI: -0.87 to 2.34), nor secondary outcomes (p > .05). However, arbidol was associated with higher adverse events (RR: 2.24; 95% CI: 1.06-4.73). Compared with lopinavir/ritonavir, arbidol showed better efficacy for primary outcomes (p < .05). Adding arbidol to lopinavir/ritonavir also led to better efficacy in terms of NR-PCR on Day 7 and PCR-NCT (p < .05). There was no significant difference between arbidol and chloroquine in primary outcomes (p > .05). No remarkable therapeutic effect was observed between arbidol and other agents (p > .05). CONCLUSION The present meta-analysis showed no significant benefit of using arbidol compared with non-antiviral treatment or other therapeutic agents against COVID-19 disease. High-quality studies are needed to establish the efficacy and safety of arbidol for COVID-19.
Collapse
Affiliation(s)
- Behnam Amani
- Department of Health Management and Economics, School of Public HealthTehran University of Medical SciencesTehranIran
| | - Bahman Amani
- Health Management and Economics Research Center, Health Management Research InstituteIran University of Medical SciencesTehranIran
| | - Sara Zareei
- Department of Cell & Molecular Biology, Faculty of Biological SciencesKharazmi UniversityTehranIran
| | - Mahsa Zareei
- Department of Health Services Management, School of Health Management and Information SciencesIran University of Medical SciencesTehranIran
| |
Collapse
|
22
|
Akter R, Rahman MH, Bhattacharya T, Kaushik D, Mittal V, Parashar J, Kumar K, Kabir MT, Tagde P. Novel coronavirus pathogen in humans and animals: an overview on its social impact, economic impact, and potential treatments. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:68071-68089. [PMID: 34664166 PMCID: PMC8523003 DOI: 10.1007/s11356-021-16809-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/25/2021] [Indexed: 04/15/2023]
Abstract
In the light of thousands of infections and deaths, the World Health Organization (WHO) has declared the outbreak of coronavirus disease (COVID-19) a worldwide pandemic. It has spread to about 22 million people worldwide, with a total of 0.45 million expiries, limiting the movement of most people worldwide in the last 6 months. However, COVID-19 became the foremost health, economic, and humanitarian challenge of the twenty-first century. Measures intended to curb the pandemic of COVID-19 included travel bans, lockdowns, and social distances through shelter orders, which will further stop human activities suddenly and eventually impact the world and the national economy. The viral disease is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). After SARS-CoV-2 virus and Middle East respiratory syndrome (MERS)-related CoV, COVID-19 is the third most significant lethal disease to humans. According to WHO, COVID-19 mortality exceeded that of SARS and MERS since COVID-19 was declared an international public health emergency. Genetic sequencing has recently established that COVID-19 is close to SARS-CoV and bat coronavirus which has not yet been recognized as the key cause of this pandemic outbreak, its transmission, and human pathogen mechanism. This review focuses on a brief introduction of novel coronavirus pathogens, including coronavirus in humans and animals, its taxonomic classification, symptoms, pathogenicity, social impact, economic impact, and potential treatment therapy for COVID-19.
Collapse
Affiliation(s)
- Rokeya Akter
- Department of Pharmacy, Jagannath University, Sadarghat, Dhaka-1100, Bangladesh
| | - Md Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka-1213, Bangladesh.
| | - Tanima Bhattacharya
- School of Chemistry & Chemical Engineering, Hubei University, Wuhan, People's Republic of China, 430062
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Jatin Parashar
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Kuldeep Kumar
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Md Tanvir Kabir
- Department of Pharmacy, Brac University, 66 Mohakhali, Dhaka, 1212, Bangladesh
| | - Priti Tagde
- Bhabha Pharmacy Research Institute, Bhabha University, Bhopal, M.P, India
| |
Collapse
|
23
|
Moraes TFS, Ferraz AC, da Cruz Nizer WS, Tótola AH, Soares DBS, Duarte LP, Vieira-Filho SA, Magalhães CLB, de Magalhães JC. A methanol extract and N,N-dimethyltryptamine from Psychotria viridis Ruiz & Pav. inhibit Zika virus infection in vitro. Arch Virol 2021; 166:3275-3287. [PMID: 34536126 DOI: 10.1007/s00705-021-05230-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/19/2021] [Indexed: 10/20/2022]
Abstract
Zika virus (ZIKV) is a public health problem due to its association with serious fetal and neurological complications and the lack of antiviral agents and licensed vaccines against this virus. Surveillance studies have alerted about the potential occurrence of a new South American epidemic episode due to the recent circulation of an African ZIKV strain detected in Brazil. Therefore, it is essential to discover antiviral agents, including natural substances, that are capable of neutralizing the action of ZIKV. Several Psychotria species have antimicrobial and anti-inflammatory properties. Thus, a methanol extract and dimethyltryptamine from Psychotria viridis were evaluated for their ability to inhibit ZIKV infection in vitro by measuring the effective concentration that protects 50% of cells and investigating their possible mechanisms of action. The tested samples showed antiviral activity against ZIKV. The extract showed virucidal activity, affecting viral and non-cellular elements, inactivating the virus before infection or when it becomes extracellular after the second cycle of infection. It was also observed that both extract and dimethyltryptamine could inhibit the virus at intracellular stages of the viral cycle. In addition to dimethyltryptamine, it is believed that other compounds also contribute to the promising virucidal effect observed for the methanol extract. To our knowledge, this is the first report of the activity of a methanolic extract and dimethyltryptamine from Psychotria viridis against cellular ZIKV infection. These two samples, extracted from natural sources, are potential candidates for use as antiviral drugs to inhibit ZIKV infections.
Collapse
Affiliation(s)
- Thaís F S Moraes
- Department of Chemistry, Biotechnology and Bioprocess Engineering, Universidade Federal de São João del-Rei, Rodovia MG 443, Km7, Ouro Branco, MG, 36420-000, Brazil.,Department of Microbiology, Universidade Federal de Minas Gerais, Avenida Antônio Carlos, Belo Horizonte, MG, 31270-901, Brazil
| | - Ariane C Ferraz
- Department of Chemistry, Biotechnology and Bioprocess Engineering, Universidade Federal de São João del-Rei, Rodovia MG 443, Km7, Ouro Branco, MG, 36420-000, Brazil.,Department of Biological Sciences, Universidade Federal de Ouro Preto, Campus Morro do Cruzeiro, Ouro Prêto, MG, 35400-000, Brazil
| | - Waleska S da Cruz Nizer
- Department of Chemistry, Biotechnology and Bioprocess Engineering, Universidade Federal de São João del-Rei, Rodovia MG 443, Km7, Ouro Branco, MG, 36420-000, Brazil.,Department of Health Sciences, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - Antônio H Tótola
- Department of Chemistry, Biotechnology and Bioprocess Engineering, Universidade Federal de São João del-Rei, Rodovia MG 443, Km7, Ouro Branco, MG, 36420-000, Brazil
| | - Débora B S Soares
- Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Lucienir P Duarte
- Department of Chemistry, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Sidney A Vieira-Filho
- Department of Pharmacy, Pharmacy's School, Universidade Federal de Ouro Preto, Campus Morro do Cruzeiro, Ouro Preto, MG, 35400-000, Brazil
| | - Cintia L B Magalhães
- Department of Biological Sciences, Universidade Federal de Ouro Preto, Campus Morro do Cruzeiro, Ouro Prêto, MG, 35400-000, Brazil
| | - José C de Magalhães
- Department of Chemistry, Biotechnology and Bioprocess Engineering, Universidade Federal de São João del-Rei, Rodovia MG 443, Km7, Ouro Branco, MG, 36420-000, Brazil.
| |
Collapse
|
24
|
Choudhary V, Gupta A, Sharma R, Parmar HS. Therapeutically effective covalent spike protein inhibitors in treatment of SARS-CoV-2. JOURNAL OF PROTEINS AND PROTEOMICS 2021; 12:257-270. [PMID: 34539131 PMCID: PMC8440732 DOI: 10.1007/s42485-021-00074-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 [coronavirus disease 2019] has resulted in over 204,644,849 confirmed cases and over 4,323,139 deaths throughout the world as of 12 August 2021, a total of 4,428,168,759 vaccine doses have been administered. The lack of potentially effective drugs against the virus is making the situation worse and dangerous. Numerous forces are working on finding an effective treatment against the virus but it is believed that a de novo drug would take several months even if huge financial support is provided. The only solution left with is drug repurposing that would not only provide effective therapy with the already used clinical drugs, but also save time and cost of the de novo drug discovery. The initiation of the COVID-19 infection starts with the attachment of spike glycoprotein of SARS-CoV-2 to the host receptor. Hence, the inhibition of the binding of the virus to the host membrane and the entry of the viral particle into the host cell are one of the main therapeutic targets. This paper not only summarizes the structure and the mechanism of spike protein, but the main focus is on the potential covalent spike protein inhibitors.
