1
|
Datta G, Rezagholizadeh N, Hasler WA, Khan N, Chen X. SLC38A9 regulates SARS-CoV-2 viral entry. iScience 2024; 27:110387. [PMID: 39071889 PMCID: PMC11277692 DOI: 10.1016/j.isci.2024.110387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/13/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
SARS-CoV-2 viral entry into host cells depends on the cleavage of spike (S) protein into S1 and S2 proteins. Such proteolytic cleavage by furin results in the exposure of a multibasic motif on S1, which is critical for SARS-CoV-2 viral infection and transmission; however, how such a multibasic motif contributes to the infection of SARS-CoV-2 remains elusive. Here, we demonstrate that the multibasic motif on S1 is critical for its interaction with SLC38A9, an endolysosome-resident arginine sensor. SLC38A9 knockdown prevents S1-induced endolysosome de-acidification and blocks the S protein-mediated entry of pseudo-SARS-CoV-2 in Calu-3, U87MG, Caco-2, and A549 cells. Our findings provide a novel mechanism in regulating SARS-CoV-2 viral entry; S1 present in endolysosome lumen could interact with SLC38A9, which mediates S1-induced endolysosome de-acidification and dysfunction, facilitating the escape of SARS-CoV-2 from endolysosomes and enhancing viral entry.
Collapse
Affiliation(s)
- Gaurav Datta
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Neda Rezagholizadeh
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Wendie A. Hasler
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Nabab Khan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| |
Collapse
|
2
|
Vieux N, Perrier Q, Bedouch P, Epaulard O. Much ado about nothing? Discrepancy between the available data on the antiviral effect of hydroxychloroquine in March 2020 and its inclusion in COVID-19 clinical trials and outpatient prescriptions. Public Health 2023; 225:35-44. [PMID: 37918175 DOI: 10.1016/j.puhe.2023.09.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 07/10/2023] [Accepted: 09/24/2023] [Indexed: 11/04/2023]
Abstract
OBJECTIVES Many of the 2020 COVID-19 clinical trials included an (hydroxy)chloroquine ((H)CQ) arm. We aimed to juxtapose the state of science before April 2020 regarding the benefits of (H)CQ for viral infections with the number and size of the clinical trials studying (H)CQ and the volume of (H)CQ dispensed in France. STUDY DESIGN We identified and analysed published scientific material regarding the antiviral activity of (H)CQ and publicly available data regarding clinical trials and drug dispensation in France. METHODS We conducted a review of scientific publications available before April 2020 and a systematic analysis of COVID-19 clinical trials featuring (H)CQ registered on clinicaltrials.gov. RESULTS Before April 2020, 894 scientific publications mentioning (H)CQ for viruses other than coronaviruses were available, including 35 in vitro studies (reporting an inconstant inhibition of viral replication), 11 preclinical studies (reporting no or disputable positive effects), and 32 clinical trials (reporting no or disputable positive effects). Moreover, 67 publications on (H)CQ and coronavirus infections were available, including 12 in vitro studies (reporting an inconstant inhibition of viral replication), two preclinical studies (reporting contradictory results), and no clinical trials. Meanwhile, 253 therapeutic clinical trials featuring an HCQ arm were registered in 2020, intending to enrol 246,623 patients. CONCLUSIONS The number and size of (H)CQ clinical trials for COVID-19 launched in 2020 were not supported by the literature published before April 2020.
Collapse
Affiliation(s)
- N Vieux
- Pôle Pharmacie, Université Grenoble Alpes, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France
| | - Q Perrier
- Pôle Pharmacie, Université Grenoble Alpes, Centre Hospitalier Universitaire Grenoble Alpes, Laboratory of Fundamental and Applied Bioenergetic (LBFA), INSERM U1055, Grenoble, France
| | - P Bedouch
- Pôle Pharmacie, Université Grenoble Alpes, Centre Hospitalier Universitaire Grenoble Alpes, CNRS, UMR 5525, VetAgro Sup, Grenoble INP, CHU Grenoble Alpes, TIMC, 38000 Grenoble, France
| | - O Epaulard
- Infectious Disease Department, Université Grenoble Alpes, Centre Hospitalier Universitaire Grenoble Alpes, Grenoble, France; Groupe de Recherche en Infectiologie Clinique, CIC-1406, INSERM-UGA-CHUGA, France.
| |
Collapse
|
3
|
Aganovic A. pH-dependent endocytosis mechanisms for influenza A and SARS-coronavirus. Front Microbiol 2023; 14:1190463. [PMID: 37234537 PMCID: PMC10206014 DOI: 10.3389/fmicb.2023.1190463] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic and the influenza epidemics have revived the interest in understanding how these highly contagious enveloped viruses respond to alterations in the physicochemical properties of their microenvironment. By understanding the mechanisms and conditions by which viruses exploit the pH environment of the host cell during endocytosis, we can gain a better understanding of how they respond to pH-regulated anti-viral therapies but also pH-induced changes in extracellular environments. This review provides a detailed explanation of the pH-dependent viral structural changes preceding and initiating viral disassembly during endocytosis for influenza A (IAV) and SARS coronaviruses. Drawing upon extensive literature from the last few decades and latest research, I analyze and compare the circumstances in which IAV and SARS-coronavirus can undertake endocytotic pathways that are pH-dependent. While there are similarities in the pH-regulated patterns leading to fusion, the mechanisms and pH activation differ. In terms of fusion activity, the measured activation pH values for IAV, across all subtypes and species, vary between approximately 5.0 to 6.0, while SARS-coronavirus necessitates a lower pH of 6.0 or less. The main difference between the pH-dependent endocytic pathways is that the SARS-coronavirus, unlike IAV, require the presence of specific pH-sensitive enzymes (cathepsin L) during endosomal transport. Conversely, the conformational changes in the IAV virus under acidic conditions in endosomes occur due to the specific envelope glycoprotein residues and envelope protein ion channels (viroporins) getting protonated by H+ ions. Despite extensive research over several decades, comprehending the pH-triggered conformational alterations of viruses still poses a significant challenge. The precise mechanisms of protonation mechanisms of certain during endosomal transport for both viruses remain incompletely understood. In absence of evidence, further research is needed.
