1
|
Tamilarasi W, Balamurugan BJ. New reverse sum Revan indices for physicochemical and pharmacokinetic properties of anti-filovirus drugs. Front Chem 2024; 12:1486933. [PMID: 39749221 PMCID: PMC11693449 DOI: 10.3389/fchem.2024.1486933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/27/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Ebola and Marburg viruses, biosafety level 4 pathogens, cause severe hemorrhaging and organ failure with high mortality. Although some FDA-approved vaccines or therapeutics like Ervebo for Zaire Ebola virus exist, still there is a lack of effective therapeutics that cover all filoviruses, including both Ebola and Marburg viruses. Therefore, some anti-filovirus drugs such as Pinocembrin, Favipiravir, Remdesivir and others are used to manage infections. In theoretical chemistry, a chemical molecule is converted into an isomorphic molecular graph, G ( V , E ) by considering atom set V as vertices and bond set E as edges. A topological index is a molecular descriptor derived from the molecular graph of a chemical compound that characterizes its topology. The relationship between a compound's chemical structure and its properties is investigated through the quantitative structure-property relationship (QSPR). This article introduces new reverse sum Revan degree based indices to explore the physicochemical and pharmacokinetic properties of anti-filovirus drugs via multilinear regression. The findings reveal a strong correlation between these proposed indices and the properties of anti-filovirus drugs when compared to reverse and Revan degree-based indices. Thus, reverse sum Revan indices offer valuable insights for analyzing the drugs properties used to treat Ebola and Marburg virus infections. Moreover, the multilinear regression (MLR) results through reverse sum Revan indices are compared with Artificial Neural Network (ANN) modelling technique and it provides the better prediction of the physicochemical and pharmacokinetic properties of anti-filovirus drugs.
Collapse
Affiliation(s)
| | - B. J. Balamurugan
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
2
|
Styles CT, Zhou J, Flight KE, Brown JC, Lewis C, Wang X, Vanden Oever M, Peacock TP, Wang Z, Millns R, O'Neill JS, Borodavka A, Grove J, Barclay WS, Tregoning JS, Edgar RS. Propylene glycol inactivates respiratory viruses and prevents airborne transmission. EMBO Mol Med 2023; 15:e17932. [PMID: 37970627 PMCID: PMC10701621 DOI: 10.15252/emmm.202317932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/28/2023] [Revised: 10/16/2023] [Accepted: 10/17/2023] [Indexed: 11/17/2023] Open
Abstract
Viruses are vulnerable as they transmit between hosts, and we aimed to exploit this critical window. We found that the ubiquitous, safe, inexpensive and biodegradable small molecule propylene glycol (PG) has robust virucidal activity. Propylene glycol rapidly inactivates a broad range of viruses including influenza A, SARS-CoV-2 and rotavirus and reduces disease burden in mice when administered intranasally at concentrations commonly found in nasal sprays. Most critically, vaporised PG efficiently abolishes influenza A virus and SARS-CoV-2 infectivity within airborne droplets, potently preventing infection at levels well below those tolerated by mammals. We present PG vapour as a first-in-class non-toxic airborne virucide that can prevent transmission of existing and emergent viral pathogens, with clear and immediate implications for public health.
Collapse
Affiliation(s)
| | - Jie Zhou
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - Katie E Flight
- Department of Infectious DiseaseImperial College LondonLondonUK
- Present address:
University College LondonLondonUK
| | | | - Charlotte Lewis
- MRC‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Xinyu Wang
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| | - Michael Vanden Oever
- Department of Infectious DiseaseImperial College LondonLondonUK
- Present address:
Life Edit TherapeuticsMorrisvilleNCUSA
| | | | - Ziyin Wang
- Department of Infectious DiseaseImperial College LondonLondonUK
| | - Rosie Millns
- Department of Infectious DiseaseImperial College LondonLondonUK
| | | | | | - Joe Grove
- MRC‐University of Glasgow Centre for Virus ResearchGlasgowUK
| | - Wendy S Barclay
- Department of Infectious DiseaseImperial College LondonLondonUK
| | | | - Rachel S Edgar
- Department of Infectious DiseaseImperial College LondonLondonUK
| |
Collapse
|
3
|
Proposal to Consider Chemical/Physical Microenvironment as a New Therapeutic Off-Target Approach. Pharmaceutics 2022; 14:pharmaceutics14102084. [PMID: 36297518 PMCID: PMC9611316 DOI: 10.3390/pharmaceutics14102084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/01/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Abstract
The molecular revolution could lead drug discovery from chance observation to the rational design of new classes of drugs that could simultaneously be more effective and less toxic. Unfortunately, we are witnessing some failure in this sense, and the causes of the crisis involve a wide range of epistemological and scientific aspects. In pharmacology, one key point is the crisis of the paradigm the “magic bullet”, which is to design therapies based on specific molecular targets. Drug repurposing is one of the proposed ways out of the crisis and is based on the off-target effects of known drugs. Here, we propose the microenvironment as the ideal place to direct the off-targeting of known drugs. While it has been extensively investigated in tumors, the generation of a harsh microenvironment is also a phenotype of the vast majority of chronic diseases. The hostile microenvironment, on the one hand, reduces the efficacy of both chemical and biological drugs; on the other hand, it dictates a sort of “Darwinian” selection of those cells armed to survive in such hostile conditions. This opens the way to the consideration of the microenvironment as a convenient target for pharmacological action, with a clear example in proton pump inhibitors.
Collapse
|
4
|
Eskandarzade N, Ghorbani A, Samarfard S, Diaz J, Guzzi PH, Fariborzi N, Tahmasebi A, Izadpanah K. Network for network concept offers new insights into host- SARS-CoV-2 protein interactions and potential novel targets for developing antiviral drugs. Comput Biol Med 2022; 146:105575. [PMID: 35533462 PMCID: PMC9055686 DOI: 10.1016/j.compbiomed.2022.105575] [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] [Academic Contribution Register] [Received: 11/13/2021] [Revised: 04/16/2022] [Accepted: 04/27/2022] [Indexed: 01/08/2023]
Abstract
SARS-CoV-2, the causal agent of COVID-19, is primarily a pulmonary virus that can directly or indirectly infect several organs. Despite many studies carried out during the current COVID-19 pandemic, some pathological features of SARS-CoV-2 have remained unclear. It has been recently attempted to address the current knowledge gaps on the viral pathogenicity and pathological mechanisms via cellular-level tropism of SARS-CoV-2 using human proteomics, visualization of virus-host protein-protein interactions (PPIs), and enrichment analysis of experimental results. The synergistic use of models and methods that rely on graph theory has enabled the visualization and analysis of the molecular context of virus/host PPIs. We review current knowledge on the SARS-COV-2/host interactome cascade involved in the viral pathogenicity through the graph theory concept and highlight the hub proteins in the intra-viral network that create a subnet with a small number of host central proteins, leading to cell disintegration and infectivity. Then we discuss the putative principle of the "gene-for-gene and "network for network" concepts as platforms for future directions toward designing efficient anti-viral therapies.
Collapse
Affiliation(s)
- Neda Eskandarzade
- Department of Basic Sciences, School of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Abozar Ghorbani
- Nuclear Agriculture Research School, Nuclear Science and Technology Research Institute (NSTRI), Karaj, Iran,Corresponding author
| | - Samira Samarfard
- Berrimah Veterinary Laboratory, Department of Primary Industry and Resources, Berrimah, NT, 0828, Australia
| | - Jose Diaz
- Laboratorio de Dinámica de Redes Genéticas, Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | - Pietro H. Guzzi
- Department of Medical and Surgical Sciences, Laboratory of Bioinformatics Unit, Italy
| | - Niloofar Fariborzi
- Department of Medical Entomology and Vector Control, School of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Tahmasebi
- Institute of Biotechnology, College of Agriculture, Shiraz University, Shiraz, Iran
| | | |
Collapse
|
5
|
Calvo-Alvarez E, Dolci M, Perego F, Signorini L, Parapini S, D’Alessandro S, Denti L, Basilico N, Taramelli D, Ferrante P, Delbue S. Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey. Microorganisms 2022; 10:1284. [PMID: 35889004 PMCID: PMC9320270 DOI: 10.3390/microorganisms10071284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/05/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 01/09/2023] Open
Abstract
More than two years have passed since the viral outbreak that led to the novel infectious respiratory disease COVID-19, caused by the SARS-CoV-2 coronavirus. Since then, the urgency for effective treatments resulted in unprecedented efforts to develop new vaccines and to accelerate the drug discovery pipeline, mainly through the repurposing of well-known compounds with broad antiviral effects. In particular, antiparasitic drugs historically used against human infections due to protozoa or helminth parasites have entered the main stage as a miracle cure in the fight against SARS-CoV-2. Despite having demonstrated promising anti-SARS-CoV-2 activities in vitro, conflicting results have made their translation into clinical practice more difficult than expected. Since many studies involving antiparasitic drugs are currently under investigation, the window of opportunity might be not closed yet. Here, we will review the (controversial) journey of these old antiparasitic drugs to combat the human infection caused by the novel coronavirus SARS-CoV-2.
Collapse
Affiliation(s)
- Estefanía Calvo-Alvarez
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Maria Dolci
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Federica Perego
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Lucia Signorini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Silvia Parapini
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| | - Sarah D’Alessandro
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (S.D.); (D.T.)
| | - Luca Denti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Nicoletta Basilico
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Donatella Taramelli
- Department of Pharmacological and Biomolecular Sciences, University of Milan, 20133 Milan, Italy; (S.D.); (D.T.)
| | - Pasquale Ferrante
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| | - Serena Delbue
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (M.D.); (F.P.); (L.S.); (L.D.); (N.B.); (P.F.); (S.D.)
| |
Collapse
|
6
|
Galão RP, Wilson H, Schierhorn KL, Debeljak F, Bodmer BS, Goldhill D, Hoenen T, Wilson SJ, Swanson CM, Neil SJD. TRIM25 and ZAP target the Ebola virus ribonucleoprotein complex to mediate interferon-induced restriction. PLoS Pathog 2022; 18:e1010530. [PMID: 35533151 PMCID: PMC9119685 DOI: 10.1371/journal.ppat.1010530] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2021] [Revised: 05/19/2022] [Accepted: 04/18/2022] [Indexed: 12/14/2022] Open
Abstract
Ebola virus (EBOV) causes highly pathogenic disease in primates. Through screening a library of human interferon-stimulated genes (ISGs), we identified TRIM25 as a potent inhibitor of EBOV transcription-and-replication-competent virus-like particle (trVLP) propagation. TRIM25 overexpression inhibited the accumulation of viral genomic and messenger RNAs independently of the RNA sensor RIG-I or secondary proinflammatory gene expression. Deletion of TRIM25 strongly attenuated the sensitivity of trVLPs to inhibition by type-I interferon. The antiviral activity of TRIM25 required ZAP and the effect of type-I interferon was modulated by the CpG dinucleotide content of the viral genome. We find that TRIM25 interacts with the EBOV vRNP, resulting in its autoubiquitination and ubiquitination of the viral nucleoprotein (NP). TRIM25 is recruited to incoming vRNPs shortly after cell entry and leads to dissociation of NP from the vRNA. We propose that TRIM25 targets the EBOV vRNP, exposing CpG-rich viral RNA species to restriction by ZAP.
Collapse
Affiliation(s)
- Rui Pedro Galão
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| | - Harry Wilson
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| | - Kristina L. Schierhorn
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| | - Franka Debeljak
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| | - Bianca S. Bodmer
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Daniel Goldhill
- Section of Virology, Department of Medicine, Imperial College London, London, United Kingdom
| | - Thomas Hoenen
- Institute for Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut, Greifswald, Germany
| | - Sam J. Wilson
- MRC Centre for Virus Research, University of Glasgow, United Kingdom
| | - Chad M. Swanson
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| | - Stuart J. D. Neil
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King’s College London, United Kingdom
| |
Collapse
|
7
|
Liu CH, Hu YT, Wong SH, Lin LT. Therapeutic Strategies against Ebola Virus Infection. Viruses 2022; 14:v14030579. [PMID: 35336986 PMCID: PMC8954160 DOI: 10.3390/v14030579] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 12/10/2022] Open
Abstract
Since the 2014–2016 epidemic, Ebola virus (EBOV) has spread to several countries and has become a major threat to global health. EBOV is a risk group 4 pathogen, which imposes significant obstacles for the development of countermeasures against the virus. Efforts have been made to develop anti-EBOV immunization and therapeutics, with three vaccines and two antibody-based therapeutics approved in recent years. Nonetheless, the high fatality of Ebola virus disease highlights the need to continuously develop antiviral strategies for the future management of EBOV outbreaks in conjunction with vaccination programs. This review aims to highlight potential EBOV therapeutics and their target(s) of inhibition, serving as a summary of the literature to inform readers of the novel candidates available in the continued search for EBOV antivirals.