Collapse
Affiliation(s)
- Vikram Choudhary
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road (Ring Road), Indore, 452001 Madhya Pradesh India
| | - Amisha Gupta
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road (Ring Road), Indore, 452001 Madhya Pradesh India
| | - Rajesh Sharma
- School of Pharmacy, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road (Ring Road), Indore, 452001 Madhya Pradesh India
| | - Hamendra Singh Parmar
- School of Biotechnology, Devi Ahilya Vishwavidyalaya, Takshila Campus, Khandwa Road, Indore, 452001 Madhya Pradesh India
| |
Collapse
|
25
|
Dowarah J, Marak BN, Yadav UCS, Singh VP. Potential drug development and therapeutic approaches for clinical intervention in COVID-19. Bioorg Chem 2021; 114:105016. [PMID: 34144277 PMCID: PMC8143914 DOI: 10.1016/j.bioorg.2021.105016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 05/20/2021] [Indexed: 01/25/2023]
Abstract
While the vaccination is now available to many countries and will slowly dissipate to others, effective therapeutics for COVID-19 is still illusive. The SARS-CoV-2 pandemic has posed an unprecedented challenge to researchers, scientists, and clinicians and affected the wellbeing of millions of people worldwide. Since the beginning of the pandemic, a multitude of existing anti-viral, antibiotic, antimalarial, and anticancer drugs have been tested, and some have shown potency in the treatment and management of COVID-19, albeit others failed to leave any positive impact and a few also became controversial as they showed mixed clinical outcomes. In the present article, we have brought together some of the candidate therapeutic drugs being repurposed or used in the clinical trials and discussed their clinical efficacy and safety for COVID-19.
Collapse
Affiliation(s)
- Jayanta Dowarah
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | - Brilliant N Marak
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India
| | | | - Ved Prakash Singh
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India; Department of Industrial Chemistry, School of Physical Sciences, Mizoram University, Aizawl 796004, Mizoram, India.
| |
Collapse
|
26
|
Leneva I, Kartashova N, Poromov A, Gracheva A, Korchevaya E, Glubokova E, Borisova O, Shtro A, Loginova S, Shchukina V, Khamitov R, Faizuloev E. Antiviral Activity of Umifenovir In Vitro against a Broad Spectrum of Coronaviruses, Including the Novel SARS-CoV-2 Virus. Viruses 2021; 13:1665. [PMID: 34452529 PMCID: PMC8402645 DOI: 10.3390/v13081665] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/12/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022] Open
Abstract
An escalating pandemic of the novel SARS-CoV-2 virus is impacting global health, and effective antivirals are needed. Umifenovir (Arbidol) is an indole-derivative molecule, licensed in Russia and China for prophylaxis and treatment of influenza and other respiratory viral infections. It has been shown that umifenovir has broad spectrum activity against different viruses. We evaluated the sensitivity of different coronaviruses, including the novel SARS-CoV-2 virus, to umifenovir using in vitro assays. Using a plaque assay, we revealed an antiviral effect of umifenovir against seasonal HCoV-229E and HCoV-OC43 coronaviruses in Vero E6 cells, with estimated 50% effective concentrations (EC50) of 10.0 ± 0.5 µM and 9.0 ± 0.4 µM, respectively. Umifenovir at 90 µM significantly suppressed plaque formation in CMK-AH-1 cells infected with SARS-CoV. Umifenovir also inhibited the replication of SARS-CoV-2 virus, with EC50 values ranging from 15.37 ± 3.6 to 28.0 ± 1.0 µM. In addition, 21-36 µM of umifenovir significantly suppressed SARS-CoV-2 virus titers (≥2 log TCID50/mL) in the first 24 h after infection. Repurposing of antiviral drugs is very helpful in fighting COVID-19. A safe, pan-antiviral drug such as umifenovir could be extremely beneficial in combating the early stages of a viral pandemic.
Collapse
Affiliation(s)
- Irina Leneva
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Nadezhda Kartashova
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Artem Poromov
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Anastasiia Gracheva
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Ekaterina Korchevaya
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Ekaterina Glubokova
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Olga Borisova
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Anna Shtro
- Smorodintsev Research Institute of Influenza, 197376 Saint-Petersburg, Russia;
| | - Svetlana Loginova
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Veronika Shchukina
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| | - Ravil Khamitov
- International Biotechnology Center IBC “GENERIUM”, Volginsky Village, Petushinsky District, 601125 Vladimir, Russia;
| | - Evgeny Faizuloev
- Mechnikov Research Institute of Vaccines and Sera, 105064 Moscow, Russia; (N.K.); (A.P.); (A.G.); (E.K.); (E.G.); (O.B.); (S.L.); (V.S.); (E.F.)
| |
Collapse
|
27
|
Mohamadi Yarijani Z, Najafi H. Kidney injury in COVID-19 patients, drug development and their renal complications: Review study. Biomed Pharmacother 2021; 142:111966. [PMID: 34333286 PMCID: PMC8313500 DOI: 10.1016/j.biopha.2021.111966] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/15/2021] [Accepted: 07/23/2021] [Indexed: 01/08/2023] Open
Abstract
Since December 2019, the world was encountered a new disease called coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although SARS-CoV-2 initially causes lung damage, it also affects many other organs, including the kidneys, and on average, 5–23% of people with COVID-19 develop the symptoms of acute kidney injury (AKI), including elevated blood creatinine and urea, hematuria, proteinuria, and histopathological damages. The exact mechanism is unknown, but the researchers believe that SARS-CoV-2 directly and indirectly affects the kidneys. The direct pathway is by binding the virus to ACE2 receptor in the kidney, damage to cells, the renin-angiotensin system disturbances, activating coagulation pathways, and damaging the renal vascular endothelium. The initial evidence from studying the kidney tissue in postmortem patients is more in favor of the direct pathway. The indirect pathway is created by increased cytokines and cytokine storm, sepsis, circulatory disturbances, hypoxemia, as well as using the nephrotoxic drugs. Using renal tissue biopsy and autopsy in the patients with COVID-19, recent studies found evidence for a predominant indirect pathway in AKI induction by SARS-CoV-2. Besides, some studies showed that the degree of acute tubular injury (ATI) in autopsies from COVID-19 victims is milder compared to AKI degree. We review the mechanism of AKI induction and the renal side effects of the most common drugs used to treat COVID-19 after the overview of the latest findings on SARS-CoV-2 pathogenicity.