Collapse
Affiliation(s)
- Amar Aganovic
- Faculty of Engineering Science and Technology, UiT The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
4
|
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: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
5
|
Antiviral Effects of ABMA and DABMA against Influenza Virus In Vitro and In Vivo via Regulating the Endolysosomal Pathway and Autophagy. Int J Mol Sci 2022; 23:ijms23073940. [PMID: 35409297 PMCID: PMC8999625 DOI: 10.3390/ijms23073940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/27/2022] [Accepted: 03/30/2022] [Indexed: 02/04/2023] Open
Abstract
Influenza virus is an acute and highly contagious respiratory pathogen that causes great concern to public health and for which there is a need for extensive drug discovery. The small chemical compound ABMA and its analog DABMA, containing an adamantane or a dimethyl-adamantane group, respectively, have been demonstrated to inhibit multiple toxins (diphtheria toxin, Clostridium difficile toxin B, Clostridium sordellii lethal toxin) and viruses (Ebola, rabies virus, HSV-2) by acting on the host’s vesicle trafficking. Here, we showed that ABMA and DABMA have antiviral effects against both amantadine-sensitive influenza virus subtypes (H1N1 and H3N2), amantadine-resistant subtypes (H3N2), and influenza B virus with EC50 values ranging from 2.83 to 7.36 µM (ABMA) and 1.82 to 6.73 µM (DABMA), respectively. ABMA and DABMA inhibited the replication of influenza virus genomic RNA and protein synthesis by interfering with the entry stage of the virus. Molecular docking evaluation together with activity against amantadine-resistant influenza virus strains suggested that ABMA and DABMA were not acting as M2 ion channel blockers. Subsequently, we found that early internalized H1N1 virions were retained in accumulated late endosome compartments after ABMA treatment. Additionally, ABMA disrupted the early stages of the H1N1 life cycle or viral RNA synthesis by interfering with autophagy. ABMA and DABMA protected mice from an intranasal H1N1 challenge with an improved survival rate of 67%. The present study suggests that ABMA and DABMA are potential antiviral leads for the development of a host-directed treatment against influenza virus infection.
Collapse
|
6
|
da Gama ANS, Soeiro MNC. Quinoline-based Compounds as Key Candidates to Tackle Drug Discovery Programs of Microbicidal Agents. Curr Pharm Des 2021; 27:1757-1762. [PMID: 33023440 DOI: 10.2174/1381612826666201006125644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/04/2020] [Accepted: 08/11/2020] [Indexed: 11/22/2022]
Abstract
Quinolines are heterocyclic nitrogen compounds, ubiquitous in nature and largely used as a structural component of dyes, solvent for resins, terpenes as well as during the production of several other chemical stuffs, including pesticides. Quinolines, such as quinine and chloroquine, exhibit various pharmacological properties, acting as antimalarial drugs, antiparasitic, antibacterial, antiviral, antifungal, and anticancer agents, besides being in clinical use for autoimmune diseases. A brief review has been presented regarding the biological effect and clinical use of quinolines and derivatives upon three trypanosomatids agents of important neglected tropical diseases; Trypanosoma cruzi, Trypanosoma brucei spp and Leishmania spp, which trigger Chagas disease, sleeping sickness and leishmaniasis, respectively, also extending to a glance update of their potential application towards other microbes relevant for emerging illness caused by fungi, bacteria and virus, including the pandemic Covid-19.
Collapse
Affiliation(s)
- Aline N Silva da Gama
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Maria N C Soeiro
- Laboratorio de Biologia Celular, Instituto Oswaldo Cruz, Fundacao Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Das A, Pandita D, Jain GK, Agarwal P, Grewal AS, Khar RK, Lather V. Role of phytoconstituents in the management of COVID-19. Chem Biol Interact 2021; 341:109449. [PMID: 33798507 PMCID: PMC8008820 DOI: 10.1016/j.cbi.2021.109449] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 03/07/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND COVID-19, a severe global pandemic caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has emerged as one of the most threatening transmissible disease. As a great threat to global public health, the development of treatment options has become vital, and a rush to find a cure has mobilized researchers globally from all areas. SCOPE AND APPROACH This review focuses on deciphering the potential of different secondary metabolites from medicinal plants as therapeutic options either as inhibitors of therapeutic targets of SARS-CoV-2 or as blockers of viral particles entry through host cell receptors. The use of medicinal plants containing specific phytomoieties could be seen in providing a safer and long-term solution for the population with lesser side effects. Key Findings and Conclusions: Considering the high cost and time-consuming drug discovery process, therapeutic repositioning of existing drugs was explored as treatment option in COVID-19, however several molecules have been retracted as therapeutics either due to no positive outcomes or the severe side effects. These effects call for exploring the alternate treatment options which are therapeutically effective as well as safe. Keeping this in mind, phytopharmaceuticals derived from medicinal plants could be explored as important resources in the development of COVID-19 treatment, as their role in the past for treatment of viral diseases like HIV, MERS-CoV, and influenza has been well reported. Considering this fact, different phytoconstituents such as flavonoids, alkaloids, tannins and glycosides etc. Possessing antiviral properties against coronaviruses and possessing potential against SARS-CoV-2 have been reviewed in the present work.
Collapse
Affiliation(s)
- Amiya Das
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313, India
| | - Deepti Pandita
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India.
| | - Gaurav Kumar Jain
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, 110017, India
| | - Pallavi Agarwal
- Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida, 201313, India
| | | | - Roop K Khar
- BS Anangpuria Institute of Pharmacy, Faridabad, Haryana, India
| | - Viney Lather
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Sector-125, Noida, 201313, India.