Collapse
Affiliation(s)
- Ching-Hsuan Liu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Yee-Tung Hu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Shu Hui Wong
- International Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
| | - Liang-Tzung Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan;
- Correspondence:
| |
Collapse
|
8
|
Xin Y, Chen S, Tang K, Wu Y, Guo Y. Identification of Nifurtimox and Chrysin as Anti-Influenza Virus Agents by Clinical Transcriptome Signature Reversion. Int J Mol Sci 2022; 23:ijms23042372. [PMID: 35216485 PMCID: PMC8876279 DOI: 10.3390/ijms23042372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2022] [Revised: 02/12/2022] [Accepted: 02/18/2022] [Indexed: 12/28/2022] Open
Abstract
The rapid development in the field of transcriptomics provides remarkable biomedical insights for drug discovery. In this study, a transcriptome signature reversal approach was conducted to identify the agents against influenza A virus (IAV) infection through dissecting gene expression changes in response to disease or compounds’ perturbations. Two compounds, nifurtimox and chrysin, were identified by a modified Kolmogorov–Smirnov test statistic based on the transcriptional signatures from 81 IAV-infected patients and the gene expression profiles of 1309 compounds. Their activities were verified in vitro with half maximal effective concentrations (EC50s) from 9.1 to 19.1 μM against H1N1 or H3N2. It also suggested that the two compounds interfered with multiple sessions in IAV infection by reversing the expression of 28 IAV informative genes. Through network-based analysis of the 28 reversed IAV informative genes, a strong synergistic effect of the two compounds was revealed, which was confirmed in vitro. By using the transcriptome signature reversion (TSR) on clinical datasets, this study provides an efficient scheme for the discovery of drugs targeting multiple host factors regarding clinical signs and symptoms, which may also confer an opportunity for decelerating drug-resistant variant emergence.
Collapse
Affiliation(s)
- Yijing Xin
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Shubing Chen
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ke Tang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - You Wu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Guo
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (Y.X.); (S.C.); (K.T.); (Y.W.)
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Correspondence: ; Tel.: +86-010-63161716
| |
Collapse
|
9
|
Abel L, Perera SM, Yam D, Garbern S, Kennedy SB, Massaquoi M, Sahr F, Woldemichael D, Liu T, Levine AC, Aluisio AR. Association between oral antimalarial medication administration and mortality among patients with Ebola virus disease: a multisite cohort study. BMC Infect Dis 2022; 22:71. [PMID: 35057753 PMCID: PMC8772178 DOI: 10.1186/s12879-021-06811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/18/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Empiric antimalarial treatment is a component of protocol-based management of Ebola virus disease (EVD), yet this approach has limited clinical evidence for patient-centered benefits. METHODS This retrospective cohort study evaluated the association between antimalarial treatment and mortality among patients with confirmed EVD. The data was collected from five International Medical Corps operated Ebola Treatment Units (ETUs) in Sierra Leone and Liberia from 2014 through 2015. The standardized protocol used for patient care included empiric oral treatment with combination artemether and lumefantrine, twice daily for three days; however, only a subset of patients received treatment due to resource variability. The outcome of interest was mortality, comparing patients treated with oral antimalarials within 48-h of admission to those not treated. Analysis was conducted with logistic regression to generate adjusted odds ratios (aORs). Multivariable analyses controlled for ETU country, malaria rapid diagnostic test result, age, EVD cycle threshold value, symptoms of bleeding, diarrhea, dysphagia and dyspnea, and additional standard clinical treatments. RESULTS Among the 424 cases analyzed, 376 (88.7%) received early oral antimalarials. Across all cases, mortality occurred in 57.5% (244). In comparing unadjusted mortality prevalence, early antimalarial treated cases yielded 55.1% mortality versus 77.1% mortality for those untreated (p = 0.005). Multivariable analysis demonstrated evidence of reduced aOR for mortality with early oral antimalarial treatment versus non-treatment (aOR = 0.34, 95% Confidence Interval: 0.12, 0.92, p = 0.039). CONCLUSION Early oral antimalarial treatment in an EVD outbreak was associated with reduced mortality. Further study is warranted to investigate this association between early oral antimalarial treatment and mortality in EVD patients.
Collapse
Affiliation(s)
- Logan Abel
- Warren Alpert Medical School of Brown University, Providence, RI, USA
| | | | - Derrick Yam
- Center for Statistical Sciences, Department of Biostatistics, Brown University School of Public Health, Providence, RI, USA
| | - Stephanie Garbern
- Department of Emergency Medicine, Warren Alpert Medical School of Brown University, 55 Claverick Street, Room 274, Providence, RI, 02903, USA
| | | | | | - Foday Sahr
- College of Medicine and Allied Health Sciences, University of Sierra Leone, Freetown, Sierra Leone
| | | | - Tao Liu
- Center for Statistical Sciences, Department of Biostatistics, Brown University School of Public Health, Providence, RI, USA
| | - Adam C Levine
- Department of Emergency Medicine, Warren Alpert Medical School of Brown University, 55 Claverick Street, Room 274, Providence, RI, 02903, USA
| | - Adam R Aluisio
- Department of Emergency Medicine, Warren Alpert Medical School of Brown University, 55 Claverick Street, Room 274, Providence, RI, 02903, USA.
| |
Collapse
|
10
|
Jaber S, Nemska V, Iliev I, Ivanova E, Foteva T, Georgieva N, Givechev I, Naydenova E, Karadjova V, Danalev D. Synthesis and Biological Studies on (KLAKLAK) 2-NH 2 Analog Containing Unnatural Amino Acid β-Ala and Conjugates with Second Pharmacophore. Molecules 2021; 26:7321. [PMID: 34885902 PMCID: PMC8658989 DOI: 10.3390/molecules26237321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 11/25/2022] Open
Abstract
(1) Background: Peptides are good candidates for anticancer drugs due to their natural existence in the body and lack of secondary effects. (KLAKLAK)2 is an antimicrobial peptide that also shows good anticancer properties. (2) Methods: The Solid Phase Peptide Synthesis (Fmoc-strategy) was used for the synthesis of target molecules, analogs of (KLAKLAK)2-NH2. The purity of all compounds was monitored by HPLC, and their structures were proven using mass spectrometry. Cytotoxicity and antiproliferative effects were studied using 3T3 NRU and MTT tests, respectively. For determination of antimicrobial activity, the disc-diffusion method was used. Hydrolytic stability at three pH values, which mimic the physiological pH in the body, was investigated by means of the HPLC technique. (3) Results: A good selective index against MCF-7 tumor cell lines, combined with good cytotoxicity and antiproliferative properties, was revealed for conjugates NphtG-(KLAKLAK)2-NH2 and Caf-(KLAKLAK)2-NH2. The same compounds showed very good antifungal properties and complete hydrolytic stability for 72 h. The compound Caf-(KLβ-AKLβ-AK)2-NH2 containing β-Ala in its structures exhibited good antimicrobial activity against Escherichia coli K12 407 and Bacillus subtilis 3562, in combination with very good antiproliferative and cytotoxic properties, as well as hydrolytic stability. (4) Conclusions: The obtained results reveal that all synthesized conjugates could be useful for medical practice as anticancer or antimicrobial agents.
Collapse
Affiliation(s)
- Sirine Jaber
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Veronica Nemska
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Ivan Iliev
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 25 A, 1113 Sofia, Bulgaria; (I.I.); (E.I.)
| | - Elena Ivanova
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 25 A, 1113 Sofia, Bulgaria; (I.I.); (E.I.)
| | - Tsvetelina Foteva
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Nelly Georgieva
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Ivan Givechev
- Testing Center Global Test Ltd., 31 Krushovski vrah Street, 1618 Sofia, Bulgaria;
| | - Emilia Naydenova
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Veronika Karadjova
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| | - Dancho Danalev
- University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd, 1756 Sofia, Bulgaria; (S.J.); (V.N.); (T.F.); (N.G.); (E.N.); (V.K.)
| |
Collapse
|
11
|
Prabhakara C, Godbole R, Sil P, Jahnavi S, Gulzar SEJ, van Zanten TS, Sheth D, Subhash N, Chandra A, Shivaraj A, Panikulam P, U I, Nuthakki VK, Puthiyapurayil TP, Ahmed R, Najar AH, Lingamallu SM, Das S, Mahajan B, Vemula P, Bharate SB, Singh PP, Vishwakarma R, Guha A, Sundaramurthy V, Mayor S. Strategies to target SARS-CoV-2 entry and infection using dual mechanisms of inhibition by acidification inhibitors. PLoS Pathog 2021; 17:e1009706. [PMID: 34252168 PMCID: PMC8297935 DOI: 10.1371/journal.ppat.1009706] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2021] [Revised: 07/22/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022] Open
Abstract
Many viruses utilize the host endo-lysosomal network for infection. Tracing the endocytic itinerary of SARS-CoV-2 can provide insights into viral trafficking and aid in designing new therapeutic strategies. Here, we demonstrate that the receptor binding domain (RBD) of SARS-CoV-2 spike protein is internalized via the pH-dependent CLIC/GEEC (CG) endocytic pathway in human gastric-adenocarcinoma (AGS) cells expressing undetectable levels of ACE2. Ectopic expression of ACE2 (AGS-ACE2) results in RBD traffic via both CG and clathrin-mediated endocytosis. Endosomal acidification inhibitors like BafilomycinA1 and NH4Cl, which inhibit the CG pathway, reduce the uptake of RBD and impede Spike-pseudoviral infection in both AGS and AGS-ACE2 cells. The inhibition by BafilomycinA1 was found to be distinct from Chloroquine which neither affects RBD uptake nor alters endosomal pH, yet attenuates Spike-pseudovirus entry. By screening a subset of FDA-approved inhibitors for functionality similar to BafilomycinA1, we identified Niclosamide as a SARS-CoV-2 entry inhibitor. Further validation using a clinical isolate of SARS-CoV-2 in AGS-ACE2 and Vero cells confirmed its antiviral effect. We propose that Niclosamide, and other drugs which neutralize endosomal pH as well as inhibit the endocytic uptake, could provide broader applicability in subverting infection of viruses entering host cells via a pH-dependent endocytic pathway.
Collapse
Affiliation(s)
| | - Rashmi Godbole
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- University of Trans-Disciplinary Health Sciences and Technology (TDU), Bengaluru, India
| | - Parijat Sil
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Sowmya Jahnavi
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Shah-e-Jahan Gulzar
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- SASTRA University, Thanjavur, India
| | | | - Dhruv Sheth
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | - Neeraja Subhash
- National Centre for Biological Sciences (TIFR), Bengaluru, India
- SASTRA University, Thanjavur, India
| | - Anchal Chandra
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | | | - Ibrahim U
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | | | - Riyaz Ahmed
- CSIR—Indian Institute of Integrative Medicine, Jammu, India
| | | | - Sai Manoz Lingamallu
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
- Manipal Academy of Higher Education (MAHE), Madhav Nagar, Manipal, Karnataka, India
| | - Snigdhadev Das
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| | | | - Praveen Vemula
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
| | | | | | | | - Arjun Guha
- Institute for Stem Cell Science and Regenerative Medicine (inSTEM), Bengaluru, India
| | | | - Satyajit Mayor
- National Centre for Biological Sciences (TIFR), Bengaluru, India
| |
Collapse
|
12
|
The antiandrogen enzalutamide downregulates TMPRSS2 and reduces cellular entry of SARS-CoV-2 in human lung cells. Nat Commun 2021; 12:4068. [PMID: 34210968 PMCID: PMC8249423 DOI: 10.1038/s41467-021-24342-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/09/2020] [Accepted: 06/09/2021] [Indexed: 12/18/2022] Open
Abstract
SARS-CoV-2 attacks various organs, most destructively the lung, and cellular entry requires two host cell surface proteins: ACE2 and TMPRSS2. Downregulation of one or both of these is thus a potential therapeutic approach for COVID-19. TMPRSS2 is a known target of the androgen receptor, a ligand-activated transcription factor; androgen receptor activation increases TMPRSS2 levels in various tissues, most notably prostate. We show here that treatment with the antiandrogen enzalutamide—a well-tolerated drug widely used in advanced prostate cancer—reduces TMPRSS2 levels in human lung cells and in mouse lung. Importantly, antiandrogens significantly reduced SARS-CoV-2 entry and infection in lung cells. In support of this experimental data, analysis of existing datasets shows striking co-expression of AR and TMPRSS2, including in specific lung cell types targeted by SARS-CoV-2. Together, the data presented provides strong evidence to support clinical trials to assess the efficacy of antiandrogens as a treatment option for COVID-19. TMPRSS2 is regulated by androgen receptor signalling in the prostate, however it is unclear if blocking this signalling is beneficial in the context of SARS-CoV-2 lung infection. Here the authors show that antiandrogen treatment downregulates TMPRSS2 in the lung and reduces viral entry and infection.