Collapse
Affiliation(s)
- Zeynab Mohamadi Yarijani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Houshang Najafi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
28
|
Chen R, Wang T, Song J, Pu D, He D, Li J, Yang J, Li K, Zhong C, Zhang J. Antiviral Drug Delivery System for Enhanced Bioactivity, Better Metabolism and Pharmacokinetic Characteristics. Int J Nanomedicine 2021; 16:4959-4984. [PMID: 34326637 PMCID: PMC8315226 DOI: 10.2147/ijn.s315705] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/11/2021] [Indexed: 12/11/2022] Open
Abstract
Antiviral drugs (AvDs) are the primary resource in the global battle against viruses, including the recent fight against corona virus disease 2019 (COVID-19). Most AvDs require multiple medications, and their use frequently leads to drug resistance, since they have poor oral bioavailability and low efficacy due to their low solubility/low permeability. Characterizing the in vivo metabolism and pharmacokinetic characteristics of AvDs may help to solve the problems associated with AvDs and enhance their efficacy. In this review of AvDs, we systematically investigated their structure-based metabolic reactions and related enzymes, their cellular pharmacology, and the effects of metabolism on AvD pharmacodynamics and pharmacokinetics. We further assessed how delivery systems achieve better metabolism and pharmacology of AvDs. This review suggests that suitable nanosystems may help to achieve better pharmacological activity and pharmacokinetic behavior of AvDs by altering drug metabolism through the utilization of advanced nanotechnology and appropriate administration routes. Notably, such AvDs as ribavirin, remdesivir, favipiravir, chloroquine, lopinavir and ritonavir have been confirmed to bind to the severe acute respiratory syndrome-like coronavirus (SARS-CoV-2) receptor and thus may represent anti-COVID-19 treatments. Elucidating the metabolic and pharmacokinetic characteristics of AvDs may help pharmacologists to identify new formulations with high bioavailability and efficacy and help physicians to better treat virus-related diseases, including COVID-19.
Collapse
Affiliation(s)
- Ran Chen
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Tingting Wang
- Biochemistry and Molecular Biology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jie Song
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Daojun Pu
- Pharmaceutical Institute, Southwest Pharmaceutical Limited Company, Chongqing, 400038, People's Republic of China
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jianjun Li
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jie Yang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Kailing Li
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, School of Pharmacy, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| |
Collapse
|
29
|
Rahman MM, Ahmed M, Islam MT, Khan MR, Sultana S, Maeesa SK, Hasan S, Hossain MA, Ferdous KS, Mathew B, Rauf A, Uddin MS. Nanotechnology-Based Approaches and Investigational Therapeutics against COVID-19. Curr Pharm Des 2021; 28:948-968. [PMID: 34218774 DOI: 10.2174/1381612827666210701150315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/30/2021] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a novel coronavirus which is currently responsible for the global pandemic since December 2019. This class of coronavirus has affected 217 countries around the world. Most of the countries have taken some non-remedial preventive actions like country lockdown, work from home, travel bans, and the most significant one is social isolation. Pharmacists, doctors, nurses, technologists, and all other healthcare professionals are playing a pivotal role during this pandemic. Unluckily, there is no specific drug that can treat patients who are confirmed with COVID-19, though favipiravir and remdesivir have appeared as favorable antiviral drugs. Some vaccines have already developed, and vaccination has started worldwide. Different nanotechnologies are in the developing stage in many countries for preventing SARS-COV-2 and treating COVID-19 conditions. In this article, we review the COVID-19 pandemic situation as well as the nanotechnology-based approaches and investigational therapeutics against COVID-19.
Collapse
Affiliation(s)
- Md Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Mohammad Touhidul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Md Robin Khan
- Bangladesh Reference Institute for Chemical Measurements, Dhaka. Bangladesh
| | - Sharifa Sultana
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Saila Kabir Maeesa
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Sakib Hasan
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Md Abid Hossain
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Kazi Sayma Ferdous
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka. Bangladesh
| | - Bijo Mathew
- Department of Pharmaceutical Chemistry, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi 682041, India
| | - Abdur Rauf
- Department of Chemistry, University of Swabi, Swabi, Anbar, Khyber Pakhtunkhwa. Bangladesh
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka. Bangladesh
| |
Collapse
|
30
|
Kim YJ, Venturini V, de la Torre JC. Progress in Anti-Mammarenavirus Drug Development. Viruses 2021; 13:v13071187. [PMID: 34206216 PMCID: PMC8310104 DOI: 10.3390/v13071187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/12/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022] Open
Abstract
Mammarenaviruses are prevalent pathogens distributed worldwide, and several strains cause severe cases of human infections with high morbidity and significant mortality. Currently, there is no FDA-approved antiviral drugs and vaccines against mammarenavirus and the potential treatment option is limited to an off-label use of ribavirin that shows only partial protective effect and associates with side effects. For the past few decades, extensive research has reported potential anti-mammarenaviral drugs and their mechanisms of action in host as well as vaccine candidates. This review describes current knowledge about mammarenavirus virology, progress of antiviral drug development, and technical strategies of drug screening.
Collapse
Affiliation(s)
- Yu-Jin Kim
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-J.K.); (V.V.)
| | - Victor Venturini
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-J.K.); (V.V.)
- Department of Biotechnology, Faculty of Experimental Sciences, Francisco de Vitoria University (UFV), Carretera Pozuelo-Majadahonda, Km 1,800, Pozuelo de Alarcón, 28223 Madrid, Spain
| | - Juan C. de la Torre
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA; (Y.-J.K.); (V.V.)
- Correspondence:
| |
Collapse
|
31
|
Chen Z, Cui Q, Caffrey M, Rong L, Du R. Small Molecule Inhibitors of Influenza Virus Entry. Pharmaceuticals (Basel) 2021; 14:ph14060587. [PMID: 34207368 PMCID: PMC8234048 DOI: 10.3390/ph14060587] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/13/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
Hemagglutinin (HA) plays a critical role during influenza virus receptor binding and subsequent membrane fusion process, thus HA has become a promising drug target. For the past several decades, we and other researchers have discovered a series of HA inhibitors mainly targeting its fusion machinery. In this review, we summarize the advances in HA-targeted development of small molecule inhibitors. Moreover, we discuss the structural basis and mode of action of these inhibitors, and speculate upon future directions toward more potent inhibitors of membrane fusion and potential anti-influenza drugs.
Collapse
Affiliation(s)
- Zhaoyu Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
| | - Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
- Correspondence: (L.R.); (R.D.); Tel.: +1-312-355-0203 (L.R.); +86-0531-89628505 (R.D.)
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Z.C.); (Q.C.)
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
- Correspondence: (L.R.); (R.D.); Tel.: +1-312-355-0203 (L.R.); +86-0531-89628505 (R.D.)
| |
Collapse
|
32
|
Ungogo MA, Mohammed M, Umar BN, Bala AA, Khalid GM. Review of pharmacologic and immunologic agents in the management of COVID-19. BIOSAFETY AND HEALTH 2021; 3:148-155. [PMID: 33458647 PMCID: PMC7796672 DOI: 10.1016/j.bsheal.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 01/01/2021] [Accepted: 01/07/2021] [Indexed: 12/15/2022] Open
Abstract
The novel coronavirus disease 2019 (COVID-19) is the third coronavirus outbreak in the last two decades. Emerging and re-emerging infections like COVID-19 pose serious challenges of the paucity of information and lack of specific cure or vaccines. This leaves utilisation of existing scientific data on related viral infections and repurposing relevant aetiologic and supportive therapies as the best control approach while novel strategies are developed and trialled. Many promising antiviral agents including lopinavir, ritonavir, remdesivir, umifenovir, darunavir, and oseltamivir have been repurposed and are currently trialled for the care for COVID-19 patients. Adjunct therapies for the management of symptoms and to provide support especially in severe and critically ill patients have also been identified. This review provides an appraisal of the current evidence for the rational use of frontline therapeutics in the management of COVID-19. It also includes updates regarding COVID-19 immunotherapy and vaccine development.