| |
Collapse
|
8
|
Affiliation(s)
- Miao Zhang
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xueting Yao
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Cheng Cui
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Xu Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| | - Haiyan Li
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
| | - Dongyang Liu
- Drug Clinical Trial Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Islamuddin M, Afzal O, Khan WH, Hisamuddin M, Altamimi ASA, Husain I, Kato K, Alamri MA, Parveen S. Inhibition of Chikungunya Virus Infection by 4-Hydroxy-1-Methyl-3-(3-morpholinopropanoyl)quinoline-2(1 H)-one (QVIR) Targeting nsP2 and E2 Proteins. ACS OMEGA 2021; 6:9791-9803. [PMID: 33869959 PMCID: PMC8047676 DOI: 10.1021/acsomega.1c00447] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/23/2021] [Indexed: 06/12/2023]
Abstract
The re-emergence of Chikungunya virus (CHIKV) infection in humans with no approved antiviral therapies or vaccines is one of the major problems with global significance. In the present investigation, we screened 80 in-house quinoline derivatives for their anti-CHIKV activity by computational techniques and found 4-hydroxy-1-methyl-3-(3-morpholinopropanoyl)quinoline-2(1H)-one (QVIR) to have potential binding affinities with CHIKV nsP2 and E2 glycoproteins. QVIR was evaluated in vitro for its anti-CHIKV potential. QVIR showed strong inhibition of CHIKV infection with an EC50 (50% effective concentration) value of 2.2 ± 0.49 μM without significant cytotoxicity (CC50 > 200 μM) and was chosen for further elucidation of its antiviral mechanism. The infectious viral particle formation was abolished by approximately 72% at a QVIR concentration of 20 μM during infection in the BHK-21 cell line, and the CHIKV RNA synthesis was diminished by 84% for nsP2 as well as 74% for E2, whereas the levels of viral proteins were decreased by 69.9% for nsP2 and 53.9% for E2. Flow cytometry analysis confirmed a huge decline in the expression of viral nsP2 and E2 proteins by 71.84 and 67.7%, respectively. Time of addition experiments indicated that QVIR inhibited viral infection at early and late stages of viral replication cycle, and the optimal inhibition was observed at 16 h post infection. The present study advocates for the first time that QVIR acts as a substantial and potent inhibitor against CHIKV and might be as an auspicious novel drug candidate for the development of therapeutic agents against CHIKV infections.
Collapse
Affiliation(s)
- Mohammad Islamuddin
- Molecular
Virology Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, New Delhi 110025, India
- Laboratory
of Sustainable Animal Environment, Graduate School of Agricultural
Science, Tohoku University, Osaki, Miyagi 989-6711, Japan
| | - Obaid Afzal
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Wajihul Hasan Khan
- Kusuma
School of Biological Sciences, Indian Institute
of Technology (IIT), New Delhi 110016, India
| | - Malik Hisamuddin
- Molecular
Virology Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | | | - Ibraheem Husain
- Department
of Pharmacology, School of Pharmaceutical and Research, Hamdard University, New Delhi 110062, India
| | - Kentaro Kato
- Laboratory
of Sustainable Animal Environment, Graduate School of Agricultural
Science, Tohoku University, Osaki, Miyagi 989-6711, Japan
| | - Mubarak A. Alamri
- Department
of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Shama Parveen
- Molecular
Virology Laboratory, Centre for Interdisciplinary Research in Basic
Sciences, Jamia Millia Islamia, New Delhi 110025, India
| |
Collapse
|
10
|
Chepur SV, Pluzhnikov NN, Chubar OV, Bakulina LS, Litvinenko IV, Makarov VA, Gogolevsky AS, Myasnikov VA, Myasnikova IA, Al-Shehadat RI. Respiratory RNA Viruses: How to Be Prepared for an Encounter with New Pandemic Virus Strains. BIOLOGY BULLETIN REVIEWS 2021; 11. [PMCID: PMC8078390 DOI: 10.1134/s207908642102002x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The characteristics of the biology of influenza viruses and coronavirus that determine the implementation of the infectious process are presented. With provision for pathogenesis of infection possible effects of serine proteinase inhibitors, heparin, and inhibitors of heparan sulfate receptors in the prevention of cell contamination by viruses are examined. It has been determined that chelators of metals of variable valency and antioxidants should be used for the reduction of replicative activity of viruses and anti-inflammatory therapy. The possibility of a pH-dependent impairment of glycosylation of cellular and viral proteins was traced for chloroquine and its derivatives. The use of low-toxicity drugs as part of adjunct therapy increases the effectiveness of synthetic antiviral drugs and interferons and ensures the safety of baseline therapy.
Collapse
Affiliation(s)
- S. V. Chepur
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - N. N. Pluzhnikov
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - O. V. Chubar
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - L. S. Bakulina
- Burdenko Voronezh State Medical University, 394036 Voronezh, Russia
| | | | - V. A. Makarov
- Fundamentals of Biotechnology Federal Research Center, 119071 Moscow, Russia
| | - A. S. Gogolevsky
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - V. A. Myasnikov
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - I. A. Myasnikova
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| | - R. I. Al-Shehadat
- State Scientific Research Test Institute of Military Medicine of the Ministry of Defense of the Russian Federation, 195043 St. Petersburg, Russia
| |
Collapse
|
11
|
Barzkar F, Ranjbar M, Sioofy-Khojine AB, Khajehazad M, Vesal Azad R, Moradi Y, Baradaran HR. Efficacy and safety of chloroquine and hydroxychloroquine for COVID-19: A comprehensive evidence synthesis of clinical, animal, and in vitro studies. Med J Islam Repub Iran 2020; 34:171. [PMID: 33816370 PMCID: PMC8004577 DOI: 10.47176/mjiri.34.171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Indexed: 12/24/2022] Open
Abstract
Background: The world is facing a pandemic of COVID-19, a respiratory disease caused by a novel coronavirus which is now called SARS-CoV-2. Current treatment recommendations for the infection are mainly repurposed drugs based on experience with other clinically similar conditions and are not backed by direct evidence. Chloroquine (CQ) and its derivative Hydroxychloroquine (HCQ) are among the candidates. We aimed to synthesize current evidence systematically for in vitro, animal, and human studies on the efficacy and safety of chloroquine in patients with COVID-19. Methods: The Cochrane Library, Google Scholar, PubMed (via Medline), Embase, Scopus, and Web of Science, MedRxiv, clinical trial registries including clinicaltrials.gov, ChiCTR (Chinese Clinical Trial Registry), IRCT (Iranian Registry of Clinical Trials), and the EU Clinical Trials Register. We used the Cochrane tool for risk of bias assessment in randomized studies, the ROBINS tool for non-randomized studies, and the GRADE methodology to summarize the evidence and certainty in effect estimates. Results: The initial database searching retrieved 24,752 studies. Of these, 15,435 abstracts were screened and 115 were selected for full-text review. Finally, 20 human studies, 3 animal studies, and 4 in vitro studies were included in this systematic review. The risk of bias within studies was unclear to high and the overall certainty in evidence-based on GRADES- was very low. HCQ may be effective in clinical improvement in a subset of patients with COVID-19. However, the frequency of adverse events was higher in patients taking HCQ compared to standard of care alone. In contrast, animal studies, did not report any adverse effects. Furthermore, clear benefit of the drug in the survival of the animals has been reported. Most in vitro studies indicated a high selectivity index for the drug and one study that used a human coronavirus reported blockage of virus replication. Conclusion: Current evidence background is limited to six poorly conducted clinical studies with inconsistent findings which fail to show significant efficacy for HCQ. Safety data is also limited but the drug may increase adverse outcomes. Routine use of the drug is not recommended based on limited efficacy and concerns about the drug safety especially in high-risk populations.