Collapse
|
13
|
The furin cleavage site in the SARS-CoV-2 spike protein is required for transmission in ferrets. Nat Microbiol 2021; 6:899-909. [PMID: 33907312 DOI: 10.1038/s41564-021-00908-w] [Citation(s) in RCA: 484] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2020] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 entry requires sequential cleavage of the spike glycoprotein at the S1/S2 and the S2' cleavage sites to mediate membrane fusion. SARS-CoV-2 has a polybasic insertion (PRRAR) at the S1/S2 cleavage site that can be cleaved by furin. Using lentiviral pseudotypes and a cell-culture-adapted SARS-CoV-2 virus with an S1/S2 deletion, we show that the polybasic insertion endows SARS-CoV-2 with a selective advantage in lung cells and primary human airway epithelial cells, but impairs replication in Vero E6, a cell line used for passaging SARS-CoV-2. Using engineered spike variants and live virus competition assays and by measuring growth kinetics, we find that the selective advantage in lung and primary human airway epithelial cells depends on the expression of the cell surface protease TMPRSS2, which enables endosome-independent virus entry by a route that avoids antiviral IFITM proteins. SARS-CoV-2 virus lacking the S1/S2 furin cleavage site was shed to lower titres from infected ferrets and was not transmitted to cohoused sentinel animals, unlike wild-type virus. Analysis of 100,000 SARS-CoV-2 sequences derived from patients and 24 human postmortem tissues showed low frequencies of naturally occurring mutants that harbour deletions at the polybasic site. Taken together, our findings reveal that the furin cleavage site is an important determinant of SARS-CoV-2 transmission.
Collapse
|
14
|
Bazotte RB, Hirabara SM, Serdan TAD, Gritte RB, Souza-Siqueira T, Gorjao R, Masi LN, Antunes MM, Cruzat V, Pithon-Curi TC, Curi R. 4-Aminoquinoline compounds from the Spanish flu to COVID-19. Biomed Pharmacother 2021; 135:111138. [PMID: 33360781 PMCID: PMC7973050 DOI: 10.1016/j.biopha.2020.111138] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/11/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/23/2022] Open
Abstract
In 1918, quinine was used as one of the unscientifically based treatments against the H1N1 virus during the Spanish flu pandemic. Originally, quinine was extracted from the bark of Chinchona trees by South American natives of the Amazon forest, and it has been used to treat fever since the seventeenth century. The recent COVID-19 pandemic caused by Sars-Cov-2 infection has forced researchers to search for ways to prevent and treat this disease. Based on the antiviral potential of two 4-aminoquinoline compounds derived from quinine, known as chloroquine (CQ) and hydroxychloroquine (HCQ), clinical investigations for treating COVID-19 are being conducted worldwide. However, there are some discrepancies among the clinical trial outcomes.Thus, even after one hundred years of quinine use during the Spanish flu pandemic, the antiviral properties promoted by 4-aminoquinoline compounds remain unclear. The underlying molecular mechanisms by which CQ and HCQ inhibit viral replication open up the possibility of developing novel analogs of these drugs to combat COVID-19 and other viruses.
Collapse
Affiliation(s)
| | - Sandro Massao Hirabara
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | | | - Raquel Bragante Gritte
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | - Talita Souza-Siqueira
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | - Renata Gorjao
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | - Laureane Nunes Masi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | | | - Vinicius Cruzat
- Faculty of Health, Torrens University Australia, Melbourne, Australia.
| | - Tania Cristina Pithon-Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| | - Rui Curi
- Interdisciplinary Post-Graduate Program in Health Sciences, Cruzeiro do Sul University, São Paulo, SP, Brazil.
| |
Collapse
|
15
|
Homolak J, Kodvanj I, Trkulja V. An Additional Perspective on Proton Pump Inhibitors as Risk Factors for COVID-19. Clin Drug Investig 2021; 41:287-289. [PMID: 33606199 PMCID: PMC7892720 DOI: 10.1007/s40261-021-01007-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 01/23/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Jan Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, 10 000, Zagreb, Croatia.
| | - Ivan Kodvanj
- Department of Pharmacology, University of Zagreb School of Medicine, 10 000, Zagreb, Croatia
| | - Vladimir Trkulja
- Department of Pharmacology, University of Zagreb School of Medicine, 10 000, Zagreb, Croatia
| |
Collapse
|
16
|
Toon K, Bentley EM, Mattiuzzo G. More Than Just Gene Therapy Vectors: Lentiviral Vector Pseudotypes for Serological Investigation. Viruses 2021; 13:217. [PMID: 33572589 PMCID: PMC7911487 DOI: 10.3390/v13020217] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/22/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
Serological assays detecting neutralising antibodies are important for determining the immune responses following infection or vaccination and are also often considered a correlate of protection. The target of neutralising antibodies is usually located in the Envelope protein on the viral surface, which mediates cell entry. As such, presentation of the Envelope protein on a lentiviral particle represents a convenient alternative to handling of a potentially high containment virus or for those viruses with no established cell culture system. The flexibility, relative safety and, in most cases, ease of production of lentiviral pseudotypes, have led to their use in serological assays for many applications such as the evaluation of candidate vaccines, screening and characterization of anti-viral therapeutics, and sero-surveillance. Above all, the speed of production of the lentiviral pseudotypes, once the envelope sequence is published, makes them important tools in the response to viral outbreaks, as shown during the COVID-19 pandemic in 2020. In this review, we provide an overview of the landscape of the serological applications of pseudotyped lentiviral vectors, with a brief discussion on their production and batch quality analysis. Finally, we evaluate their role as surrogates for the real virus and possible alternatives.
Collapse
Affiliation(s)
- Kamilla Toon
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
- Division of Infection and Immunity, University College London, London WC1E 6BT, UK
| | - Emma M. Bentley
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| | - Giada Mattiuzzo
- Division of Virology, National Institute for Biological Standards and Control-MHRA, Blanche Lane, South Mimms EN6 3QG, UK;
| |
Collapse
|
17
|
Chang R, Sun WZ. Repositioning chloroquine as antiviral prophylaxis against COVID-19: potential and challenges. Drug Discov Today 2020; 25:1786-1792. [PMID: 32629169 PMCID: PMC7831558 DOI: 10.1016/j.drudis.2020.06.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/26/2020] [Revised: 06/11/2020] [Accepted: 06/30/2020] [Indexed: 12/23/2022]
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic is advancing globally, and pharmaceutical prophylaxis is one solution. Here, we propose repositioning chloroquine (CQ) as prophylaxis against COVID-19. CQ blocks viral attachment and entry to host cells and demonstrates efficacy against a variety of viruses, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19. Furthermore, CQ is safe, inexpensive, and available. Here, we review the antiviral mechanisms of CQ, its in vitro activity against coronaviruses, its pharmacokinetics (PK) and adverse effects, and why it could be more efficacious as a prophylactic rather than as a therapeutic, given the infection dynamics of SARS-CoV-2. We propose two prophylactic regimens based on efficacy and risk considerations. Although it is largely preclinical data that suggest the potential of CQ, properly planned prophylactic trials and further research are urgently needed.
Collapse
Affiliation(s)
| | - Wei-Zen Sun
- Institute of East-West Medicine, New York, NY, USA; National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
18
|
El Kantar S, Nehmeh B, Saad P, Mitri G, Estephan C, Mroueh M, Akoury E, Taleb RI. Derivatization and combination therapy of current COVID-19 therapeutic agents: a review of mechanistic pathways, adverse effects, and binding sites. Drug Discov Today 2020; 25:1822-1838. [PMID: 32801052 PMCID: PMC7422796 DOI: 10.1016/j.drudis.2020.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/10/2020] [Revised: 07/07/2020] [Accepted: 08/06/2020] [Indexed: 12/12/2022]
Abstract
Current treatment of patients with coronavirus 2019 (COVID-19) involves repurposed drugs that inhibit viral infection by either binding to their respective targets or via modulating cellular signal transduction. However, there is still a great deal of efficacy enhancement through combination therapy and derivatization. Combination therapy should involve agents with significant activity and different mechanisms of action. The structural map of the interaction between a drug and its target protein will help guide drug discovery for devising safe and effective ways to treat COVID-19. Herein, we report numerous synthetic designs based on enhanced affinity to the viral carbohydrate-rich protein spikes and protein-binding sites of COVID-19.
Collapse
Affiliation(s)
- Sally El Kantar
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Bilal Nehmeh
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos Campus, Blat, Lebanon
| | - Philippe Saad
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos Campus, Blat, Lebanon
| | - Gabie Mitri
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos Campus, Blat, Lebanon
| | - Celine Estephan
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos Campus, Blat, Lebanon
| | - Mohamad Mroueh
- School of Pharmacy, Lebanese American University, Byblos Campus, Blat, Lebanon
| | - Elias Akoury
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Beirut 1102-2801, Lebanon
| | - Robin I. Taleb
- School of Arts and Sciences, Department of Natural Sciences, Lebanese American University, Byblos Campus, Blat, Lebanon,Corresponding author:
| |
Collapse
|
19
|
Ray A, Sharma S, Sadasivam B. The Potential Therapeutic Role of Proton Pump Inhibitors in COVID-19: Hypotheses Based on Existing Evidences. Drug Res (Stuttg) 2020; 70:484-488. [PMID: 32877948 PMCID: PMC7672704 DOI: 10.1055/a-1236-3041] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022]
Abstract
Although the major therapeutic uses of the proton pump inhibitors are in gastric-acid related diseases, evidences are suggestive of a pleiotropic nature of the compounds. We comment on the probable pathways and cellular machineries via which proton pump inhibitors could show beneficial therapeutic effects against SARS-CoV-2 based on the existing evidences. Proton pump inhibitors have shown antiviral potencies in various in vivo and in vitro studies. Some of the major possible ways through which they can act against SARS-CoV-2 are by exerting anti-inflammatory and anti-fibrotic effects, via vacuolar ATPase pumps leading to raised endolysosomal pH and by targeting endosomal complexes. The current pandemic has put forward a challenge to find treatment options. Although the potential roles of proton pump inhibitors against SARS-CoV-2 have been discussed in recent publications, the clinical evidences for their real-world effectiveness do not point towards a beneficial effect clearly yet. We suggest that although proton pump inhibitors should strongly be considered as potential therapeutic options for COVID-19, larger studies in the form of randomized controlled trials would be required to arrive at a definite conclusion.
Collapse
Affiliation(s)
- Avik Ray
- Department of Pharmacology, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Swati Sharma
- Department of Pharmacology, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Balakrishnan Sadasivam
- Department of Pharmacology, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| |
Collapse
|
20
|
Abstract
BACKGROUND Chronic obstructive pulmonary disease (COPD) is a common and progressive disease characterised by chronic cough, airflow limitation and recurrent exacerbations. Since COPD exacerbations are linked to rising mortality and reduced quality of life, the condition poses a substantial burden on individuals, society and the healthcare system. Effective management of COPD exacerbations that includes treatment of related conditions in people with COPD is thus recognised as a relevant clinical question and an important research topic. Gastroesophageal reflux disease (GERD) is a known comorbidity of COPD, and pulmonary microaspiration of gastric acid is thought to be a possible cause of COPD exacerbations. Therefore, reducing gastric acid secretion may lead to a reduction in COPD exacerbations. Proton pump inhibitors (PPIs) are one of the most commonly prescribed medications and are recommended as first-line therapy for people with GERD because of their inhibitory effects on gastric acid secretion. Treatment with PPIs may present a viable treatment option for people with COPD. OBJECTIVES To evaluate the efficacy and safety of PPI administration for people with COPD, focusing on COPD-specific outcomes. SEARCH METHODS We searched the Cochrane Airways Register of Trials and conventional clinical trial registers from inception to 22 May 2020. We also screened bibliographies of relevant studies. SELECTION CRITERIA Parallel-group and cluster-randomised controlled trials (RCTs) that compared oral PPIs versus placebo, usual care or low-dose PPIs in adults with COPD were eligible for inclusion. We excluded cross-over RCTs, as well as studies with a duration of less than two months. DATA COLLECTION AND ANALYSIS Two independent review authors screened search results, selected studies for inclusion, extracted study characteristics and outcome data, and assessed risk of bias according to standard Cochrane methodology. We resolved discrepancies by involving a third review author. Primary outcomes of interest were COPD exacerbations, pneumonia and other serious adverse events. Secondary outcomes were quality of life, lung function test indices, acute respiratory infections and disease-specific adverse events. We extracted data on these outcome measures and entered into them into Review Manager software for analysis. MAIN RESULTS The search identified 99 records, and we included one multicentre RCT that randomised 103 adults with COPD. The 12-month RCT compared an oral PPI (lansoprazole) and usual care versus usual care alone. It was conducted at one tertiary care hospital and three secondary care hospitals in Japan. This study recruited participants with a mean age of 75 years, and excluded people with symptoms or history of GERD. No placebo was used in the usual care arm. Among the primary and secondary outcomes of this review, the study only reported data on COPD exacerbations and acute respiratory infections (the common cold). As we only included one study, we could not conduct a meta-analysis. The included study reported that 12 of the 50 people on lansoprazole had at least one exacerbation over a year, compared to 26 out of 50 on usual care (risk ratio 0.46, 95% CI 0.26 to 0.81). The frequency of COPD exacerbations per person in a year was also lower in the PPI plus usual care group than in the usual care alone group(0.34 ± 0.72 vs 1.18 ± 1.40; P < 0.001). The number of people with at least one cold over the year was similar in both groups: 26 people on lansoprazole and 27 people in the usual care group. We judged the evidence to be of low to very low certainty, according to GRADE criteria. The study reported no data on pneumonia and other serious adverse events, quality of life, lung function test indices or disease-specific adverse events. The risk of bias was largely low or unclear for the majority of domains, though the performance bias was a high risk, as the study was not blinded. AUTHORS' CONCLUSIONS Evidence identified by this review is insufficient to determine whether treatment with PPIs is a potential option for COPD. The sample size of the included trial is small, and the evidence is low to very low-certainty. The efficacy and safety profile of PPIs for people with COPD remains uncertain. Future large-scale, high-quality studies are warranted, which investigate major clinical outcomes such as COPD exacerbation rate, serious adverse events and quality of life.