Collapse
Affiliation(s)
- Marzuq A Ungogo
- Department of Veterinary Pharmacology and Toxicology, Ahmadu Bello University, Zaria 810107, Kaduna State, Nigeria
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Mustapha Mohammed
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Penang City 11800, Pulau Pinang State, Malaysia
- Department of Clinical Pharmacy and Pharmacy Practice, Ahmadu Bello University, Zaria 810107, Kaduna State, Nigeria
| | - Bala N Umar
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria 810107, Kaduna State, Nigeria
| | - Auwal A Bala
- Department of Pharmacology, College of Medicine and Health Sciences, Federal University, Dutse 720231, Jigawa State, Nigeria
| | - Garba M Khalid
- Faculty of Pharmaceutical Sciences, Bayero University, Kano P.M.B. 3011, Kano State, Nigeria
- Department of Pharmaceutical Sciences, Università Degli Studi di Milano, Via G. Colombo, 71, Milano 20133, Italy
| |
Collapse
|
33
|
Clinical Management of COVID-19: A Review of Pharmacological Treatment Options. Pharmaceuticals (Basel) 2021; 14:ph14060520. [PMID: 34071185 PMCID: PMC8229327 DOI: 10.3390/ph14060520] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023] Open
Abstract
Since the outbreak and subsequent declaration of COVID-19 as a global pandemic in March 2020, concerted efforts have been applied by the scientific community to curtail the spread of the disease and find a cure. While vaccines constitute a vital part of the public health strategy to reduce the burden of COVID-19, the management of this disease will continue to rely heavily on pharmacotherapy. This study aims to provide an updated review of pharmacological agents that have been developed and/or repurposed for the treatment of COVID-19. To this end, a comprehensive literature search was conducted using the PubMed, Google Scholar, and LitCovid databases. Relevant clinical studies on drugs used in the management of COVID-19 were identified and evaluated in terms of evidence of efficacy and safety. To date, the FDA has approved three therapies for the treatment of COVID-19 Emergency Use Authorization: convalescent plasma, remdesivir, and casirivimab/imdevimab (REGN-COV2). Drugs such as lopinavir/ritonavir, umifenovir, favipiravir, anakinra, chloroquine, hydroxychloroquine, tocilizumab, interferons, tissue plasminogen activator, intravenous immunoglobulins, and nafamosat have been used off-label with mixed therapeutic results. Adjunctive administration of corticosteroids is also very common. The clinical experience with these approved and repurposed drugs is limited, and data on efficacy for the new indication are not strong. Overall, the response of the global scientific community to the COVID-19 pandemic has been impressive, as evident from the volume of scientific literature elucidating the molecular biology and pathophysiology of SARS-CoV-2 and the approval of three new drugs for clinical management. Reviewed studies have shown mixed data on efficacy and safety of the currently utilized drugs. The lack of standard treatment for COVID-19 has made it difficult to interpret results from most of the published studies due to the risk of attribution error. The long-term effects of drugs can only be assessed after several years of clinical experience; therefore, the efficacy and safety of current COVID-19 therapeutics should continue to be rigorously monitored as part of post-marketing studies.
Collapse
|
34
|
Bauso LV, Imbesi C, Irene G, Calì G, Bitto A. New Approaches and Repurposed Antiviral Drugs for the Treatment of the SARS-CoV-2 Infection. Pharmaceuticals (Basel) 2021; 14:503. [PMID: 34070359 PMCID: PMC8228036 DOI: 10.3390/ph14060503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/02/2021] [Accepted: 05/08/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the virus that causes coronavirus disease 2019 (COVID-19). The outbreak of this coronavirus was first identified in Wuhan (Hubei, China) in December 2019, and it was declared as pandemic by the World Health Organization (WHO) in March 2020. Today, several vaccines against SARS-CoV-2 have been approved, and some neutralizing monoclonal antibodies are being tested as therapeutic approaches for COVID-19 but, one of the key questions is whether both vaccines and monoclonal antibodies could be effective against infections by new SARS-CoV-2 variants. Nevertheless, there are currently more than 1000 ongoing clinical trials focusing on the use and effectiveness of antiviral drugs as a possible therapeutic treatment. Among the classes of antiviral drugs are included 3CL protein inhibitors, RNA synthesis inhibitors and other small molecule drugs which target the ability of SARS-COV-2 to interact with host cells. Considering the need to find specific treatment to prevent the emergent outbreak, the aim of this review is to explain how some repurposed antiviral drugs, indicated for the treatment of other viral infections, could be potential candidates for the treatment of COVID-19.
Collapse
Affiliation(s)
- Luana Vittoria Bauso
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (L.V.B.); (C.I.); (G.I.); (G.C.)
| | - Chiara Imbesi
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (L.V.B.); (C.I.); (G.I.); (G.C.)
- Laboratori Campisi, Corso Vittorio Emanuele 231, 96012 Avola, Italy
| | - Gasparo Irene
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (L.V.B.); (C.I.); (G.I.); (G.C.)
| | - Gabriella Calì
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (L.V.B.); (C.I.); (G.I.); (G.C.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy; (L.V.B.); (C.I.); (G.I.); (G.C.)
| |
Collapse
|
35
|
Xu J, Xue Y, Zhou R, Shi PY, Li H, Zhou J. Drug repurposing approach to combating coronavirus: Potential drugs and drug targets. Med Res Rev 2021; 41:1375-1426. [PMID: 33277927 PMCID: PMC8044022 DOI: 10.1002/med.21763] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/03/2020] [Accepted: 11/20/2020] [Indexed: 01/18/2023]
Abstract
In the past two decades, three highly pathogenic human coronaviruses severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus, and, recently, SARS-CoV-2, have caused pandemics of severe acute respiratory diseases with alarming morbidity and mortality. Due to the lack of specific anti-CoV therapies, the ongoing pandemic of coronavirus disease 2019 (COVID-19) poses a great challenge to clinical management and highlights an urgent need for effective interventions. Drug repurposing is a rapid and feasible strategy to identify effective drugs for combating this deadly infection. In this review, we summarize the therapeutic CoV targets, focus on the existing small molecule drugs that have the potential to be repurposed for existing and emerging CoV infections of the future, and discuss the clinical progress of developing small molecule drugs for COVID-19.
Collapse
Affiliation(s)
- Jimin Xu
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Yu Xue
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Hongmin Li
- Wadsworth Center, New York State Department of Health, Albany, New York, USA
- Department of Biomedical Sciences, School of Public Health, University at Albany, Albany, New York, USA
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
36
|
Disease-drug and drug-drug interaction in COVID-19: Risk and assessment. Biomed Pharmacother 2021; 139:111642. [PMID: 33940506 PMCID: PMC8078916 DOI: 10.1016/j.biopha.2021.111642] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/11/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is announced as a global pandemic in 2020. Its mortality and morbidity rate are rapidly increasing, with limited medications. The emergent outbreak of COVID-19 prompted by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) keeps spreading. In this infection, a patient's immune response plays pivotal role in the pathogenesis. This inflammatory factor was shown by its mediators that, in severe cases, reach the cytokine at peaks. Hyperinflammatory state may sparks significant imbalances in transporters and drug metabolic machinery, and subsequent alteration of drug pharmacokinetics may result in unexpected therapeutic response. The present scenario has accounted for the requirement for therapeutic opportunities to relive and overcome this pandemic. Despite the diminishing developments of COVID-19, there is no drug still approved to have significant effects with no side effect on the treatment for COVID-19 patients. Based on the evidence, many antiviral and anti-inflammatory drugs have been authorized by the Food and Drug Administration (FDA) to treat the COVID-19 patients even though not knowing the possible drug-drug interactions (DDI). Remdesivir, favipiravir, and molnupiravir are deemed the most hopeful antiviral agents by improving infected patient’s health. Dexamethasone is the first known steroid medicine that saved the lives of seriously ill patients. Some oligopeptides and proteins have also been using. The current review summarizes medication updates to treat COVID-19 patients in an inflammatory state and their interaction with drug transporters and drug-metabolizing enzymes. It gives an opinion on the potential DDI that may permit the individualization of these drugs, thereby enhancing the safety and efficacy.