Collapse
Affiliation(s)
- Farzaneh Barzkar
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mitra Ranjbar
- Department of Infectious Diseases, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Amir-Babak Sioofy-Khojine
- Department of Virology, Faculty of Medicine and Life Sciences, PL 100, 33014 University of Tampere, Tampere, Finland
| | - Mohammadamin Khajehazad
- Department of Infectious Diseases, School of Medicine, Iran University of Medical Science, Tehran, Iran
| | - Roya Vesal Azad
- School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Yousef Moradi
- Social Determinants of Health Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Hamid Reza Baradaran
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
- Ageing Clinical & Experimental Research Team, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
12
|
Lei ZN, Wu ZX, Dong S, Yang DH, Zhang L, Ke Z, Zou C, Chen ZS. Chloroquine and hydroxychloroquine in the treatment of malaria and repurposing in treating COVID-19. Pharmacol Ther 2020; 216:107672. [PMID: 32910933 PMCID: PMC7476892 DOI: 10.1016/j.pharmthera.2020.107672] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/28/2020] [Accepted: 08/31/2020] [Indexed: 12/14/2022]
Abstract
Chloroquine (CQ) and Hydroxychloroquine (HCQ) have been commonly used for the treatment and prevention of malaria, and the treatment of autoimmune diseases for several decades. As their new mechanisms of actions are identified in recent years, CQ and HCQ have wider therapeutic applications, one of which is to treat viral infectious diseases. Since the pandemic of the coronavirus disease 2019 (COVID-19), CQ and HCQ have been subjected to a number of in vitro and in vivo tests, and their therapeutic prospects for COVID-19 have been proposed. In this article, the applications and mechanisms of action of CQ and HCQ in their conventional fields of anti-malaria and anti-rheumatism, as well as their repurposing prospects in anti-virus are reviewed. The current trials and future potential of CQ and HCQ in combating COVID-19 are discussed.
Collapse
Affiliation(s)
- Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Shaowei Dong
- Key Laboratory of medical electrophysiology of education ministry, School of Pharmacy, Southwest Medical University, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China
| | - Dong-Hua Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Litu Zhang
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, Guangdong, China.
| | - Chang Zou
- Key Laboratory of medical electrophysiology of education ministry, School of Pharmacy, Southwest Medical University, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Southern University of Science and Technology, Shenzhen 518020, Guangdong, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| |
Collapse
|
13
|
Makarov V, Riabova O, Ekins S, Pluzhnikov N, Chepur S. The past, present and future of RNA respiratory viruses: influenza and coronaviruses. Pathog Dis 2020; 78:ftaa046. [PMID: 32860686 PMCID: PMC7499567 DOI: 10.1093/femspd/ftaa046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022] Open
Abstract
Influenza virus and coronaviruses continue to cause pandemics across the globe. We now have a greater understanding of their functions. Unfortunately, the number of drugs in our armory to defend us against them is inadequate. This may require us to think about what mechanisms to address. Here, we review the biological properties of these viruses, their genetic evolution and antiviral therapies that can be used or have been attempted. We will describe several classes of drugs such as serine protease inhibitors, heparin, heparan sulfate receptor inhibitors, chelating agents, immunomodulators and many others. We also briefly describe some of the drug repurposing efforts that have taken place in an effort to rapidly identify molecules to treat patients with COVID-19. While we put a heavy emphasis on the past and present efforts, we also provide some thoughts about what we need to do to prepare for respiratory viral threats in the future.
Collapse
Affiliation(s)
- Vadim Makarov
- Federal Research Center Fundamentals of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow 119071, Russia
| | - Olga Riabova
- Federal Research Center Fundamentals of Biotechnology of the Russian Academy of Sciences, 33-2 Leninsky Prospect, Moscow 119071, Russia
| | - Sean Ekins
- Collaborations Pharmaceuticals, Inc., 840 Main Campus Drive, Lab 3510, Raleigh, NC 27606, USA
| | - Nikolay Pluzhnikov
- State Research Institute of Military Medicine of the Ministry of Defence of the Russian Federation, St Petersburg 195043, Russia
| | - Sergei Chepur
- State Research Institute of Military Medicine of the Ministry of Defence of the Russian Federation, St Petersburg 195043, Russia
| |
Collapse
|
14
|
Chen X, Geiger JD. Janus sword actions of chloroquine and hydroxychloroquine against COVID-19. Cell Signal 2020; 73:109706. [PMID: 32629149 PMCID: PMC7333634 DOI: 10.1016/j.cellsig.2020.109706] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 06/26/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Chloroquine (CQ) and its analogue hydroxychloroquine (HCQ) have been thrust into our everyday vernacular because some believe, based on very limited basic and clinical data, that they might be helpful in preventing and/or lessening the severity of the pandemic coronavirus disease 2019 (COVID-19). However, lacking is a temperance in enthusiasm for their possible use as well as sufficient perspective on their effects and side-effects. CQ and HCQ have well-known properties of being diprotic weak bases that preferentially accumulate in acidic organelles (endolysosomes and Golgi apparatus) and neutralize luminal pH of acidic organelles. These primary actions of CQ and HCQ are responsible for their anti-malarial effects; malaria parasites rely on acidic digestive vacuoles for survival. Similarly, de-acidification of endolysosomes and Golgi by CQ and HCQ may block severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) integration into host cells because SARS-CoV-2 may require an acidic environment for its entry and for its ability to bud and infect bystander cells. Further, de-acidification of endolysosomes and Golgi may underly the immunosuppressive effects of these two drugs. However, modern cell biology studies have shown clearly that de-acidification results in profound changes in the structure, function and cellular positioning of endolysosomes and Golgi, in signaling between these organelles and other subcellular organelles, and in fundamental cellular functions. Thus, studying the possible therapeutic effects of CQ and HCQ against COVID-19 must occur concurrent with studies of the extent to which these drugs affect organellar and cell biology. When comprehensively examined, a better understanding of the Janus sword actions of these and other drugs might yield better decisions and better outcomes.