Collapse
Affiliation(s)
- Shino Kikuchi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hissei Imai
- Department of Health Promotion and Human Behavior, Kyoto University Graduate School of Medicine/School of Public Health, Kyoto, Japan
| | - Yoko Tani
- Department of Respiratory Medicine, Graduate School of Medicine,Osaka City University, Osaka, Japan
| | - Tomoko Tajiri
- Department of Respiratory Medicine, Allergy and Clinical Immunology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Norio Watanabe
- Department of Health Promotion and Human Behavior, Kyoto University School of Public Health, Kyoto, Japan
| |
Collapse
|
21
|
Sivaraman H, Er SY, Choong YK, Gavor E, Sivaraman J. Structural Basis of SARS-CoV-2- and SARS-CoV-Receptor Binding and Small-Molecule Blockers as Potential Therapeutics. Annu Rev Pharmacol Toxicol 2020; 61:465-493. [PMID: 32574109 DOI: 10.1146/annurev-pharmtox-061220-093932] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/18/2023]
Abstract
Over the past two decades, deadly coronaviruses, with the most recent being the severe acute respiratory syndrome-related coronavirus-2 (SARS-CoV-2) 2019 pandemic, have majorly challenged public health. The path for virus invasion into humans and other hosts is mediated by host-pathogen interactions, specifically virus-receptor binding. An in-depth understanding of the virus-receptor binding mechanism is a prerequisite for the discovery of vaccines, antibodies, and small-molecule inhibitors that can interrupt this interaction and prevent or cure infection. In this review, we discuss the viral entry mechanism, the known structural aspects of virus-receptor interactions (SARS-CoV-2 S/humanACE2, SARS-CoV S/humanACE2, and MERS-CoV S/humanDPP4), the key protein domains and amino acid residues involved in binding, and the small-molecule inhibitors and other drugs that have (as of June 2020) exhibited therapeutic potential. Specifically, we review the potential clinical utility of two transmembrane serine protease 2 (TMPRSS2)-targeting protease inhibitors, nafamostat mesylate and camostat mesylate, as well as two novel potent fusion inhibitors and the repurposed Ebola drug, remdesivir, which is specific to RNA-dependent RNA polymerase, against human coronaviruses, including SARS-CoV-2.
Collapse
Affiliation(s)
- Hariharan Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore 117543;
| | - Shi Yin Er
- Department of Biological Sciences, National University of Singapore, Singapore 117543;
| | - Yeu Khai Choong
- Department of Biological Sciences, National University of Singapore, Singapore 117543;
| | - Edem Gavor
- Department of Biological Sciences, National University of Singapore, Singapore 117543;
| | - J Sivaraman
- Department of Biological Sciences, National University of Singapore, Singapore 117543;
| |
Collapse
|
22
|
Rodrigo C, Fernando SD, Rajapakse S. Clinical evidence for repurposing chloroquine and hydroxychloroquine as antiviral agents: a systematic review. Clin Microbiol Infect 2020; 26:979-987. [PMID: 32470568 PMCID: PMC7250111 DOI: 10.1016/j.cmi.2020.05.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/08/2020] [Revised: 05/05/2020] [Accepted: 05/16/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Repurposing hydroxychloroquine (HCQ) and chloroquine (CQ) as antiviral agents is a re-emerging topic with the advent of new viral epidemics. AIMS To summarize evidence from human clinical studies for using HCQ or CQ as antiviral agents for any viral infection. SOURCES PubMed, EMBASE, Scopus, Web of Science for published studies without time or language restrictions; Cochrane Clinical Trial Registry and Chinese Clinical Trials Registry for trials registered after 2015; MedRxiv for preprints within the last 12 months. CONTENT Study eligibility criteria were interventional and prospective observational studies (with or without a control group). Participants were adults and children with a confirmed viral infection. Interventions included the use of CQ or HCQ as antiviral agent in one or more groups of the study. Two authors independently screened abstracts, and all authors agreed on eligible studies. A meta-analysis was planned if studies were available which were similar in terms of participants, intervention, comparator and outcomes. Nineteen studies (including two preprints) were eligible (HIV 8, HCV 2, dengue 2, chikungunya 1, COVID-19 6). Nine and ten studies assessed CQ and HCQ respectively. Benefits of either drug for viral load suppression in HIV are inconsistent. CQ is ineffective in curing dengue (high-certainty evidence) and may have little or no benefit in curing chikungunya (low-certainty evidence). The evidence for COVID-19 infection is rapidly evolving but at this stage we are unsure whether either CQ or HCQ has any benefit in clearing viraemia (very-low-certainty evidence). IMPLICATIONS Using HCQ or CQ for HIV/HCV infections is now clinically irrelevant as other effective antivirals are available for viral load suppression (HIV) and cure (HCV). There is no benefit of CQ in dengue, and the same conclusion is likely for chikungunya. More evidence is needed to confirm whether either HCQ or CQ is beneficial in COVID-19 infection.
Collapse
Affiliation(s)
- C Rodrigo
- Department of Pathology, School of Medical Sciences, UNSW Sydney, Australia
| | - S D Fernando
- Department of Parasitology, Faculty of Medicine, University of Colombo, Sri Lanka
| | - S Rajapakse
- Department of Clinical Medicine, Faculty of Medicine, University of Colombo, Sri Lanka.
| |
Collapse
|
23
|
Tripathy S, Dassarma B, Roy S, Chabalala H, Matsabisa MG. A review on possible modes of action of chloroquine/hydroxychloroquine: repurposing against SAR-CoV-2 (COVID-19) pandemic. Int J Antimicrob Agents 2020; 56:106028. [PMID: 32450198 PMCID: PMC7243790 DOI: 10.1016/j.ijantimicag.2020.106028] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2020] [Revised: 05/14/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
Chloroquine (CQ) and its analogue hydroxychloroquine (HCQ) have long been used worldwide as frontline drugs for the treatment and prophylaxis of human malaria. Since the first reported cases in Wuhan, China, in late December 2019, humans have been under threat from coronavirus disease 2019 (COVID-19) caused by the novel coronavirus SARS-CoV-2 (previously known as 2019-nCoV), subsequently declared a pandemic. While the world is searching for expedited approval for a vaccine, which may be only preventative and not a cure, physicians and country leaders are considering several concerted clinical trials suggesting that the age-old antimalarial drugs CQ/HCQ could be a potent therapeutic against COVID-19. Based on accumulating scientific reports, here we highlight the possible modes of action of CQ/HCQ that could justify its use against viral infections. Considering the global health crisis of the COVID-19 pandemic, the option of repurposing old drugs, e.g. CQ/HCQ, particularly HCQ, for the treatment of SARS-CoV-2 infection could be a good choice. CQ/HCQ has diverse modes of action, including alteration of the acidic environment inside lysosomes and late endosomes, preventing endocytosis, exosome release and phagolysosomal fusion, and inhibition of the host cytokine storm. One or more diverse mechanisms might work against viral infections and reduce mortality. As there is no cure for COVID-19, clinical testing of HCQ is urgently required to determine its potency against SARS-CoV-2, as this is the currently available treatment option. There remains a need to find other innovative drug candidates as possible candidates to enter clinical evaluation and testing.
Collapse
Affiliation(s)
- Satyajit Tripathy
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Barsha Dassarma
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| | - Somenath Roy
- Ex-Professor, Department of Human Physiology with Community Health, Vidyasagar University, Paschim Medinipur 721102, India
| | - Hlupheka Chabalala
- IK-based Technology Innovations, Department of Science and Innovations, Brummeria, Pretoria 0001, South Africa
| | - Motlalepula Gilbert Matsabisa
- Department of Pharmacology, School of Medicine, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa.
| |
Collapse
|
24
|
Chen L, Chen H, Dong S, Huang W, Chen L, Wei Y, Shi L, Li J, Zhu F, Zhu Z, Wang Y, Lv X, Yu X, Li H, Wei W, Zhang K, Zhu L, Qu C, Hong J, Hu C, Dong J, Qi R, Lu D, Wang H, Peng S, Hao G. The Effects of Chloroquine and Hydroxychloroquine on ACE2-Related Coronavirus Pathology and the Cardiovascular System: An Evidence-Based Review. FUNCTION 2020; 1:zqaa012. [PMID: 38626250 PMCID: PMC7454642 DOI: 10.1093/function/zqaa012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/26/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 01/08/2023] Open
Abstract
The ongoing pandemic of coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a serious threat to global public health and there is currently no effective antiviral therapy. It has been suggested that chloroquine (CQ) and hydroxychloroquine (HCQ), which were primarily employed as prophylaxis and treatment for malaria, could be used to treat COVID-19. CQ and HCQ may be potential inhibitors of SARS-CoV-2 entry into host cells, which are mediated via the angiotensin-converting enzyme 2 (ACE2), and may also inhibit subsequent intracellular processes which lead to COVID-19, including damage to the cardiovascular (CV) system. However, paradoxically, CQ and HCQ have also been reported to cause damage to the CV system. In this review, we provide a critical examination of the published evidence. CQ and HCQ could potentially be useful drugs in the treatment of COVID-19 and other ACE2 involved virus infections, but the antiviral effects of CQ and HCQ need to be tested in more well-designed clinical randomized studies and their actions on the CV system need to be further elucidated. However, even if it were to turn out that CQ and HCQ are not useful drugs in practice, further studies of their mechanism of action could be helpful in improving our understanding of COVID-19 pathology.
Collapse
Affiliation(s)
- Li Chen
- Department of Medicine, Georgia Prevention Institute, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - Haiyan Chen
- Department of Endemic Disease, Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
| | - Shan Dong
- Guangzhou First People’s Hospital, The Second Affiliated Hospital of South China University of Technology, Guangzhou 510180, China
| | - Wei Huang
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Li Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yuan Wei
- Center for Scientific Research and Institute of Exercise and Health, Guangzhou Sports University, Guangzhou 510500, China
| | - Liping Shi
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinying Li
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Fengfeng Zhu
- Department of Hepatobiliary and Pancreas Surgery, The First Affiliated Hospital Of University of South China, Hengyang 421001, China
| | - Zhu Zhu
- Department of Hepatobiliary and Pancreas Surgery, The First Affiliated Hospital Of University of South China, Hengyang 421001, China
| | - Yiyang Wang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiuxiu Lv
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Xiaohui Yu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Hongmei Li
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Wei Wei
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Keke Zhang
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Lihong Zhu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chen Qu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jian Hong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Chaofeng Hu
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Jun Dong
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Renbin Qi
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Daxiang Lu
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People’s Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Huadong Wang
- Department of Pathophysiology, Key Laboratory of State Administration of Traditional Chinese Medicine of the People’s Republic of China, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Shuang Peng
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Guang Hao
- Department of Epidemiology, School of Medicine, Jinan University, Guangzhou 510632, China
| |
Collapse
|
25
|
Naghipour S, Ghodousi M, Rahsepar S, Elyasi S. Repurposing of well-known medications as antivirals: hydroxychloroquine and chloroquine - from HIV-1 infection to COVID-19. Expert Rev Anti Infect Ther 2020; 18:1119-1133. [PMID: 32631083 DOI: 10.1080/14787210.2020.1792291] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Chloroquine (CQ) and hydroxychloroquine (HCQ) originally were prescribed for prevention or treatment of malaria, but now successfully are used in several rheumatologic diseases. In addition, in recent decades considering their immunomodulatory effects, high tolerably, and low cost, they are evaluated for various viral infections from HIV to COVID-19. AREAS COVERED In this review, we tried to summarize all available studies on HCQ and CQ efficacy for management of viral infections and the probable mechanisms of action. The data were collected by searching 'Hydroxychloroquine,' 'Chloroquine,' 'Viral infection,' and names of various viral infections in PubMed/MEDLINE, Scopus, and Google Scholar databases from commencement to June 2020. Out of 95 search results, 74 most relevant works were gathered. EXPERT OPINION HCQ/CQ showed acceptable efficacy in HIV especially as an adjuvant treatment beside routine HAART. However, for some viral infections such as ZIKA, EBOLA, SARS-CoV, and MERS-CoV, human studies are lacking. In the COVID-19 pandemic, in vitro and preliminary human studies showed encouraging findings. However, later well-designed trials and retrospective studies with large sample size not only reported non-significant efficacy but also showed more cardiac adverse reactions. Alkalinization of acid vesicles is the most important mechanism of action.