Collapse
|
37
|
Hansen F, Feldmann H, Jarvis MA. Targeting Ebola virus replication through pharmaceutical intervention. Expert Opin Investig Drugs 2021; 30:201-226. [PMID: 33593215 DOI: 10.1080/13543784.2021.1881061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Introduction. The consistent emergence/reemergence of filoviruses into a world that previously lacked an approved pharmaceutical intervention parallels an experience repeatedly played-out for most other emerging pathogenic zoonotic viruses. Investment to preemptively develop effective and low-cost prophylactic and therapeutic interventions against viruses that have high potential for emergence and societal impact should be a priority.Areas covered. Candidate drugs can be characterized into those that interfere with cellular processes required for Ebola virus (EBOV) replication (host-directed), and those that directly target virally encoded functions (direct-acting). We discuss strategies to identify pharmaceutical interventions for EBOV infections. PubMed/Web of Science databases were searched to establish a detailed catalog of these interventions.Expert opinion. Many drug candidates show promising in vitro inhibitory activity, but experience with EBOV shows the general lack of translation to in vivo efficacy for host-directed repurposed drugs. Better translation is seen for direct-acting antivirals, in particular monoclonal antibodies. The FDA-approved monoclonal antibody treatment, Inmazeb™ is a success story that could be improved in terms of impact on EBOV-associated disease and mortality, possibly by combination with other direct-acting agents targeting distinct aspects of the viral replication cycle. Costs need to be addressed given EBOV emergence primarily in under-resourced countries.
Collapse
Affiliation(s)
- Frederick Hansen
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Michael A Jarvis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA.,School of Biomedical Sciences, University of Plymouth, Plymouth, Devon, UK.,The Vaccine Group, Ltd, Plymouth, Devon, UK
| |
Collapse
|
38
|
Herring S, Oda JM, Wagoner J, Kirchmeier D, O'Connor A, Nelson EA, Huang Q, Liang Y, DeWald LE, Johansen LM, Glass PJ, Olinger GG, Ianevski A, Aittokallio T, Paine MF, Fink SL, White JM, Polyak SJ. Inhibition of Arenaviruses by Combinations of Orally Available Approved Drugs. Antimicrob Agents Chemother 2021; 65:e01146-20. [PMID: 33468464 PMCID: PMC8097473 DOI: 10.1128/aac.01146-20] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 12/24/2020] [Indexed: 02/07/2023] Open
Abstract
Neglected diseases caused by arenaviruses such as Lassa virus (LASV) and filoviruses like Ebola virus (EBOV) primarily afflict resource-limited countries, where antiviral drug development is often minimal. Previous studies have shown that many approved drugs developed for other clinical indications inhibit EBOV and LASV and that combinations of these drugs provide synergistic suppression of EBOV, often by blocking discrete steps in virus entry. We hypothesize that repurposing of combinations of orally administered approved drugs provides effective suppression of arenaviruses. In this report, we demonstrate that arbidol, an approved influenza antiviral previously shown to inhibit EBOV, LASV, and many other viruses, inhibits murine leukemia virus (MLV) reporter viruses pseudotyped with the fusion glycoproteins (GPs) of other arenaviruses (Junin virus [JUNV], lymphocytic choriomeningitis virus [LCMV], and Pichinde virus [PICV]). Arbidol and other approved drugs, including aripiprazole, amodiaquine, sertraline, and niclosamide, also inhibit infection of cells by infectious PICV, and arbidol, sertraline, and niclosamide inhibit infectious LASV. Combining arbidol with aripiprazole or sertraline results in the synergistic suppression of LASV and JUNV GP-bearing pseudoviruses. This proof-of-concept study shows that arenavirus infection in vitro can be synergistically inhibited by combinations of approved drugs. This approach may lead to a proactive strategy with which to prepare for and control known and new arenavirus outbreaks.
Collapse
Affiliation(s)
- Shawn Herring
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jessica M Oda
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Jessica Wagoner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Delaney Kirchmeier
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Aidan O'Connor
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Elizabeth A Nelson
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Qinfeng Huang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Yuying Liang
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Minnesota, USA
| | - Lisa Evans DeWald
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | | | - Pamela J Glass
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Frederick, Maryland, USA
| | | | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Mary F Paine
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA
| | - Susan L Fink
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen J Polyak
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
39
|
Padhi AK, Seal A, Khan JM, Ahamed M, Tripathi T. Unraveling the mechanism of arbidol binding and inhibition of SARS-CoV-2: Insights from atomistic simulations. Eur J Pharmacol 2021; 894:173836. [PMID: 33387467 PMCID: PMC7773528 DOI: 10.1016/j.ejphar.2020.173836] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/16/2022]
Abstract
The COVID-19 pandemic has spread rapidly and poses an unprecedented threat to the global economy and human health. Broad-spectrum antivirals are currently being administered to treat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). China's prevention and treatment guidelines suggest the use of an anti-influenza drug, arbidol, for the clinical treatment of COVID-19. Reports indicate that arbidol could neutralize SARS-CoV-2. Monotherapy with arbidol is superior to lopinavir-ritonavir or favipiravir for treating COVID-19. In SARS-CoV-2 infection, arbidol acts by interfering with viral binding to host cells. However, the detailed mechanism by which arbidol induces the inhibition of SARS-CoV-2 is not known. Here, we present atomistic insights into the mechanism underlying membrane fusion inhibition of SARS-CoV-2 by arbidol. Molecular dynamics (MD) simulation-based analyses demonstrate that arbidol binds and stabilizes at the receptor-binding domain (RBD)/ACE2 interface with a high affinity. It forms stronger intermolecular interactions with the RBD than ACE2. Analyses of the detailed decomposition of energy components and binding affinities revealed a substantial increase in the affinity between the RBD and ACE2 in the arbidol-bound RBD/ACE2 complex, suggesting that arbidol generates favorable interactions between them. Based on our MD simulation results, we propose that the binding of arbidol induces structural rigidity in the viral glycoprotein, thus restricting the conformational rearrangements associated with membrane fusion and virus entry. Furthermore, key residues of the RBD and ACE2 that interact with arbidol were identified, opening the door for developing therapeutic strategies and higher-efficacy arbidol derivatives or lead drug candidates.