Collapse
Affiliation(s)
- Xuesong Chen
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America.
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota, United States of America
| |
Collapse
|
15
|
Patil VM, Singhal S, Masand N. A systematic review on use of aminoquinolines for the therapeutic management of COVID-19: Efficacy, safety and clinical trials. Life Sci 2020; 254:117775. [PMID: 32418894 PMCID: PMC7211740 DOI: 10.1016/j.lfs.2020.117775] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023]
Abstract
Recent global outbreak of the pandemic caused by coronavirus (COVID-19) emphasizes the urgent need for novel antiviral therapeutics. It can be supplemented by utilization of efficient and validated drug discovery approaches such as drug repurposing/repositioning. The well reported and clinically used anti-malarial aminoquinoline drugs (chloroquine and hydroxychloroquine) have shown potential to be repurposed to control the present pandemic by inhibition of COVID-19. The review elaborates the mechanism of action, safety (side effects, adverse effects, toxicity) and details of clinical trials for chloroquine and hydroxychloroquine to benefit the clinicians, medicinal chemist, pharmacologist actively involved in controlling the pandemic and to provide therapeutics for the treatment of COVID-19 infection.
Collapse
Affiliation(s)
- Vaishali M Patil
- Computer Aided Drug Design Lab, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India.
| | - Shipra Singhal
- Computer Aided Drug Design Lab, KIET School of Pharmacy, KIET Group of Institutions, Delhi-NCR, Ghaziabad, India
| | - Neeraj Masand
- Department of Pharmacy, Lala Lajpat Rai Memorial Medical College, Meerut, Uttar Pradesh, India
| |
Collapse
|
16
|
Chen Y, Shen T, Zhong L, Liu Z, Dong X, Huang T, Wang Q, Xiao H. Research Progress of Chloroquine and Hydroxychloroquine on the COVID-19 and Their Potential Risks in Clinic Use. Front Pharmacol 2020; 11:1167. [PMID: 32848774 PMCID: PMC7412992 DOI: 10.3389/fphar.2020.01167] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/17/2020] [Indexed: 01/12/2023] Open
Abstract
In December 2019, a severe outbreak of a novel coronavirus (COVID-19) occurred in the whole world, posing a great threat to people's health. With the outbreak and development of the epidemic, how to improve the cure rate, find effective drugs against this virus, has been the most urgent problem. Chloroquine (CQ) was verified effective against COVID-19 in vitro. As CQ's analogue, hydroxychloroquine (HCQ) was also reminded as a potential candidate for treating COVID-19. This review summarizes the latest clinical trials of CQ and HCQ against COVID-19 and its therapeutic regimen in China aiming to share their current usage to the whole world and provide insight into its appropriate future use in the treatment of COVID-19. Through searching the CNKI and Wangfang databases in Chinese language and PubMed, EMBASE, and Ovid databases in English language to identify published reports with the keywords including "coronavirus/COVID, chloroquine, hyroxychloroquine" in alone or combined, we found out the potential preclinical or clinical evidence for using CQ and HCQ against COVID-19. Consequently, we also searched the website of Chinese Clinical Trial Registry (http://www.chictr.org.cn/) till the day on 27th, June, 2020. This review found that there are 23 programs aimed to treat the different phases under COVID-19 pipeline in clinic with CQ and HCQ, totally. The inclusion criteria, exclusion criteria and therapeutic regimen were all shared to consult. Among them, seven have been canceled due to lack of patients or other objective factors. There are two trials have completed, which the potential relationship between usage and adverse reactions was discussed emphatically. Through literature research, we suggested that paid close attention to retinal toxicity and ophthalmologic adverse symptom of CQ and HCQ. And the outcome of HCQ in clinic shows better than CQ especially in protective effect with low dosage.
Collapse
Affiliation(s)
- Yan Chen
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - TaiPeng Shen
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - LiJun Zhong
- Department of Information, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - ZhiXi Liu
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - XinWei Dong
- Department of Information, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - TingWenLi Huang
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - QiuJu Wang
- Department of Clinical Laboratory, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - HongTao Xiao
- Department of Pharmacy, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Chengdu, China
| |
Collapse
|
17
|
Nirk EL, Reggiori F, Mauthe M. Hydroxychloroquine in rheumatic autoimmune disorders and beyond. EMBO Mol Med 2020; 12:e12476. [PMID: 32715647 PMCID: PMC7411564 DOI: 10.15252/emmm.202012476] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/02/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Initially used as antimalarial drugs, hydroxychloroquine (HCQ) and, to a lesser extent, chloroquine (CQ) are currently being used to treat several diseases. Due to its cost‐effectiveness, safety and efficacy, HCQ is especially used in rheumatic autoimmune disorders (RADs), such as systemic lupus erythematosus, primary Sjögren's syndrome and rheumatoid arthritis. Despite this widespread use in the clinic, HCQ molecular modes of action are still not completely understood. By influencing several cellular pathways through different mechanisms, CQ and HCQ inhibit multiple endolysosomal functions, including autophagy, as well as endosomal Toll‐like receptor activation and calcium signalling. These effects alter several aspects of the immune system with the synergistic consequence of reducing pro‐inflammatory cytokine production and release, one of the most marked symptoms of RADs. Here, we review the current knowledge on the molecular modes of action of these drugs and the circumstances under which they trigger side effects. This is of particular importance as the therapeutic use of HCQ is expanding beyond the treatment of malaria and RADs.
Collapse
Affiliation(s)
- Eliise Laura Nirk
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Mario Mauthe
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
18
|
|
19
|
Kim AHJ, Sparks JA, Liew JW, Putman MS, Berenbaum F, Duarte-García A, Graef ER, Korsten P, Sattui SE, Sirotich E, Ugarte-Gil MF, Webb K, Grainger R. A Rush to Judgment? Rapid Reporting and Dissemination of Results and Its Consequences Regarding the Use of Hydroxychloroquine for COVID-19. Ann Intern Med 2020; 172:819-821. [PMID: 32227189 PMCID: PMC7138335 DOI: 10.7326/m20-1223] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hydroxychloroquine, an essential treatment for many patients with rheumatologic conditions, has recently garnered widespread attention as a potential treatment for COVID-19 infection. The authors appraise the study generating this interest and highlight the potential consequences of rapid dissemination of overinterpreted data, particularly for people with conditions for which hydroxychloroquine has demonstrated benefits in preventing organ damage and life-threatening disease flares.