Collapse
Affiliation(s)
- Sara Naghipour
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences , Mashhad, Iran
| | - Mahsa Ghodousi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences , Mashhad, Iran
| | - Sara Rahsepar
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences , Mashhad, Iran
| | - Sepideh Elyasi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences , Mashhad, Iran
| |
Collapse
|
26
|
Homolak J, Kodvanj I. Widely available lysosome targeting agents should be considered as potential therapy for COVID-19. Int J Antimicrob Agents 2020; 56:106044. [PMID: 32522674 PMCID: PMC7275137 DOI: 10.1016/j.ijantimicag.2020.106044] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/30/2020] [Revised: 05/21/2020] [Accepted: 05/31/2020] [Indexed: 02/06/2023]
Abstract
Lysosome targeting agents can disrupt endolysosomal maturation and inhibit viral replication Lysosome targeting agents and lysosomotropic drugs should be explored as antiviral drugs for severe acute respiratory syndrome coronavirus-2 Some lysosomotropic drugs are commonly used pharmacological agents Particular attention should be directed towards macrolides (e.g. azithromycin), non-steroidal anti-inflammatory drugs (e.g. indomethacin) and other lysosomotoropic agents
While the coronavirus disease 2019 (COVID-19) pandemic advances, the scientific community continues to struggle in the search for treatments. Several improvements have been made, including discovery of the clinical efficacy of chloroquine (CQ) in patients with COVID-19, but effective treatment protocols remain elusive. In the search for novel treatment options, many scientists have used the in-silico approach to identify compounds that could interfere with the key molecules involved in entrance, replication or dissemination of severe acute respiratory syndrome coronavirus-2. However, most of the identified molecules are not available as pharmacological agents at present, and assessment of their safety and efficacy could take many months. This review took a different approach based on the proposed pharmacodynamic model of CQ in COVID-19. The main mechanism of action responsible for the favourable outcome of patients with COVID-19 treated with CQ seems to be related to a pH-modulation-mediated effect on endolysosomal trafficking, a characteristic of chemical compounds often called ‘lysosomotropic agents’ because of the physico-chemical properties that enable them to diffuse passively through the endosomal membrane and undergo protonation-based trapping in the lumen of the acidic vesicles. This review discusses lysosomotropic and lysosome targeting drugs that are already in clinical use and are characterized by good safety profiles, low cost and wide availability. Some of these drugs –particularly azithromycin and other macrolides, indomethacin and some other non-steroidal anti-inflammatory drugs, proton pump inhibitors and fluoxetine – could provide additional therapeutic benefits in addition to the potential antiviral effect that is still to be confirmed by well-controlled clinical trials. As some of these drugs have probably been used empirically in the treatment of COVID-19, it is hoped that colleagues worldwide will publish patient data to enable evaluation of the potential efficacy of these agents in the clinical context, and rapid implementation in therapeutic protocols if they are shown to have a beneficial effect on clinical outcome.
Collapse
Affiliation(s)
- J Homolak
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb 10000, Croatia.
| | - I Kodvanj
- Department of Pharmacology, University of Zagreb School of Medicine, Salata 11, Zagreb 10000, Croatia
| |
Collapse
|
27
|
Sebastian S, Flaxman A, Cha KM, Ulaszewska M, Gilbride C, Sharpe H, Wright E, Spencer AJ, Dowall S, Hewson R, Gilbert S, Lambe T. A Multi-Filovirus Vaccine Candidate: Co-Expression of Ebola, Sudan, and Marburg Antigens in a Single Vector. Vaccines (Basel) 2020; 8:E241. [PMID: 32455764 PMCID: PMC7349952 DOI: 10.3390/vaccines8020241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/06/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 01/08/2023] Open
Abstract
In the infectious diseases field, protective immunity against individual virus species or strains does not always confer cross-reactive immunity to closely related viruses, leaving individuals susceptible to disease after exposure to related virus species. This is a significant hurdle in the field of vaccine development, in which broadly protective vaccines represent an unmet need. This is particularly evident for filoviruses, as there are multiple family members that can cause lethal haemorrhagic fever, including Zaire ebolavirus, Sudan ebolavirus, and Marburg virus. In an attempt to address this need, both pre-clinical and clinical studies previously used mixed or co-administered monovalent vaccines to prevent filovirus mediated disease. However, these multi-vaccine and multi-dose vaccination regimens do not represent a practical immunisation scheme when considering the target endemic areas. We describe here the development of a single multi-pathogen filovirus vaccine candidate based on a replication-deficient simian adenoviral vector. Our vaccine candidate encodes three different filovirus glycoproteins in one vector and induces strong cellular and humoral immunity to all three viral glycoproteins after a single vaccination. Crucially, it was found to be protective in a stringent Zaire ebolavirus challenge in guinea pigs in a one-shot vaccination regimen. This trivalent filovirus vaccine offers a tenable vaccine product that could be rapidly translated to the clinic to prevent filovirus-mediated viral haemorrhagic fever.
Collapse
Affiliation(s)
- Sarah Sebastian
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
- Current address: Vaccitech Ltd., Oxford Science Park, Oxford OX4 4GE, UK
| | - Amy Flaxman
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Kuan M. Cha
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Marta Ulaszewska
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Ciaran Gilbride
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Hannah Sharpe
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Edward Wright
- School of Life Sciences, University of Sussex, Falmer BN1 9QG, UK;
| | - Alexandra J. Spencer
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Stuart Dowall
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK; (S.D.); (R.H.)
| | - Roger Hewson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK; (S.D.); (R.H.)
| | - Sarah Gilbert
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| | - Teresa Lambe
- Nuffield Department of Medicine, Jenner Institute, University of Oxford, Oxford OX3 7DQ, UK; (S.S.); (A.F.); (K.M.C.); (M.U.); (C.G.); (H.S.); (A.J.S.); (S.G.)
| |
Collapse
|
28
|
Watanabe SM, Ehrlich LS, Strickland M, Li X, Soloveva V, Goff AJ, Stauft CB, Bhaduri-McIntosh S, Tjandra N, Carter C. Selective Targeting of Virus Replication by Proton Pump Inhibitors. Sci Rep 2020; 10:4003. [PMID: 32132561 PMCID: PMC7055211 DOI: 10.1038/s41598-020-60544-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/20/2019] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Two proton pump inhibitors, tenatoprazole and esomeprazole, were previously shown to inhibit HIV-1 egress by blocking the interaction between Tsg101, a member of the ESCRT-I complex, and ubiquitin. Here, we deepen our understanding of prazole budding inhibition by studying a range of viruses in the presence of tenatoprazole. Furthermore, we investigate the relationship between the chemistry of prodrug activation and HIV-1 inhibition for diverse prazoles currently on the market. We report that tenatoprazole is capable of inhibiting the replication of members of the enveloped filo, alpha, and herpes virus families but not the flavivirus group and not the non-enveloped poliovirus. Another key finding is that prazole prodrugs must be activated inside the cell, while their rate of activation in vitro correlated to their efficacy in cells. Our study lays the groundwork for future efforts to repurpose prazole-based compounds as antivirals that are both broad-spectrum and selective in nature.
Collapse
Affiliation(s)
- Susan M Watanabe
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794-5222, USA
| | - Lorna S Ehrlich
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794-5222, USA
| | - Madeleine Strickland
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xiaofan Li
- Department of Pediatrics, Division of Infectious Diseases and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Veronica Soloveva
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD, 21702-5011, USA
| | - Arthur J Goff
- U.S. Army Medical Research Institute of Infectious Diseases, Frederick, MD, 21702-5011, USA
| | - Charles B Stauft
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794-5222, USA
| | - Sumita Bhaduri-McIntosh
- Department of Pediatrics, Division of Infectious Diseases and Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Nico Tjandra
- Laboratory of Molecular Biophysics, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Carol Carter
- Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, 11794-5222, USA.
| |
Collapse
|
29
|
Stankova I, Chuchkov K, Chayrov R, Mukova L, Galabov A, Marinkova D, Danalev D. Adamantane Derivatives Containing Thiazole Moiety: Synthesis, Antiviral and Antibacterial Activity. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09983-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
|
30
|
Abstract
The process of entry into a host cell is a key step in the life cycle of most viruses. In recent years, there has been a significant increase in our understanding of the routes and mechanisms of entry for a number of these viruses. This has led to the development of novel broad-spectrum antiviral approaches that target host cell proteins and pathways, in addition to strategies focused on individual viruses or virus families. Here we consider a number of these approaches and their broad-spectrum potential.
Collapse
Affiliation(s)
- Michela Mazzon
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| | - Mark Marsh
- MRC Laboratory for Molecular Cell Biology, University College London, London, UK
| |
Collapse
|
31
|
The Natural Product Eugenol Is an Inhibitor of the Ebola Virus In Vitro. Pharm Res 2019; 36:104. [PMID: 31101988 DOI: 10.1007/s11095-019-2629-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/21/2019] [Accepted: 04/18/2019] [Indexed: 12/30/2022]
Abstract
PURPOSE Since the 2014 Ebola virus (EBOV) outbreak in West Africa there has been considerable effort towards developing drugs to treat Ebola virus disease and yet to date there is no FDA approved treatment. This is important as at the time of writing this manuscript there is an ongoing outbreak in the Democratic Republic of the Congo which has killed over 1000. METHODS We have evaluated a small number of natural products, some of which had shown antiviral activity against other pathogens. This is exemplified with eugenol, which is found in high concentrations in multiple essential oils, and has shown antiviral activity against feline calicivirus, tomato yellow leaf curl virus, Influenza A virus, Herpes Simplex virus type 1 and 2, and four airborne phages. RESULTS Four compounds possessed EC50 values less than or equal to 11 μM. Of these, eugenol, had an EC50 of 1.3 μM against EBOV and is present in several plants including clove, cinnamon, basil and bay. Eugenol is much smaller and structurally unlike any compound that has been previously identified as an inhibitor of EBOV, therefore it may provide new mechanistic insights. CONCLUSION This compound is readily accessible in bulk quantities, is inexpensive, and has a long history of human consumption, which endorses the idea for further assessment as an antiviral therapeutic. This work also suggests that a more exhaustive assessment of natural product libraries against EBOV and other viruses is warranted to improve our ability to identify compounds that are so distinct from FDA approved drugs.
Collapse
|
32
|
Edwards MR, Basler CF. Current status of small molecule drug development for Ebola virus and other filoviruses. Curr Opin Virol 2019; 35:42-56. [PMID: 31003196 PMCID: PMC6556423 DOI: 10.1016/j.coviro.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/29/2019] [Accepted: 03/12/2019] [Indexed: 12/16/2022]
Abstract
The filovirus family includes some of the deadliest viruses known, including Ebola virus and Marburg virus. These viruses cause periodic outbreaks of severe disease that can be spread from person to person, making the filoviruses important public health threats. There remains a need for approved drugs that target all or most members of this virus family. Small molecule inhibitors that target conserved functions hold promise as pan-filovirus therapeutics. To date, compounds that effectively target virus entry, genome replication, gene expression, and virus egress have been described. The most advanced inhibitors are nucleoside analogs that target viral RNA synthesis reactions.
Collapse
Affiliation(s)
- Megan R Edwards
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States
| | - Christopher F Basler
- Center for Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, United States.
| |
Collapse
|
33
|
Improving Encapsulation of Hydrophilic Chloroquine Diphosphate into Biodegradable Nanoparticles: A Promising Approach against Herpes Virus Simplex-1 Infection. Pharmaceutics 2018; 10:pharmaceutics10040255. [PMID: 30513856 PMCID: PMC6320969 DOI: 10.3390/pharmaceutics10040255] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/28/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023] Open
Abstract
Chloroquine diphosphate (CQ) is a hydrophilic drug with low entrapment efficiency in hydrophobic nanoparticles (NP). Herpes simplex virus type 1 (HSV-1) is an enveloped double-stranded DNA virus worldwide known as a common human pathogen. This study aims to develop chloroquine-loaded poly(lactic acid) (PLA) nanoparticles (CQ-NP) to improve the chloroquine anti- HSV-1 efficacy. CQ-NP were successfully prepared using a modified emulsification-solvent evaporation method. Physicochemical properties of the NP were monitored using dynamic light scattering, atomic force microscopy, drug loading efficiency, and drug release studies. Spherical nanoparticles were produced with modal diameter of <300 nm, zeta potential of −20 mv and encapsulation efficiency of 64.1%. In vitro assays of CQ-NP performed in Vero E6 cells, using the MTT-assay, revealed different cytotoxicity levels. Blank nanoparticles (B-NP) were biocompatible. Finally, the antiviral activity tested by the plaque reduction assay revealed greater efficacy for CQ-NP compared to CQ at concentrations equal to or lower than 20 µg mL−1 (p < 0.001). On the other hand, the B-NP had no antiviral activity. The CQ-NP has shown feasible properties and great potential to improve the antiviral activity of drugs.