Collapse
Affiliation(s)
- Aditya K. Padhi
- Laboratory for Structural Bioinformatics, Center for Biosystems Dynamics Research, RIKEN, Yokohama, Japan
| | - Aniruddha Seal
- School of Chemical Sciences, National Institute of Science Education and Research Bhubaneswar, Khurda, Odisha, India,Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, Maharashtra, India
| | - Javed Masood Khan
- Department of Food Science and Nutrition, Faculty of Food and Agricultural Sciences, King Saud University, 2460, Riyadh, 11451, Saudi Arabia
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Timir Tripathi
- Molecular and Structural Biophysics Laboratory, Department of Biochemistry, North-Eastern Hill University, Shillong, India,Corresponding author. Department of Biochemistry North-Eastern Hill University Shillong- 793022, India
| |
Collapse
|
40
|
Cai L, Guo X, Cao Y, Ying P, Hong L, Zhang Y, Yi G, Fu M. Determining available strategies for prevention and therapy: Exploring COVID‑19 from the perspective of ACE2 (Review). Int J Mol Med 2021; 47:43. [PMID: 33576441 PMCID: PMC7891831 DOI: 10.3892/ijmm.2021.4876] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 12/07/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an acute infectious pneumonia caused by a novel type of coronavirus infection. There are currently no clinically available specific drugs for the treatment of this virus. The process of host invasion is the key to viral infection, and it is a mechanism that needs to be considered when exploring antiviral drugs. At present, studies have confirmed that angiotensin-converting enzyme II (ACE2) is the main functional receptor through which severe acute respiratory syndrome coronavirus (SARS-CoV-2) invades host cells. Therefore, a number of studies have focused on this field. However, as ACE2 may play a dual role in mediating susceptibility and immunity to SARS-CoV-2 infection, the role of ACE2 in viral infection is controversial. Beginning with the physiological function of ACE2, the present review article summarizes the influence of the ACE2 content on the susceptibility to the virus and acute lung injury. Drug mechanisms were taken as the starting point, combined with the results of clinical trials, specifically elaborating upon and analyzing the efficacy of several ACE2-centered therapeutic drugs and their potential effects. In addition, the current status of ACE2 as a targeted therapy for COVID-19 is discussed in order to provide new insight into the clinical prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Liyang Cai
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xi Guo
- Medical College of Rehabilitation, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yuchen Cao
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Peixi Ying
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Libing Hong
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Yuxi Zhang
- The Second Clinical School, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guoguo Yi
- Department of Ophthalmology, The Sixth Affiliated Hospital of Sun‑Yat‑Sen University, Guangzhou, Guangdong 510655, P.R. China
| | - Min Fu
- Department of Ophthalmology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
41
|
Españo E, Kim D, Kim J, Park SK, Kim JK. COVID-19 Antiviral and Treatment Candidates: Current Status. Immune Netw 2021; 21:e7. [PMID: 33728100 PMCID: PMC7937511 DOI: 10.4110/in.2021.21.e7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by the severe acute respiratory syndrome coronavirus 2 has severely impacted global health and economy. There is currently no effective approved treatment for COVID-19; although vaccines have been granted emergency use authorization in several countries, they are currently only administered to high-risk individuals, thereby leaving a gap in virus control measures. The scientific and clinical communities and drug manufacturers have collaborated to speed up the discovery of potential therapies for COVID-19 by taking advantage of currently approved drugs as well as investigatory agents in clinical trials. In this review, we stratified some of these candidates based on their potential targets in the progression of COVID-19 and discuss some of the results of ongoing clinical evaluations.
Collapse
Affiliation(s)
- Erica Españo
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Dajung Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Jiyeon Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Song-Kyu Park
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| | - Jeong-Ki Kim
- Department of Pharmacy, Korea University College of Pharmacy, Sejong 30019, Korea
| |
Collapse
|
42
|
Lopes-Pacheco M, Silva PL, Cruz FF, Battaglini D, Robba C, Pelosi P, Morales MM, Caruso Neves C, Rocco PRM. Pathogenesis of Multiple Organ Injury in COVID-19 and Potential Therapeutic Strategies. Front Physiol 2021; 12:593223. [PMID: 33584343 PMCID: PMC7876335 DOI: 10.3389/fphys.2021.593223] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/08/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory disease coronavirus 2 (SARS-CoV-2, formerly 2019-nCoV) is a novel coronavirus that has rapidly disseminated worldwide, causing the coronavirus disease 2019 (COVID-19) pandemic. As of January 6th, 2021, there were over 86 million global confirmed cases, and the disease has claimed over 1.87 million lives (a ∼2.2% case fatality rate). SARS-CoV-2 is able to infect human cells by binding its spike (S) protein to angiotensin-conversing enzyme 2 (ACE2), which is expressed abundantly in several cell types and tissues. ACE2 has extensive biological activities as a component of the renin-angiotensin-aldosterone system (RAAS) and plays a pivotal role as counter-regulator of angiotensin II (Ang II) activity by converting the latter to Ang (1-7). Virion binding to ACE2 for host cell entry leads to internalization of both via endocytosis, as well as activation of ADAM17/TACE, resulting in downregulation of ACE2 and loss of its protective actions in the lungs and other organs. Although COVID-19 was initially described as a purely respiratory disease, it is now known that infected individuals can rapidly progress to a multiple organ dysfunction syndrome. In fact, all human structures that express ACE2 are susceptible to SARS-CoV-2 infection and/or to the downstream effects of reduced ACE2 levels, namely systemic inflammation and injury. In this review, we aim to summarize the major features of SARS-CoV-2 biology and the current understanding of COVID-19 pathogenesis, as well as its clinical repercussions in the lung, heart, kidney, bowel, liver, and brain. We also highlight potential therapeutic targets and current global efforts to identify safe and effective therapies against this life-threatening condition.
Collapse
Affiliation(s)
- Miquéias Lopes-Pacheco
- Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisbon, Lisbon, Portugal
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| | - Denise Battaglini
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Chiara Robba
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
| | - Paolo Pelosi
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncology and Neuroscience, Genoa, Italy
- Department of Surgical Sciences and Integrated Diagnostic, University of Genoa, Genoa, Italy
| | - Marcelo Marcos Morales
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
- Laboratory of Cellular and Molecular Physiology, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Celso Caruso Neves
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
- Laboratory of Biochemistry and Cell Signaling, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Rieken Macedo Rocco
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
- COVID-19 Virus Network, Ministry of Science, Technology and Innovation, Brasília, Brazil
- COVID-19 Virus Network, Brazilian Council for Scientific and Technological Development, Brasília, Brazil
- COVID-19 Virus Network, Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro – FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Li H, Liu R, Zhang R, Zhang S, Wei Y, Zhang L, Zhou H, Yang C. Protective Effect of Arbidol Against Pulmonary Fibrosis and Sepsis in Mice. Front Pharmacol 2021; 11:607075. [PMID: 33584285 PMCID: PMC7873045 DOI: 10.3389/fphar.2020.607075] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/30/2020] [Indexed: 01/08/2023] Open
Abstract
From the perspective of epidemiology, viral immunology and current clinical research, pulmonary fibrosis may become one of the complications of patients with Coronavirus Disease 2019 (COVID-19). Cytokine storm is a major cause of new coronavirus death. The purpose of this study was to explore the effects of antiviral drug arbidol on cytokine storm and pulmonary fibrosis. Here, we use a mouse model of bleomycin-induced pulmonary fibrosis and a mouse model of fecal dilution-induced sepsis to evaluate the effects of arbidol on pulmonary fibrosis and cytokine storm. The results showed that arbidol significantly reduced the area of pulmonary fibrosis and improved lung function (reduced inspiratory resistance, lung dynamic compliance and forced vital capacity increased). Treatment with arbidol promoted reduced sepsis severity 48 h after sepsis induction, based on weight, murine sepsis score and survival rate. Arbidol observably alleviates inflammatory infiltrates and injury in the lungs and liver. Finally, we also found that arbidol reduced serum levels of pro-inflammatory factors such as TNF-α and IL-6 induced by fecal dilution. In conclusion, our results indicate that arbidol can alleviate the severity of pulmonary fibrosis and sepsis, and provide some reference for the treatment of cytokine storm and sequelae of pulmonary fibrosis in patients with COVID-19.
Collapse
Affiliation(s)
- Hailong Li
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Rui Liu
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Ruotong Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Shanshan Zhang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yiying Wei
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Liang Zhang
- Department of Thoracic Surgery, Tian Jin First Central Hospital, Tianjin, China
| | - Honggang Zhou
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| | - Cheng Yang
- The State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China.,High-Throughput Molecular Drug Screening Centre, Tianjin International Joint Academy of Biomedicine, Tianjin, China
| |
Collapse
|
44
|
Romano A, Casazza M, Gonella F. Addressing Non-linear System Dynamics of Single-Strand RNA Virus-Host Interaction. Front Microbiol 2021; 11:600254. [PMID: 33519741 PMCID: PMC7843927 DOI: 10.3389/fmicb.2020.600254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 12/09/2020] [Indexed: 12/27/2022] Open
Abstract
Positive single-strand ribonucleic acid [(+)ssRNA] viruses can cause multiple outbreaks, for which comprehensive tailored therapeutic strategies are still missing. Virus and host cell dynamics are tightly connected, generating a complex dynamics that conveys in virion assembly to ensure virus spread in the body. Starting from the knowledge of relevant processes in (+ss)RNA virus replication, transcription, translation, virions budding and shedding, and their respective energy costs, we built up a systems thinking (ST)-based diagram of the virus-host interaction, comprehensive of stocks, flows, and processes as well-described in literature. In ST approach, stocks and flows are expressed by a proxy of the energy embedded and transmitted, respectively, whereas processes are referred to the energy required for the system functioning. In this perspective, healthiness is just a particular configuration, in which stocks relevant for the system (equivalent but not limited to proteins, RNA, DNA, and all metabolites required for the survival) are constant, and the system behavior is stationary. At time of infection, the presence of additional stocks (e.g., viral protein and RNA and all metabolites required for virion assembly and spread) confers a complex network of feedbacks leading to new configurations, which can evolve to maximize the virions stock, thus changing the system structure, output, and purpose. The dynamic trajectories will evolve to achieve a new stationary status, a phenomenon described in microbiology as integration and symbiosis when the system is resilient enough to the changes, or the system may stop functioning and die. Application of external driving forces, acting on processes, can affect the dynamic trajectories adding a further degree of complexity, which can be captured by ST approach, used to address these new configurations. Investigation of system configurations in response to external driving forces acting is developed by computational analysis based on ST diagrams, with the aim at designing novel therapeutic approaches.