Collapse
Affiliation(s)
- Alfred H J Kim
- Washington University School of Medicine, St. Louis, Missouri (A.H.K.)
| | - Jeffrey A Sparks
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (J.A.S.)
| | - Jean W Liew
- University of Washington, Seattle, Washington (J.W.L.)
| | | | - Francis Berenbaum
- Sorbonne University, Inserm CRSA, AP-HP Saint-Antoine Hospital, Paris, France (F.B.)
| | | | - Elizabeth R Graef
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts (E.R.G.)
| | - Peter Korsten
- University Medical Center Göttingen, Göttingen, Germany (P.K.)
| | | | - Emily Sirotich
- McMaster University, Hamilton, and Canadian Arthritis Patient Alliance, Toronto, Ontario, Canada (E.S.)
| | - Manuel F Ugarte-Gil
- School of Medicine, Universidad Científica del Sur and Hospital Nacional Guillermo Almenara Irigoyen, EsSalud, Lima, Peru (M.F.U.)
| | - Kate Webb
- University of Cape Town, Cape Town, South Africa, and the Francis Crick Institute, London, United Kingdom (K.W.)
| | | | | |
Collapse
|
20
|
Hashem AM, Alghamdi BS, Algaissi AA, Alshehri FS, Bukhari A, Alfaleh MA, Memish ZA. Therapeutic use of chloroquine and hydroxychloroquine in COVID-19 and other viral infections: A narrative review. Travel Med Infect Dis 2020; 35:101735. [PMID: 32387694 PMCID: PMC7202851 DOI: 10.1016/j.tmaid.2020.101735] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/17/2022]
Abstract
The rapidly spreading Coronavirus Disease (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus (SARS-CoV-2), represents an unprecedented serious challenge to the global public health community. The extremely rapid international spread of the disease with significant morbidity and mortality made finding possible therapeutic interventions a global priority. While approved specific antiviral drugs against SARS-CoV-2 are still lacking, a large number of existing drugs are being explored as a possible treatment for COVID-19 infected patients. Recent publications have re-examined the use of Chloroquine (CQ) and/or Hydroxychloroquine (HCQ) as a potential therapeutic option for these patients. In an attempt to explore the evidence that supports their use in COVID-19 patients, we comprehensively reviewed the previous studies which used CQ or HCQ as an antiviral treatment. Both CQ and HCQ demonstrated promising in vitro results, however, such data have not yet been translated into meaningful in vivo studies. While few clinical trials have suggested some beneficial effects of CQ and HCQ in COVID-19 patients, most of the reported data are still preliminary. Given the current uncertainty, it is worth being mindful of the potential risks and strictly rationalise the use of these drugs in COVID-19 patients until further high quality randomized clinical trials are available to clarify their role in the treatment or prevention of COVID-19.
Collapse
Affiliation(s)
- Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Vaccines and Immunnotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Badrah S Alghamdi
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia; Pre-Clinical Research Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Abdullah A Algaissi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia; Medical Research Center, Jazan University, Jazan, Saudi Arabia.
| | - Fahad S Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Abdullah Bukhari
- Department of Medicine, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Riyadh, Saudi Arabia.
| | - Mohamed A Alfaleh
- Vaccines and Immunnotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia; Faculty of Pharmacy; King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ziad A Memish
- King Saud Medical City, Research & Innovation Center, Ministry of Health, Saudi Arabia; Al-Faisal University, Riyadh, Saudi Arabia; Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA, USA.
| |
Collapse
|
21
|
Meyerowitz EA, Vannier AGL, Friesen MGN, Schoenfeld S, Gelfand JA, Callahan MV, Kim AY, Reeves PM, Poznansky MC. Rethinking the role of hydroxychloroquine in the treatment of COVID-19. FASEB J 2020; 34:6027-6037. [PMID: 32350928 PMCID: PMC7267640 DOI: 10.1096/fj.202000919] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/07/2023]
Abstract
There are currently no proven or approved treatments for coronavirus disease 2019 (COVID-19). Early anecdotal reports and limited in vitro data led to the significant uptake of hydroxychloroquine (HCQ), and to lesser extent chloroquine (CQ), for many patients with this disease. As an increasing number of patients with COVID-19 are treated with these agents and more evidence accumulates, there continues to be no high-quality clinical data showing a clear benefit of these agents for this disease. Moreover, these agents have the potential to cause harm, including a broad range of adverse events including serious cardiac side effects when combined with other agents. In addition, the known and potent immunomodulatory effects of these agents which support their use in the treatment of auto-immune conditions, and provided a component in the original rationale for their use in patients with COVID-19, may, in fact, undermine their utility in the context of the treatment of this respiratory viral infection. Specifically, the impact of HCQ on cytokine production and suppression of antigen presentation may have immunologic consequences that hamper innate and adaptive antiviral immune responses for patients with COVID-19. Similarly, the reported in vitro inhibition of viral proliferation is largely derived from the blockade of viral fusion that initiates infection rather than the direct inhibition of viral replication as seen with nucleoside/tide analogs in other viral infections. Given these facts and the growing uncertainty about these agents for the treatment of COVID-19, it is clear that at the very least thoughtful planning and data collection from randomized clinical trials are needed to understand what if any role these agents may have in this disease. In this article, we review the datasets that support or detract from the use of these agents for the treatment of COVID-19 and render a data informed opinion that they should only be used with caution and in the context of carefully thought out clinical trials, or on a case-by-case basis after rigorous consideration of the risks and benefits of this therapeutic approach.