Collapse
|
34
|
Bramble MS, Hoff N, Gilchuk P, Mukadi P, Lu K, Doshi RH, Steffen I, Nicholson BP, Lipson A, Vashist N, Sinai C, Spencer D, Olinger G, Wemakoy EO, Illunga BK, Pettitt J, Logue J, Marchand J, Varughese J, Bennett RS, Jahrling P, Cavet G, Serafini T, Ollmann Saphire E, Vilain E, Muyembe-Tamfum JJ, Hensely LE, Simmons G, Crowe JE, Rimoin AW. Pan-Filovirus Serum Neutralizing Antibodies in a Subset of Congolese Ebolavirus Infection Survivors. J Infect Dis 2018; 218:1929-1936. [PMID: 30107445 PMCID: PMC6217721 DOI: 10.1093/infdis/jiy453] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/28/2018] [Accepted: 07/31/2018] [Indexed: 11/30/2022] Open
Abstract
One year after a Zaire ebolavirus (EBOV) outbreak occurred in the Boende Health Zone of the Democratic Republic of the Congo during 2014, we sought to determine the breadth of immune response against diverse filoviruses including EBOV, Bundibugyo (BDBV), Sudan (SUDV), and Marburg (MARV) viruses. After assessing the 15 survivors, 5 individuals demonstrated some degree of reactivity to multiple ebolavirus species and, in some instances, Marburg virus. All 5 of these survivors had immunoreactivity to EBOV glycoprotein (GP) and EBOV VP40, and 4 had reactivity to EBOV nucleoprotein (NP). Three of these survivors showed serologic responses to the 3 species of ebolavirus GPs tested (EBOV, BDBV, SUDV). All 5 samples also exhibited ability to neutralize EBOV using live virus, in a plaque reduction neutralization test. Remarkably, 3 of these EBOV survivors had plasma antibody responses to MARV GP. In pseudovirus neutralization assays, serum antibodies from a subset of these survivors also neutralized EBOV, BDBV, SUDV, and Taï Forest virus as well as MARV. Collectively, these findings suggest that some survivors of naturally acquired ebolavirus infection mount not only a pan-ebolavirus response, but also in less frequent cases, a pan-filovirus neutralizing response.
Collapse
Affiliation(s)
- Matthew S Bramble
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | - Nicole Hoff
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Pavlo Gilchuk
- Vanderbilt Vaccine Center, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Patrick Mukadi
- Institut National de Recherche Biomedicale, Kinshasa, Democratic Republic of the Congo
| | - Kai Lu
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - Reena H Doshi
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Imke Steffen
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - Bradly P Nicholson
- Institute for Medical Research, Durham Veterans Affairs Medical Center, North Carolina
| | - Allen Lipson
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Neerja Vashist
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | - Cyrus Sinai
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - D’andre Spencer
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| | - Garrard Olinger
- Boston University, School of Medicine, Department of Medicine, Massachusetts
| | | | - Benoit Kebela Illunga
- Direction de la Lutte Contre les Maladies, Ministère de la Sante, Kinshasa, Democratic Republic of the Congo
| | - James Pettitt
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - James Logue
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Jonathan Marchand
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Justin Varughese
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Richard S Bennett
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | - Peter Jahrling
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
| | | | | | - Erica Ollmann Saphire
- Skaggs Institute for Chemical Biology, La Jolla, California
- Department of Immunology and Microbial Science, Scripps Research Institute, La Jolla, California
| | - Eric Vilain
- Department of Genetic Medicine Research, Children’s Research Institute, Children’s National Medical Center, Washington, District of Columbia
| | | | - Lisa E Hensely
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Frederick, Maryland
- Emerging Viral Pathogens Section, NIAID, NIH, Frederick, Maryland
| | - Graham Simmons
- Blood Systems Research Institute, and Department of Laboratory Medicine, University of California, San Francisco
| | - James E Crowe
- Vanderbilt Vaccine Center, and Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Departments of Pediatrics and Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anne W Rimoin
- Department of Epidemiology, School of Public Health, University of California, Los Angeles
| |
Collapse
|
35
|
Dhama K, Karthik K, Khandia R, Chakraborty S, Munjal A, Latheef SK, Kumar D, Ramakrishnan MA, Malik YS, Singh R, Malik SVS, Singh RK, Chaicumpa W. Advances in Designing and Developing Vaccines, Drugs, and Therapies to Counter Ebola Virus. Front Immunol 2018; 9:1803. [PMID: 30147687 PMCID: PMC6095993 DOI: 10.3389/fimmu.2018.01803] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2018] [Accepted: 07/23/2018] [Indexed: 01/10/2023] Open
Abstract
Ebola virus (EBOV), a member of the family Filoviridae, is responsible for causing Ebola virus disease (EVD) (formerly named Ebola hemorrhagic fever). This is a severe, often fatal illness with mortality rates varying from 50 to 90% in humans. Although the virus and associated disease has been recognized since 1976, it was only when the recent outbreak of EBOV in 2014-2016 highlighted the danger and global impact of this virus, necessitating the need for coming up with the effective vaccines and drugs to counter its pandemic threat. Albeit no commercial vaccine is available so far against EBOV, a few vaccine candidates are under evaluation and clinical trials to assess their prophylactic efficacy. These include recombinant viral vector (recombinant vesicular stomatitis virus vector, chimpanzee adenovirus type 3-vector, and modified vaccinia Ankara virus), Ebola virus-like particles, virus-like replicon particles, DNA, and plant-based vaccines. Due to improvement in the field of genomics and proteomics, epitope-targeted vaccines have gained top priority. Correspondingly, several therapies have also been developed, including immunoglobulins against specific viral structures small cell-penetrating antibody fragments that target intracellular EBOV proteins. Small interfering RNAs and oligomer-mediated inhibition have also been verified for EVD treatment. Other treatment options include viral entry inhibitors, transfusion of convalescent blood/serum, neutralizing antibodies, and gene expression inhibitors. Repurposed drugs, which have proven safety profiles, can be adapted after high-throughput screening for efficacy and potency for EVD treatment. Herbal and other natural products are also being explored for EVD treatment. Further studies to better understand the pathogenesis and antigenic structures of the virus can help in developing an effective vaccine and identifying appropriate antiviral targets. This review presents the recent advances in designing and developing vaccines, drugs, and therapies to counter the EBOV threat.
Collapse
Affiliation(s)
- Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Rekha Khandia
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Sandip Chakraborty
- Department of Veterinary Microbiology, College of Veterinary Sciences and Animal Husbandry, Agartala, India
| | - Ashok Munjal
- Department of Biochemistry and Genetics, Barkatullah University, Bhopal, India
| | - Shyma K. Latheef
- Immunology Section, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Deepak Kumar
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | | | - Yashpal Singh Malik
- Division of Biological Standardization, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Satya Veer Singh Malik
- Division of Veterinary Public Health, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Raj Kumar Singh
- ICAR-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, India
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine SIriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
36
|
Cong Y, Hart BJ, Gross R, Zhou H, Frieman M, Bollinger L, Wada J, Hensley LE, Jahrling PB, Dyall J, Holbrook MR. MERS-CoV pathogenesis and antiviral efficacy of licensed drugs in human monocyte-derived antigen-presenting cells. PLoS One 2018; 13:e0194868. [PMID: 29566060 PMCID: PMC5864050 DOI: 10.1371/journal.pone.0194868] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/27/2017] [Accepted: 03/12/2018] [Indexed: 12/21/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) presents an emerging threat to public health worldwide by causing severe respiratory disease in humans with high virulence and case fatality rate (about 35%) since 2012. Little is known about the pathogenesis and innate antiviral response in primary human monocyte-derived macrophages (MDMs) and dendritic cells (MDDCs) upon MERS-CoV infection. In this study, we assessed MERS-CoV replication as well as induction of inflammatory cytokines and chemokines in MDMs and immature and mature MDDCs. Immature MDDCs and MDMs were permissive for MERS-CoV infection, while mature MDDCs were not, with stimulation of proinflammatory cytokine and chemokine upregulation in MDMs, but not in MDDCs. To further evaluate the antiviral activity of well-defined drugs in primary antigen presenting cells (APCs), three compounds (chloroquine, chlorpromazine and toremifine), each with broad-spectrum antiviral activity in immortalized cell lines, were evaluated in MDMs and MDDCs to determine their antiviral effect on MERS-CoV infection. While chloroquine was not active in these primary cells, chlorpromazine showed strong anti-MERS-CoV activity, but it was associated with high cytotoxicity narrowing the potential window for drug utilization. Unlike in established cells, toremifene had marginal activity when tested in antigen presenting cells, with high apparent cytotoxicity, also limiting its potential as a therapeutic option. These results demonstrate the value of testing drugs in primary cells, in addition to established cell lines, before initiating preclinical or clinical studies for MERS treatment and the importance of carefully assessing cytotoxicity in drug screen assays. Furthermore, these studies also highlight the role of APCs in stimulating a robust protective immune response to MERS-CoV infection.
Collapse
Affiliation(s)
- Yu Cong
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Brit J. Hart
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Robin Gross
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Huanying Zhou
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Matthew Frieman
- Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, United States of America
| | - Laura Bollinger
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Jiro Wada
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Lisa E. Hensley
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Peter B. Jahrling
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
- Emerging Viral Pathogen Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Julie Dyall
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| | - Michael R. Holbrook
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Ft. Detrick, Frederick, Maryland, United States of America
| |
Collapse
|
37
|
Yu LY, Sun LN, Zhang XH, Li YQ, Yu L, Yuan ZQY, Meng L, Zhang HW, Wang YQ. A Review of the Novel Application and Potential Adverse Effects of Proton Pump Inhibitors. Adv Ther 2017; 34:1070-1086. [PMID: 28429247 PMCID: PMC5427147 DOI: 10.1007/s12325-017-0532-9] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/27/2017] [Indexed: 02/07/2023]
Abstract
Proton pump inhibitors (PPIs) are known as a class of pharmaceutical agents that target H+/K+-ATPase, which is located in gastric parietal cells. PPIs are widely used in the treatment of gastric acid-related diseases including peptic ulcer disease, erosive esophagitis and gastroesophageal reflux disease, and so on. These drugs present an excellent safety profile and have become one of the most commonly prescribed drugs in primary and specialty care. Except for gastric acid-related diseases, PPIs can also be used in the treatment of Helicobacter pylori infection, viral infections, respiratory system diseases, cancer and so on. Although PPIs are mainly used short term in patients with peptic ulcer disease, nowadays these drugs are increasingly used long term, and frequently for a lifetime, for instance in patients with typical or atypical symptoms of gastroesophageal reflux disease and in NSAID or aspirin users at risk of gastrotoxicity and related complications including hemorrhage, perforation and gastric outlet obstruction. Long-term use of PPIs may lead to potential adverse effects, such as osteoporotic fracture, renal damage, infection (pneumonia and clostridium difficile infection), rhabdomyolysis, nutritional deficiencies (vitamin B12, magnesium and iron), anemia and thrombocytopenia. In this article, we will review some novel uses of PPIs in other fields and summarize the underlying adverse reactions.
Collapse
|
38
|
Nie J, Wu X, Ma J, Cao S, Huang W, Liu Q, Li X, Li Y, Wang Y. Development of in vitro and in vivo rabies virus neutralization assays based on a high-titer pseudovirus system. Sci Rep 2017; 7:42769. [PMID: 28218278 PMCID: PMC5316940 DOI: 10.1038/srep42769] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/02/2016] [Accepted: 01/13/2017] [Indexed: 12/25/2022] Open
Abstract
Pseudoviruses are useful virological tools because of their safety and versatility; however the low titer of these viruses substantially limits their wider applications. We developed a highly efficient pseudovirus production system capable of yielding 100 times more rabies pseudovirus than the traditional method. Employing the high-titer pseudoviruses, we have developed robust in vitro and in vivo neutralization assays for the evaluation of rabies vaccine, which traditionally relies on live-virus based assays. Compared with current rapid fluorescent focus inhibition test (RFFIT), our in vitro pseudovirus-based neutralization assay (PBNA) is much less labor-intensive while demonstrating better reproducibility. Moreover, the in vivo PBNA assay was also found to be superior to the live virus based assay. Following intravenous administration, the pseudovirus effectively infected the mice, with dynamic viral distributions being sequentially observed in spleen, liver and brain. Furthermore, data from in vivo PBNA showed great agreement with those generated from the live virus model but with the experimental time significantly reduced from 2 weeks to 3 days. Taken together, the effective pseudovirus production system facilitated the development of novel PBNA assays which could replace live virus-based traditional assays due to its safety, rapidity, reproducibility and high throughput capacity.