Collapse
Affiliation(s)
- Alessandra Romano
- Sezione di Ematologia, Dipartimento di Chirurgia Generale e Specialità Medico Chirurgiche (CHIRMED), Università degli Studi di Catania, Catania, Italy
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Marco Casazza
- Division of Hematology, U.O.C di Ematologia, Azienda Ospedaliero Universitaria Policlinico “G.Rodolico - San Marco”, Catania, Italy
| | - Francesco Gonella
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca’ Foscari Venezia, Venezia, Italy
| |
Collapse
|
45
|
Kali S, Jallet C, Azebi S, Cokelaer T, Da Fonseca JP, Wu Y, Barbier J, Cintrat JC, Gillet D, Tordo N. Broad spectrum compounds targeting early stages of rabies virus (RABV) infection. Antiviral Res 2021; 188:105016. [PMID: 33444703 DOI: 10.1016/j.antiviral.2021.105016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/31/2020] [Accepted: 01/05/2021] [Indexed: 12/16/2022]
Abstract
ABMA and its analogue DABMA are two molecules of the adamantane family known to perturbate the endosomal pathway and to inhibit cell infection of several RNA and DNA viruses. Their activity against Rabies Virus (RABV) infection has already been demonstrated in vitro. (Wu et al., 2017, 2019). Here, we describe in more details their mechanism of action by comparison to Arbidol (umifenovir) and Ribavirin, two broad spectrum antivirals against emerging viruses such as Lassa, Ebola, influenza and Hantaan viruses. ABMA and DABMA, delivered 2 h pre-infection, inhibit RABV infection in vitro with an EC50 of 7.8 μM and 14 μM, respectively. They act at post-entry, by causing RABV accumulation within the endosomal compartment and DABMA specifically diminishes the expression of the GTPase Rab7a controlling the fusion of early endosomes to late endosomes or lysosomes. This may suggest that ABMA and DABMA act at different stages of the late endosomal pathway as supported by their different profile of synergy/antagonism with the fusion inhibitor Arbidol. This difference is further confirmed by the RABV mutants induced by successive passages under increasing selective pressure showing a particular involvement of the viral G protein in the DABMA inhibition while ABMA inhibition induces less mutations dispersed in the M, G and L viral proteins. These results suggest new therapeutic perspectives against rabies.
Collapse
Affiliation(s)
- Sabrina Kali
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Institut Pasteur D'Algérie, Dely Ibrahim, Alger, Algeria
| | - Corinne Jallet
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France
| | - Saliha Azebi
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France; Unit Viral Neuroimmunology, Institut Pasteur, 75724, Paris, France
| | - Thomas Cokelaer
- Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France; Hub de Bioinformatique et Biostatistique, Institut Pasteur, USR 3756 CNRS, 75724, Paris, France
| | - Juliana Pipoli Da Fonseca
- Plate-forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Institut Pasteur, 75724, Paris, France
| | - Yu Wu
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Julien Barbier
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Jean-Christophe Cintrat
- Service de Chimie Bioorganique et Marquage (SCBM), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Daniel Gillet
- Service D'Ingénierie Moléculaire des Protéines (SIMOPRO), CEA, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Noël Tordo
- Unit Antiviral Strategies, Institut Pasteur, 75724, Paris, France; Institut Pasteur de Guinée, BP 4416, Conakry, Guinea.
| |
Collapse
|
46
|
Pagliano P, Scarpati G, Sellitto C, Conti V, Spera AM, Ascione T, Piazza O, Filippelli A. Experimental Pharmacotherapy for COVID-19: The Latest Advances. J Exp Pharmacol 2021; 13:1-13. [PMID: 33442304 PMCID: PMC7800714 DOI: 10.2147/jep.s255209] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 12/12/2020] [Indexed: 12/17/2022] Open
Abstract
The coronavirus infectious disease-2019 (COVID-19) has overwhelmed like a shock wave in a completely unprepared world. Despite coronavirus infections were involved in previous epidemic outbreaks, no antiviral agent was developed for specific treatment. As a consequence, since the beginning of this pandemic, both repositioned and experimental drugs were used to treat the infected patients without evidence of clinical efficacy. Just based on experience coming from the use of antiviral agents to treat other viruses (eg, lopinavir/ritonavir, remdesivir) and supposed antiviral or immunomodulatory activities of drugs with no approved antiviral indications (eg hydroxychloroquine, tocilizumab), clinicians have faced the ongoing pandemic. Currently, after about 9 months from the COVID-19 spread, there is still no antiviral agent capable of ensuring the cure of this syndrome. Clinical trials are beginning to confirm the benefits of some drugs, while for other compounds, efficacy and safety have not yet been confirmed. Randomized clinical trials (RCT) have denied or downsized the beneficial effects attributed to certain molecules, such as aminoquinolines, largely used in clinical practice at the beginning of COVID-19 spread. Conversely, at the same time, they have provided evidence for unexpected effectiveness of other agents that have been underutilized, such as steroids, which were not used in SARS treatment because of the threatened effect on viral replication. Evidence deriving from pathologic studies have demonstrated that the prothrombotic effects of SARS-CoV-2 can be prevented by heparin prophylaxis, underlining the need for personalized treatment for patients with severe disease. The main aim of this review is to synthesize the available information and evidence on both repositioned and experimental drugs for the treatment of COVID-19, focusing on the need to exercise caution on the use of unproven medical therapies.
Collapse
Affiliation(s)
- Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Infectious Diseases, University of Salerno, Baronissi, Italy
| | - Giuliana Scarpati
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Anesthesiology, University of Salerno, Baronissi, Italy
| | - Carmine Sellitto
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Pharmacology, University of Salerno, Baronissi, Italy
| | - Valeria Conti
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Pharmacology, University of Salerno, Baronissi, Italy
| | - Anna Maria Spera
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Infectious Diseases, University of Salerno, Baronissi, Italy
| | - Tiziana Ascione
- Department of Medicine, Service of Infectious Diseases, Cardarelli Hospital, Naples, Italy
| | - Ornella Piazza
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Anesthesiology, University of Salerno, Baronissi, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery and Dentistry, “Scuola Medica Salernitana”, Unit of Pharmacology, University of Salerno, Baronissi, Italy
| |
Collapse
|
47
|
Hossain MJ, Rahman SMA. Repurposing therapeutic agents against SARS-CoV-2 infection: most promising and neoteric progress. Expert Rev Anti Infect Ther 2020; 19:1009-1027. [PMID: 33355520 DOI: 10.1080/14787210.2021.1864327] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION The pathogenic and highly transmissible etiological agent, SARS-CoV-2, has caused a serious threat COVID-19 pandemic. WHO has declared the epidemic a public health emergency of international concern owing to its high contagiosity, mortality rate, and morbidity. Till now, there is no approved vaccine or drug to combat the COVID-19 and avert this global crisis. AREAS COVERED In this narrative review, we summarized the updated results (January to August 2020) of the most promising repurposing therapeutic candidates to treat the SARS-CoV-2 viral infection. The repurposed drugs classified under four headlines like antivirals, anti-parasitic, immune-modulating, and miscellaneous drugs were discussed with their in vitro efficacy to recent clinical advancements against COVID-19. EXPERT OPINION Currently, palliative care, ranging from outpatient management to intensive care, including oxygen administration, ventilator support, intravenous fluids therapy, with some repurposed drugs, are the primary weapons to fight against COVID-19. Until a safe and effective vaccine is developed, an evidence-based drug repurposing strategy might be the wisest option to save people from this catastrophe. Several existing drugs are now under clinical trials, and some of them are approved in different places of the world for emergency use or as adjuvant therapy in COVID-19 with standard of care.