Collapse
Affiliation(s)
- Eric A. Meyerowitz
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Augustin G. L. Vannier
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Morgan G. N. Friesen
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Sara Schoenfeld
- Division of Allergy, Immunology and RheumatologyMGH and HMSBostonMAUSA
| | - Jeffrey A. Gelfand
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Michael V. Callahan
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
- Special Advisor to the Assistant Secretary of Public Health Preparedness and Response U.S Dept of Health and Human ServicesWashingtonDCUSA
| | - Arthur Y. Kim
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
| | - Patrick M. Reeves
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| | - Mark C. Poznansky
- Division of Infectious DiseasesMassachusetts General Hospital (MGH) and Harvard Medical School (HMS)BostonMAUSA
- Vaccine and Immunotherapy Center (VIC)MGH and HMSBostonMAUSA
| |
Collapse
|
22
|
Audi A, Soudani N, Dbaibo G, Zaraket H. Depletion of Host and Viral Sphingomyelin Impairs Influenza Virus Infection. Front Microbiol 2020; 11:612. [PMID: 32425895 PMCID: PMC7203554 DOI: 10.3389/fmicb.2020.00612] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/19/2020] [Indexed: 12/19/2022] Open
Abstract
Influenza A virus (IAV) is a major human respiratory pathogen causing annual epidemics as well as periodic pandemics. A complete understanding of the virus pathogenesis and host factors involved in the viral lifecycle is crucial for developing novel therapeutic approaches. Sphingomyelin (SM) is the most abundant membrane sphingolipid. It preferentially associates with cholesterol to form distinct domains named lipid rafts. Sphingomyelinases, including acid sphingomyelinase (ASMase), catalyzes the hydrolysis of membrane SM and consequently transform lipid rafts into ceramide-enriched membrane platforms. In this study, we investigated the effect of SM hydrolysis on IAV propagation. Depleting plasma membrane SM by exogenous bacterial SMase (bSMase) impaired virus infection and reduced virus entry, whereas exogenous SM enhanced infection. Moreover, the depletion of virus envelope SM also reduced virus infectivity and impaired its attachment and internalization. Nonetheless, inhibition of ASMase by desipramine did not affect IAV infection. Similarly, virus replication was not impaired in Niemann-Pick disease type A (NPA) cells, which lack functional ASMase. IAV infection in A549 cells was associated with suppression of ASMase activity starting at 6 h post-infection. Our data reveals that intact cellular and viral envelope SM is required for efficient IAV infection. Therefore, SM metabolism can be a potential target for therapeutic intervention against influenza virus infection.
Collapse
Affiliation(s)
- Amani Audi
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadia Soudani
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Doctoral School of Science and Technology, Research Platform for Environmental Science (PRASE), Faculty of Sciences, Lebanese University, Beirut, Lebanon
| | - Ghassan Dbaibo
- Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Department of Pediatrics and Adolescent Medicine, Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon.,Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Hassan Zaraket
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.,Center for Infectious Diseases Research, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
23
|
Chloroquine inhibits endosomal viral RNA release and autophagy-dependent viral replication and effectively prevents maternal to fetal transmission of Zika virus. Antiviral Res 2019; 169:104547. [PMID: 31251958 DOI: 10.1016/j.antiviral.2019.104547] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/25/2019] [Accepted: 06/25/2019] [Indexed: 01/15/2023]
Abstract
Zika virus (ZIKV) infection can cause neonatal microcephaly and neurological disorders. Currently, there is no designated drug for treating ZIKV infection and preventing neonatal microcephaly. In this study, we evaluated the effect of chloroquine, an anti-malaria drug, in ZIKV infected cells and mouse models. Chloroquine significantly inhibited ZIKV infection in multiple mammalian cell lines. Chloroquine treatment significantly improved the survival of ZIKV-infected 1-day old suckling SCID Beige mice and reduced viremia in adult SCID Beige mice. Importantly, chloroquine protected the fetus from maternal infection by reducing placenta to fetus viral transmission. We found that chloroquine exerts at least two mechanisms in protecting against ZIKV infection: 1) inhibiting endosomal disassembly of the internalized virus and thus reducing the release of viral RNA to the cytoplasm for replication; 2) inhibiting ZIKV RNA replication through blocking ZIKV induced autophagy. Our study suggests that chloroquine treatment warrants to be considered as a mitigation strategy for treating ZIKV infection and preventing ZIKV-associated microcephaly in pregnant women.
Collapse
|
24
|
García-Serradilla M, Risco C, Pacheco B. Drug repurposing for new, efficient, broad spectrum antivirals. Virus Res 2019; 264:22-31. [PMID: 30794895 PMCID: PMC7114681 DOI: 10.1016/j.virusres.2019.02.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 12/26/2022]
Abstract
Emerging viruses are a major threat to human health. Recent outbreaks have emphasized the urgent need for new antiviral treatments. For several pathogenic viruses, considerable efforts have focused on vaccine development. However, during epidemics infected individuals need to be treated urgently. High-throughput screening of clinically tested compounds provides a rapid means to identify undiscovered, antiviral functions for well-characterized therapeutics. Repurposed drugs can bypass part of the early cost and time needed for validation and authorization. In this review we describe recent efforts to find broad spectrum antivirals through drug repurposing. We have chosen several candidates and propose strategies to understand their mechanism of action and to determine how resistance to antivirals develops in infected cells.
Collapse
Affiliation(s)
- Moisés García-Serradilla
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain
| | - Cristina Risco
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain.
| | - Beatriz Pacheco
- Cell Structure Laboratory, National Center for Biotechnology, National Research Council, CNB-CSIC, Darwin 3, UAM, campus de Cantoblanco, 28049 Madrid, Spain.
| |
Collapse
|
25
|
de la Guardia C, Stephens DE, Dang HT, Quijada M, Larionov OV, Lleonart R. Antiviral Activity of Novel Quinoline Derivatives against Dengue Virus Serotype 2. Molecules 2018; 23:molecules23030672. [PMID: 29547522 PMCID: PMC5997395 DOI: 10.3390/molecules23030672] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Revised: 03/13/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022] Open
Abstract
Dengue virus causes dengue fever, a debilitating disease with an increasing incidence in many tropical and subtropical territories. So far, there are no effective antivirals licensed to treat this virus. Here we describe the synthesis and antiviral activity evaluation of two compounds based on the quinoline scaffold, which has shown potential for the development of molecules with various biological activities. Two of the tested compounds showed dose-dependent inhibition of dengue virus serotype 2 in the low and sub micromolar range. The compounds 1 and 2 were also able to impair the accumulation of the viral envelope glycoprotein in infected cells, while showing no sign of direct virucidal activity and acting possibly through a mechanism involving the early stages of the infection. The results are congruent with previously reported data showing the potential of quinoline derivatives as a promising scaffold for the development of new antivirals against this important virus.
Collapse
Affiliation(s)
- Carolina de la Guardia
- Institute of Scientific Research and High Technology Services (INDICASAT AIP), PO 0843-01103 City of Panama, Panama.