Collapse
Affiliation(s)
- Jianhui Nie
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Xiaohong Wu
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Jian Ma
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Shouchun Cao
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Qiang Liu
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, On K1A 0K9, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, On, Canada
| | - Yuhua Li
- Division of Arboviral Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), No. 2 Tiantanxili, Beijing 100050, China
| |
Collapse
|
39
|
Sweiti H, Ekwunife O, Jaschinski T, Lhachimi SK. Repurposed Therapeutic Agents Targeting the Ebola Virus: A Systematic Review. CURRENT THERAPEUTIC RESEARCH 2017; 84:10-21. [PMID: 28761574 PMCID: PMC5522984 DOI: 10.1016/j.curtheres.2017.01.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Accepted: 01/30/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND The Ebola virus has been responsible for numerous outbreaks since the 1970s, with the most recent outbreak taking place between 2014 and 2016 and causing an international public health emergency. Ebola virus disease (EVD) has a high mortality rate and no approved targeted treatment exists to date. A number of established drugs are being considered as potential therapeutic agents for the treatment of EVD. OBJECTIVE We aimed to identify potential drug repositioning candidates and to assess the scientific evidence available on their efficacy. METHODS We conducted a systematic literature search in MEDLINE, Embase, and other relevant trial registry platforms for studies published between January 1976 and January 2017. We included drug screening, preclinical studies, and clinical studies on repurposed drugs for the treatment of EVD. The risk of bias for animal studies and nonrandomized clinical studies was assessed. The quality of reporting for case series and case reports was evaluated. Finally, we selected drugs approved by established regulatory authorities, which have positive in vitro study outcomes and at least one additional animal or clinical trial. RESULTS We identified 3301 publications, of which 37 studies fulfilled our inclusion criteria. Studies were highly heterogeneous in terms of study type, methodology, and intervention. The risk of bias was high for 13 out of 14 animal studies. We selected 11 drugs with potential anti-EVD therapeutic effects and summarized their evidence. CONCLUSIONS Several established drugs may have therapeutic effects on EVD, but the quality and quantity of current scientific evidence is lacking. This review highlights the need for well-designed and conducted preclinical and clinical research to establish the efficacy of potential repurposed drugs against EVD.
Collapse
Affiliation(s)
- Hussein Sweiti
- Institute of Health Services Research and Health Economics, School of Medicine, Heinrich-Heine University Dû¥sseldorf, Dû¥sseldorf, Germany
- Surgical Department, Klinikum Frankfurt HûÑchst, Frankfurt, Germany
| | - Obinna Ekwunife
- Cooperative Research Group for Evidence-Based Public Health, Department of Prevention and Evaluation, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
- Department of Clinical Pharmacy and Pharmacy Management, Nnamdi Azikiwe University, Awka, Nigeria
| | - Thomas Jaschinski
- Department for Evidence-based Health Services Research, Institute for Research in Operative Medicine, Witten/Herdecke University, Witten, Germany
| | - Stefan K. Lhachimi
- Institute of Health Services Research and Health Economics, School of Medicine, Heinrich-Heine University Dû¥sseldorf, Dû¥sseldorf, Germany
- Cooperative Research Group for Evidence-Based Public Health, Department of Prevention and Evaluation, Leibniz Institute for Prevention Research and Epidemiology-BIPS, Bremen, Germany
- Institute for Public Health, Health Sciences Bremen, University of Bremen, Bremen, Germany
| |
Collapse
|
40
|
Al-Bari MAA. Targeting endosomal acidification by chloroquine analogs as a promising strategy for the treatment of emerging viral diseases. Pharmacol Res Perspect 2017; 5:e00293. [PMID: 28596841 PMCID: PMC5461643 DOI: 10.1002/prp2.293] [Citation(s) in RCA: 248] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/25/2016] [Revised: 11/15/2016] [Accepted: 12/07/2016] [Indexed: 12/13/2022] Open
Abstract
Emerging viruses such as HIV, dengue, influenza A, SARS coronavirus, Ebola, and other viruses pose a significant threat to human health. Majority of these viruses are responsible for the outbreaks of pathogenic lethal infections. To date, there are no effective therapeutic strategies available for the prophylaxis and treatment of these infections. Chloroquine analogs have been used for decades as the primary and most successful drugs against malaria. Concomitant with the emergence of chloroquine‐resistant Plasmodium strains and a subsequent decrease in the use as antimalarial drugs, other applications of the analogs have been investigated. Since the analogs have interesting biochemical properties, these drugs are found to be effective against a wide variety of viral infections. As antiviral action, the analogs have been shown to inhibit acidification of endosome during the events of replication and infection. Moreover, immunomodulatory effects of analogs have been beneficial to patients with severe inflammatory complications of several viral diseases. Interestingly, one of the successful targeting strategies is the inhibition of HIV replication by the analogs in vitro which are being tested in several clinical trials. This review focuses on the potentialities of chloroquine analogs for the treatment of endosomal low pH dependent emerging viral diseases.
Collapse
|
41
|
Miller JL, Spiro SG, Dowall SD, Taylor I, Rule A, Alonzi DS, Sayce AC, Wright E, Bentley EM, Thom R, Hall G, Dwek RA, Hewson R, Zitzmann N. Minimal In Vivo Efficacy of Iminosugars in a Lethal Ebola Virus Guinea Pig Model. PLoS One 2016; 11:e0167018. [PMID: 27880800 PMCID: PMC5120828 DOI: 10.1371/journal.pone.0167018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/25/2016] [Accepted: 11/07/2016] [Indexed: 11/29/2022] Open
Abstract
The antiviral properties of iminosugars have been reported previously in vitro and in small animal models against Ebola virus (EBOV); however, their effects have not been tested in larger animal models such as guinea pigs. We tested the iminosugars N-butyl-deoxynojirimycin (NB-DNJ) and N-(9-methoxynonyl)-1deoxynojirimycin (MON-DNJ) for safety in uninfected animals, and for antiviral efficacy in animals infected with a lethal dose of guinea pig adapted EBOV. 1850 mg/kg/day NB-DNJ and 120 mg/kg/day MON-DNJ administered intravenously, three times daily, caused no adverse effects and were well tolerated. A pilot study treating infected animals three times within an 8 hour period was promising with 1 of 4 infected NB-DNJ treated animals surviving and the remaining three showing improved clinical signs. MON-DNJ showed no protective effects when EBOV-infected guinea pigs were treated. On histopathological examination, animals treated with NB-DNJ had reduced lesion severity in liver and spleen. However, a second study, in which NB-DNJ was administered at equally-spaced 8 hour intervals, could not confirm drug-associated benefits. Neither was any antiviral effect of iminosugars detected in an EBOV glycoprotein pseudotyped virus assay. Overall, this study provides evidence that NB-DNJ and MON-DNJ do not protect guinea pigs from a lethal EBOV-infection at the dose levels and regimens tested. However, the one surviving animal and signs of improvements in three animals of the NB-DNJ treated cohort could indicate that NB-DNJ at these levels may have a marginal beneficial effect. Future work could be focused on the development of more potent iminosugars.
Collapse
Affiliation(s)
- Joanna L. Miller
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
- * E-mail: (NZ); (JLM)
| | - Simon G. Spiro
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
- The Royal Veterinary College, London, United Kingdom
| | | | - Irene Taylor
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Antony Rule
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Dominic S. Alonzi
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| | - Andrew C. Sayce
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| | - Edward Wright
- Viral Pseudotype Unit, Faculty of Science and Technology, University of Westminster, London, United Kingdom
| | - Emma M. Bentley
- Viral Pseudotype Unit, Faculty of Science and Technology, University of Westminster, London, United Kingdom
| | - Ruth Thom
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Graham Hall
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Raymond A. Dwek
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
| | - Roger Hewson
- Public Health England, Porton Down, Salisbury, United Kingdom
| | - Nicole Zitzmann
- Antiviral Research Unit, Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford, OX1 3QU, United Kingdom
- * E-mail: (NZ); (JLM)
| |
Collapse
|
42
|
Antiviral Screening of Multiple Compounds against Ebola Virus. Viruses 2016; 8:v8110277. [PMID: 27801778 PMCID: PMC5127007 DOI: 10.3390/v8110277] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2016] [Revised: 10/19/2016] [Accepted: 10/19/2016] [Indexed: 01/04/2023] Open
Abstract
In light of the recent outbreak of Ebola virus (EBOV) disease in West Africa, there have been renewed efforts to search for effective antiviral countermeasures. A range of compounds currently available with broad antimicrobial activity have been tested for activity against EBOV. Using live EBOV, eighteen candidate compounds were screened for antiviral activity in vitro. The compounds were selected on a rational basis because their mechanisms of action suggested that they had the potential to disrupt EBOV entry, replication or exit from cells or because they had displayed some antiviral activity against EBOV in previous tests. Nine compounds caused no reduction in viral replication despite cells remaining healthy, so they were excluded from further analysis (zidovudine; didanosine; stavudine; abacavir sulphate; entecavir; JB1a; Aimspro; celgosivir; and castanospermine). A second screen of the remaining compounds and the feasibility of appropriateness for in vivo testing removed six further compounds (ouabain; omeprazole; esomeprazole; Gleevec; D-LANA-14; and Tasigna). The three most promising compounds (17-DMAG; BGB324; and NCK-8) were further screened for in vivo activity in the guinea pig model of EBOV disease. Two of the compounds, BGB324 and NCK-8, showed some effect against lethal infection in vivo at the concentrations tested, which warrants further investigation. Further, these data add to the body of knowledge on the antiviral activities of multiple compounds against EBOV and indicate that the scientific community should invest more effort into the development of novel and specific antiviral compounds to treat Ebola virus disease.
Collapse
|
43
|
Integrated Computational Approach for Virtual Hit Identification against Ebola Viral Proteins VP35 and VP40. Int J Mol Sci 2016; 17:ijms17111748. [PMID: 27792169 PMCID: PMC5133775 DOI: 10.3390/ijms17111748] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/28/2016] [Revised: 09/18/2016] [Accepted: 09/22/2016] [Indexed: 12/30/2022] Open
Abstract
The Ebola virus (EBOV) has been recognised for nearly 40 years, with the most recent EBOV outbreak being in West Africa, where it created a humanitarian crisis. Mortalities reported up to 30 March 2016 totalled 11,307. However, up until now, EBOV drugs have been far from achieving regulatory (FDA) approval. It is therefore essential to identify parent compounds that have the potential to be developed into effective drugs. Studies on Ebola viral proteins have shown that some can elicit an immunological response in mice, and these are now considered essential components of a vaccine designed to protect against Ebola haemorrhagic fever. The current study focuses on chemoinformatic approaches to identify virtual hits against Ebola viral proteins (VP35 and VP40), including protein binding site prediction, drug-likeness, pharmacokinetic and pharmacodynamic properties, metabolic site prediction, and molecular docking. Retrospective validation was performed using a database of non-active compounds, and early enrichment of EBOV actives at different false positive rates was calculated. Homology modelling and subsequent superimposition of binding site residues on other strains of EBOV were carried out to check residual conformations, and hence to confirm the efficacy of potential compounds. As a mechanism for artefactual inhibition of proteins through non-specific compounds, virtual hits were assessed for their aggregator potential compared with previously reported aggregators. These systematic studies have indicated that a few compounds may be effective inhibitors of EBOV replication and therefore might have the potential to be developed as anti-EBOV drugs after subsequent testing and validation in experiments in vivo.
Collapse
|
44
|
Filovirus proteins for antiviral drug discovery: A structure/function analysis of surface glycoproteins and virus entry. Antiviral Res 2016; 135:1-14. [PMID: 27640102 PMCID: PMC7113884 DOI: 10.1016/j.antiviral.2016.09.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/29/2016] [Revised: 09/01/2016] [Accepted: 09/05/2016] [Indexed: 12/20/2022]
Abstract
This review focuses on the recent progress in our understanding of filovirus protein structure/function and its impact on antiviral research. Here we focus on the surface glycoprotein GP1,2 and its different roles in filovirus entry. We first describe the latest advances on the characterization of GP gene-overlapping proteins sGP, ssGP and Δ-peptide. Then, we compare filovirus surface GP1,2 proteins in terms of structure, synthesis and function. As they bear potential in drug-design, the discovery of small organic compounds inhibiting filovirus entry is a currently very active field. Although it is at an early stage, the development of antiviral drugs against Ebola and Marburg virus entry might prove essential to reduce outbreak-associated fatality rates through post-exposure treatment of both suspected and confirmed cases. The filovirus surface glycoprotein is the key player protein responsible for viral entry. Secreted forms of the glycoprotein have been suggested to participate to filovirus virus pathogenicity. Recent structural insights of the filovirus surface glycoprotein highlight new antiviral perspectives. Interesting compounds and innovative antiviral strategies emerge from research and development to inhibit filovirus entry.