Collapse
Affiliation(s)
- Md Jamal Hossain
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Dhaka, Dhaka-1000, Bangladesh
| | - S M Abdur Rahman
- Department of Clinical Pharmacy and Pharmacology, Faculty of Pharmacy, University of Dhaka, Dhaka-1000, Bangladesh
| |
Collapse
|
48
|
Tan J, Yuan Y, Xu C, Song C, Liu D, Ma D, Gao Q. A retrospective comparison of drugs against COVID-19. Virus Res 2020; 294:198262. [PMID: 33333102 PMCID: PMC7833729 DOI: 10.1016/j.virusres.2020.198262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/14/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022]
Abstract
Hydroxychloroquine is an efficient candidate drug against COVID-19. Oseltamivir can be prudently considered in combination therapy. Drug repurposing is a promising way to combat SARS-CoV-2 infection. Comparison of drug effects against COVID-19 is instructive in the pandemic.
Coronavirus disease 19 (COVID-19) has posed serious threats to the general population. To relieve the crisis, a comparison of drug effects against COVID-19 is instructive. Between January 27, 2020 and March 21, 2020, a total of 333 patients treated with arbidol, corticosteroids, hydroxychloroquine, lopinavir/ritonavir, or oseltamivir monotherapy, having definite outcomes and serological antibody detection results, were retrospectively analyzed. The hydroxychloroquine group had a significantly reduced duration of hospital stay than the arbidol and corticosteroids groups. The oseltamivir group had a significantly shorter length of hospital stay than the arbidol, corticosteroids, and lopinavir/ritonavir groups. The hydroxychloroquine group had a significantly higher IgM titer than the other four groups and exhibited significantly higher IgG levels than the arbidol, lopinavir/ritonavir, and oseltamivir groups. Our findings indicated that hydroxychloroquine might have the potential for efficient COVID-19 management, while oseltamivir should be prudently considered in combination therapy.
Collapse
Affiliation(s)
- Jiahong Tan
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yuan Yuan
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Cheng Xu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Chunyan Song
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Dan Liu
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ding Ma
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Qinglei Gao
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
49
|
Nojomi M, Yassin Z, Keyvani H, Makiani MJ, Roham M, Laali A, Dehghan N, Navaei M, Ranjbar M. Effect of Arbidol (Umifenovir) on COVID-19: a randomized controlled trial. BMC Infect Dis 2020; 20:954. [PMID: 33317461 PMCID: PMC7734453 DOI: 10.1186/s12879-020-05698-w] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Treatment of patients with COVID-19 has included supportive care to mainly relief symptoms of the disease. Although World Health Organization (WHO) has not recommended any effective treatments for COVID-19, there are some reports about use of antiviral drugs. The aim of this study is to determine the effect of Arbidol (ARB) on COVID-19 disease. METHODS Using an open-label randomized controlled trial, we examined the efficacy of ARB in patients with COVID-19 in a teaching hospital. One hundred eligible patients with diagnosis of COVID-19 were recruited in the study and assigned randomly to two groups of either hydroxychloroquine followed by KALETRA (Lopinavir/ritonavir) or hydroxychloroquine followed by ARB. The primary outcome was hospitalization duration and clinical improvement 7 days after admission. The criteria of improvement were relief of cough, dyspnea, and fever. Time to relief from fever was also assessed across the two groups. Without any dropouts, 100 patients were entered into the study for the final analysis at significance level of 0.05. RESULTS The mean age of patients was 56.6 (17.8) years and 56.2 (14.8) years in ARB and KALETRA groups, respectively. Majority of patients were male across two groups (66 and 54%). The duration of hospitalization in ARB group was significantly less than KALETRA arm (7.2 versus 9.6 days; P = 0.02). Time to relief fever was almost similar across two groups (2.7 versus 3.1 days in ARB and KALETRA arms, respectively). Peripheral oxygen saturation rate was significantly different after 7 days of admission across two groups (94% versus 92% in ARB and KALETRA groups respectively) (P = 0.02). Based on multiple linear regression analysis, IHD, Na level, and oxygen saturation at the time of admission and type of therapy were the independent adjusted variables that determined the duration of hospitalization in patients with COVID-19. CONCLUSION Our findings showed that Arbidol, compared to KALETRA, significantly contributes to clinical and laboratory improvements, including peripheral oxygen saturation, requiring ICU admissions, duration of hospitalization, chest CT involvements, WBC, and ESR. We suggest further studies on ARB against COVID-19 using larger sample size and multicenter design. TRIAL REGISTRATION IRCT20180725040596N2 on 18 April 2020.
Collapse
Affiliation(s)
- Marzieh Nojomi
- Preventive Medicine & Public Health Research Center, Psychosocial Health Research Institute, Department of Community and Family Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zeynab Yassin
- Antimicrobial Resistance Research Center, Department of Infectious Disease, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Keyvani
- Department of Virology, School of Medicine, Gastrointestinal and Liver Disease Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mahin Jamshidi Makiani
- Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Roham
- Antimicrobial Resistance Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Azadeh Laali
- Department of Infectious Disease, School of Medicine, Firoozgar General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Nasir Dehghan
- Department of Community and Family Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrnaz Navaei
- Department of Infectious Disease, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Ranjbar
- Department of Infectious Disease, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
50
|
Yadav M, Dhagat S, Eswari JS. Emerging strategies on in silico drug development against COVID-19: challenges and opportunities. Eur J Pharm Sci 2020; 155:105522. [PMID: 32827661 PMCID: PMC7438372 DOI: 10.1016/j.ejps.2020.105522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022]
Abstract
The importance of coronaviruses as human pathogen has been highlighted by the recent outbreak of SARS-CoV-2 leading to the search of suitable drugs to overcome respiratory infections caused by the virus. Due to the lack of specific drugs against coronavirus, the existing antiviral and antimalarial drugs are currently being administered to the patients infected with SARS-CoV-2. The scientists are also considering repurposing of some of the existing drugs as a suitable option in search of effective drugs against coronavirus till the establishment of a potent drug and/or vaccine. Computer-aided drug discovery provides a promising attempt to enable scientists to develop new and target specific drugs to combat any disease. The discovery of novel targets for COVID-19 using computer-aided drug discovery tools requires knowledge of the structure of coronavirus and various target proteins present in the virus. Targeting viral proteins will make the drug specific against the virus, thereby, increasing the chances of viral mortality. Hence, this review provides the structure of SARS-CoV-2 virus along with the important viral components involved in causing infection. It also focuses on the role of various target proteins in disease, the mechanism by which currently administered drugs act against the virus and the repurposing of few drugs. The gap arising from the absence of specific drugs is addressed by proposing potential antiviral drug targets which might provide insights into structure-based drug development against SARS-CoV-2.
Collapse
Affiliation(s)
- Manisha Yadav
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India
| | - Swasti Dhagat
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India
| | - J Satya Eswari
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India.
| |
Collapse
|