Collapse
|
45
|
Dowall SD, Bosworth A, Watson R, Bewley K, Taylor I, Rayner E, Hunter L, Pearson G, Easterbrook L, Pitman J, Hewson R, Carroll MW. Chloroquine inhibited Ebola virus replication in vitro but failed to protect against infection and disease in the in vivo guinea pig model. J Gen Virol 2016; 96:3484-3492. [PMID: 26459826 DOI: 10.1099/jgv.0.000309] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022] Open
Abstract
Ebola virus (EBOV) is highly pathogenic, with a predisposition to cause outbreaks in human populations accompanied by significant mortality. Owing to the lack of approved therapies, screening programmes of potentially efficacious drugs have been undertaken. One of these studies has demonstrated the possible utility of chloroquine against EBOV using pseudotyped assays. In mouse models of EBOV disease there are conflicting reports of the therapeutic effects of chloroquine. There are currently no reports of its efficacy using the larger and more stringent guinea pig model of infection. In this study we have shown that replication of live EBOV is impaired by chloroquine in vitro. However, no protective effects were observed in vivo when EBOV-infected guinea pigs were treated with chloroquine. These results advocate that chloroquine should not be considered as a treatment strategy for EBOV.
Collapse
Affiliation(s)
- Stuart D Dowall
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Andrew Bosworth
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Robert Watson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Kevin Bewley
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Irene Taylor
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Emma Rayner
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Laura Hunter
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Geoff Pearson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Linda Easterbrook
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - James Pitman
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Roger Hewson
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| | - Miles W Carroll
- Public Health England, Porton Down, Salisbury, Wiltshire SP4 0JG, UK
| |
Collapse
|
46
|
Akpovwa H. Chloroquine could be used for the treatment of filoviral infections and other viral infections that emerge or emerged from viruses requiring an acidic pH for infectivity. Cell Biochem Funct 2016; 34:191-6. [PMID: 27001679 PMCID: PMC5071688 DOI: 10.1002/cbf.3182] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/10/2015] [Revised: 02/16/2016] [Accepted: 02/29/2016] [Indexed: 12/24/2022]
Abstract
Viruses from the Filoviridae family, as many other virus families, require an acidic pH for successful infection and are therefore susceptible to the actions of 4‐aminoquinolines, such as chloroquine. Although the mechanisms of action of chloroquine clearly indicate that it might inhibit filoviral infections, several clinical trials that attempted to use chloroquine in the treatment of other acute viral infections – including dengue and influenza A and B – caused by low pH‐dependent viruses, have reported that chloroquine had no clinical efficacy, and these results demoted chloroquine from the potential treatments for other virus families requiring low pH for infectivity. The present review is aimed at investigating whether chloroquine could combat the present Ebola virus epidemic, and also at exploring the main reasons for the reported lack of efficacy. Literature was sourced from PubMed, Scopus, Google Scholar, reference list of articles and textbooks – Fields Virology (Volumes 1and 2), the cytokine handbook, Pharmacology in Medicine: Principles and Practice, and hydroxychloroquine and chloroquine retinopathy. The present analysis concludes that (1) chloroquine might find a place in the treatment of Ebola, either as a monotherapy or in combination therapies; (2) the ineffectiveness of chloroquine, or its analogue, hydroxychloroquine, at treating infections from low pH‐dependent viruses is a result of the failure to attain and sustain a steady state concentration sufficient to increase and keep the pH of the acidic organelles to approximately neutral levels; (3) to successfully treat filoviral infections – or other viral infections that emerge or emerged from low pH‐dependent viruses – a steady state chloroquine plasma concentration of at least 1 µg/mL(~3.125 μM/L) or a whole blood concentration of 16 μM/L must be achieved and be sustained until the patients' viraemia becomes undetectable. These concentrations, however, do not rule out the efficacy of other, higher, steady state concentrations – although such concentrations might be accompanied by severe adverse effects or toxicities. The feasibility of the conclusion in the preceding texts has recently been supported by a subsequent study that shows that amodiaquine, a derivative of CQ, is able to protect humans infected with Ebola from death.
Collapse
|
47
|
Ekins S, Freundlich JS, Clark AM, Anantpadma M, Davey RA, Madrid P. Machine learning models identify molecules active against the Ebola virus in vitro. F1000Res 2016; 4:1091. [PMID: 26834994 DOI: 10.12688/f1000research.7217.2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 12/23/2015] [Indexed: 12/15/2022] Open
Abstract
The search for small molecule inhibitors of Ebola virus (EBOV) has led to several high throughput screens over the past 3 years. These have identified a range of FDA-approved active pharmaceutical ingredients (APIs) with anti-EBOV activity in vitro and several of which are also active in a mouse infection model. There are millions of additional commercially-available molecules that could be screened for potential activities as anti-EBOV compounds. One way to prioritize compounds for testing is to generate computational models based on the high throughput screening data and then virtually screen compound libraries. In the current study, we have generated Bayesian machine learning models with viral pseudotype entry assay and the EBOV replication assay data. We have validated the models internally and externally. We have also used these models to computationally score the MicroSource library of drugs to select those likely to be potential inhibitors. Three of the highest scoring molecules that were not in the model training sets, quinacrine, pyronaridine and tilorone, were tested in vitro and had EC50 values of 350, 420 and 230 nM, respectively. Pyronaridine is a component of a combination therapy for malaria that was recently approved by the European Medicines Agency, which may make it more readily accessible for clinical testing. Like other known antimalarial drugs active against EBOV, it shares the 4-aminoquinoline scaffold. Tilorone, is an investigational antiviral agent that has shown a broad array of biological activities including cell growth inhibition in cancer cells, antifibrotic properties, α7 nicotinic receptor agonist activity, radioprotective activity and activation of hypoxia inducible factor-1. Quinacrine is an antimalarial but also has use as an anthelmintic. Our results suggest data sets with less than 1,000 molecules can produce validated machine learning models that can in turn be utilized to identify novel EBOV inhibitors in vitro.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations in Chemistry, Fuquay-Varina, NC, 27526, USA
- Collaborations Pharmaceuticals Inc, Fuquay-Varina, NC, 27526, USA
- Collaborative Drug Discovery, Burlingame, CA, 94010, USA
| | - Joel S Freundlich
- Departments of Pharmacology & Physiology and Medicine, Center for Emerging and Reemerging Pathogens, UMDNJ, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alex M Clark
- Molecular Materials Informatics, Inc., Montreal, 94025, Canada
| | - Manu Anantpadma
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Robert A Davey
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | | |
Collapse
|
48
|
Ekins S, Freundlich JS, Clark AM, Anantpadma M, Davey RA, Madrid P. Machine learning models identify molecules active against the Ebola virus in vitro. F1000Res 2015; 4:1091. [PMID: 26834994 DOI: 10.12688/f1000research.7217.1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 10/15/2015] [Indexed: 12/23/2022] Open
Abstract
The search for small molecule inhibitors of Ebola virus (EBOV) has led to several high throughput screens over the past 3 years. These have identified a range of FDA-approved active pharmaceutical ingredients (APIs) with anti-EBOV activity in vitro and several of which are also active in a mouse infection model. There are millions of additional commercially-available molecules that could be screened for potential activities as anti-EBOV compounds. One way to prioritize compounds for testing is to generate computational models based on the high throughput screening data and then virtually screen compound libraries. In the current study, we have generated Bayesian machine learning models with viral pseudotype entry assay and the EBOV replication assay data. We have validated the models internally and externally. We have also used these models to computationally score the MicroSource library of drugs to select those likely to be potential inhibitors. Three of the highest scoring molecules that were not in the model training sets, quinacrine, pyronaridine and tilorone, were tested in vitro and had EC 50 values of 350, 420 and 230 nM, respectively. Pyronaridine is a component of a combination therapy for malaria that was recently approved by the European Medicines Agency, which may make it more readily accessible for clinical testing. Like other known antimalarial drugs active against EBOV, it shares the 4-aminoquinoline scaffold. Tilorone, is an investigational antiviral agent that has shown a broad array of biological activities including cell growth inhibition in cancer cells, antifibrotic properties, α7 nicotinic receptor agonist activity, radioprotective activity and activation of hypoxia inducible factor-1. Quinacrine is an antimalarial but also has use as an anthelmintic. Our results suggest data sets with less than 1,000 molecules can produce validated machine learning models that can in turn be utilized to identify novel EBOV inhibitors in vitro.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations in Chemistry, Fuquay-Varina, NC, 27526, USA.,Collaborations Pharmaceuticals Inc, Fuquay-Varina, NC, 27526, USA.,Collaborative Drug Discovery, Burlingame, CA, 94010, USA
| | - Joel S Freundlich
- Departments of Pharmacology & Physiology and Medicine, Center for Emerging and Reemerging Pathogens, UMDNJ, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alex M Clark
- Molecular Materials Informatics, Inc., Montreal, 94025, Canada
| | - Manu Anantpadma
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Robert A Davey
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | | |
Collapse
|
49
|
Ekins S, Freundlich JS, Clark AM, Anantpadma M, Davey RA, Madrid P. Machine learning models identify molecules active against the Ebola virus in vitro. F1000Res 2015; 4:1091. [PMID: 26834994 PMCID: PMC4706063 DOI: 10.12688/f1000research.7217.3] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Accepted: 01/17/2017] [Indexed: 12/21/2022] Open
Abstract
The search for small molecule inhibitors of Ebola virus (EBOV) has led to several high throughput screens over the past 3 years. These have identified a range of FDA-approved active pharmaceutical ingredients (APIs) with anti-EBOV activity
in vitro and several of which are also active in a mouse infection model. There are millions of additional commercially-available molecules that could be screened for potential activities as anti-EBOV compounds. One way to prioritize compounds for testing is to generate computational models based on the high throughput screening data and then virtually screen compound libraries. In the current study, we have generated Bayesian machine learning models with viral pseudotype entry assay and the EBOV replication assay data. We have validated the models internally and externally. We have also used these models to computationally score the MicroSource library of drugs to select those likely to be potential inhibitors. Three of the highest scoring molecules that were not in the model training sets, quinacrine, pyronaridine and tilorone, were tested
in vitro and had EC
50 values of 350, 420 and 230 nM, respectively. Pyronaridine is a component of a combination therapy for malaria that was recently approved by the European Medicines Agency, which may make it more readily accessible for clinical testing. Like other known antimalarial drugs active against EBOV, it shares the 4-aminoquinoline scaffold. Tilorone, is an investigational antiviral agent that has shown a broad array of biological activities including cell growth inhibition in cancer cells, antifibrotic properties, α7 nicotinic receptor agonist activity, radioprotective activity and activation of hypoxia inducible factor-1. Quinacrine is an antimalarial but also has use as an anthelmintic. Our results suggest data sets with less than 1,000 molecules can produce validated machine learning models that can in turn be utilized to identify novel EBOV inhibitors
in vitro.
Collapse
Affiliation(s)
- Sean Ekins
- Collaborations in Chemistry, Fuquay-Varina, NC, 27526, USA.,Collaborations Pharmaceuticals Inc, Fuquay-Varina, NC, 27526, USA.,Collaborative Drug Discovery, Burlingame, CA, 94010, USA
| | - Joel S Freundlich
- Departments of Pharmacology & Physiology and Medicine, Center for Emerging and Reemerging Pathogens, UMDNJ, New Jersey Medical School, Newark, NJ, 07103, USA
| | - Alex M Clark
- Molecular Materials Informatics, Inc., Montreal, 94025, Canada
| | - Manu Anantpadma
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | - Robert A Davey
- Texas Biomedical Research Institute, San Antonio, TX, 78227, USA
| | | |
Collapse
|
50
|
Savarino A, Shytaj IL. Chloroquine and beyond: exploring anti-rheumatic drugs to reduce immune hyperactivation in HIV/AIDS. Retrovirology 2015; 12:51. [PMID: 26084487 PMCID: PMC4472405 DOI: 10.1186/s12977-015-0178-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/11/2015] [Accepted: 05/30/2015] [Indexed: 11/30/2022] Open
Abstract
The restoration of the immune system prompted by antiretroviral therapy (ART) has allowed drastically reducing the mortality and morbidity of HIV infection. However, one main source of clinical concern is the persistence of immune hyperactivation in individuals under ART. Chronically enhanced levels of T-cell activation are associated with several deleterious effects which lead to faster disease progression and slower CD4+ T-cell recovery during ART. In this article, we discuss the rationale, and review the results, of the use of antimalarial quinolines, such as chloroquine and its derivative hydroxychloroquine, to counteract immune activation in HIV infection. Despite the promising results of several pilot trials, the most recent clinical data indicate that antimalarial quinolines are unlikely to exert a marked beneficial effect on immune activation. Alternative approaches will likely be required to reproducibly decrease immune activation in the setting of HIV infection. If the quinoline-based strategies should nevertheless be pursued in future studies, particular care must be devoted to the dosage selection, in order to maximize the chances to obtain effective in vivo drug concentrations.
Collapse
Affiliation(s)
- Andrea Savarino
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| | - Iart Luca Shytaj
- Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161, Rome, Italy.
| |
Collapse
|