- Department of Biotechnology, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur 522510, Andhra Pradesh, India.
| | - David E Stephens
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Hang T Dang
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Mario Quijada
- Institute of Scientific Research and High Technology Services (INDICASAT AIP), PO 0843-01103 City of Panama, Panama.
| | - Oleg V Larionov
- Department of Chemistry, University of Texas at San Antonio, San Antonio, TX 78249, USA.
| | - Ricardo Lleonart
- Institute of Scientific Research and High Technology Services (INDICASAT AIP), PO 0843-01103 City of Panama, Panama.
| |
Collapse
|
26
|
Villeret B, Dieu A, Straube M, Solhonne B, Miklavc P, Hamadi S, Le Borgne R, Mailleux A, Norel X, Aerts J, Diallo D, Rouzet F, Dietl P, Sallenave JM, Garcia-Verdugo I. Silver Nanoparticles Impair Retinoic Acid-Inducible Gene I-Mediated Mitochondrial Antiviral Immunity by Blocking the Autophagic Flux in Lung Epithelial Cells. ACS NANO 2018; 12:1188-1202. [PMID: 29357226 DOI: 10.1021/acsnano.7b06934] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Silver nanoparticles (AgNPs) are microbicidal agents which could be potentially used as an alternative to antivirals to treat human infectious diseases, especially influenza virus infections where antivirals have generally proven unsuccessful. However, concerns about the use of AgNPs on humans arise from their potential toxicity, although mechanisms are not well-understood. We show here, in the context of an influenza virus infection of lung epithelial cells, that AgNPs down-regulated influenza induced CCL-5 and -IFN-β release (two cytokines important in antiviral immunity) through RIG-I inhibition, while enhancing IL-8 production, a cytokine important for mobilizing host antibacterial responses. AgNPs activity was independent of coating and was not observed with gold nanoparticles. Down-stream analysis indicated that AgNPs disorganized the mitochondrial network and prevented the antiviral IRF-7 transcription factor influx into the nucleus. Importantly, we showed that the modulation of RIG-I-IRF-7 pathway was concomitant with inhibition of either classical or alternative autophagy (ATG-5- and Rab-9 dependent, respectively), depending on the epithelial cell type used. Altogether, this demonstration of a AgNPs-mediated functional dichotomy (down-regulation of IFN-dependent antiviral responses and up-regulation of IL-8-dependent antibacterial responses) may have practical implications for their use in the clinic.
Collapse
Affiliation(s)
- Berengere Villeret
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Alexandra Dieu
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Marjolene Straube
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Brigitte Solhonne
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Pika Miklavc
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford , Salford, United Kingdom
| | - Sena Hamadi
- Université Paris Est, ICMPE (UMR7182), CNRS, UPEC , F-94320 Thiais, France
| | - Rémi Le Borgne
- ImagoSeine, Electron Microscopy Facility, Institut Jacques Monod, CNRS UMR 7592, Université Paris Diderot , Sorbonne Paris Cité, 75205 Cedex 13 Paris, France
| | - Arnaud Mailleux
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Xavier Norel
- Inserm U1148, UMR-S1148, University Paris Nord , 75018 Paris, France
| | - Joel Aerts
- AP-HP, Groupe Hospitalier Bichat-Claude Bernard, Service de Médecine Nucléaire, Université Denis Diderot-Paris 7, U1148, Inserm , 75013 Paris, France
| | - Devy Diallo
- AP-HP, Groupe Hospitalier Bichat-Claude Bernard, Service de Médecine Nucléaire, Université Denis Diderot-Paris 7, U1148, Inserm , 75013 Paris, France
| | - Francois Rouzet
- AP-HP, Groupe Hospitalier Bichat-Claude Bernard, Service de Médecine Nucléaire, Université Denis Diderot-Paris 7, U1148, Inserm , 75013 Paris, France
| | - Paul Dietl
- Institute of General Physiology, University of Ulm , Albert-Einstein Allee 11, 89081 Ulm, Germany
| | - Jean-Michel Sallenave
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| | - Ignacio Garcia-Verdugo
- INSERM, UMR U1152, Laboratoire d'Excellence Inflamex, Département Hospitalo-Universitaire FIRE (Fibrosis, Inflammation, and Remodeling), University Paris Diderot , Sorbonne Paris Cité, 75018 Paris, France
| |
Collapse
|
27
|
Hui DS, Lee N, Chan PK, Beigel JH. The role of adjuvant immunomodulatory agents for treatment of severe influenza. Antiviral Res 2018; 150:202-216. [PMID: 29325970 PMCID: PMC5801167 DOI: 10.1016/j.antiviral.2018.01.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Revised: 12/31/2017] [Accepted: 01/05/2018] [Indexed: 12/18/2022]
Abstract
A severe inflammatory immune response with hypercytokinemia occurs in patients hospitalized with severe influenza, such as avian influenza A(H5N1), A(H7N9), and seasonal A(H1N1)pdm09 virus infections. The role of immunomodulatory therapy is unclear as there have been limited published data based on randomized controlled trials (RCTs). Passive immunotherapy such as convalescent plasma and hyperimmune globulin have some studies demonstrating benefit when administered as an adjunctive therapy for severe influenza. Triple combination of oseltamivir, clarithromycin, and naproxen for severe influenza has one study supporting its use, and confirmatory studies would be of great interest. Likewise, confirmatory studies of sirolimus without concomitant corticosteroid therapy should be explored as a research priority. Other agents with potential immunomodulating effects, including non-immune intravenous immunoglobulin, N-acetylcysteine, acute use of statins, macrolides, pamidronate, nitazoxanide, chloroquine, antiC5a antibody, interferons, human mesenchymal stromal cells, mycophenolic acid, peroxisome proliferator-activated receptors agonists, non-steroidal anti-inflammatory agents, mesalazine, herbal medicine, and the role of plasmapheresis and hemoperfusion as rescue therapy have supportive preclinical or observational clinical data, and deserve more investigation preferably by RCTs. Systemic corticosteroids administered in high dose may increase the risk of mortality and morbidity in patients with severe influenza and should not be used, while the clinical utility of low dose systemic corticosteroids requires further investigation.
Collapse
Affiliation(s)
- David S Hui
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong.
| | - Nelson Lee
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong; Division of Infectious Diseases, University of Alberta, Edmonton, Canada
| | - Paul K Chan
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong
| | - John H Beigel
- Leidos Biomedical Research Inc, Support to National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
28
|
Dyall J, Gross R, Kindrachuk J, Johnson RF, Olinger GG, Hensley LE, Frieman MB, Jahrling PB. Middle East Respiratory Syndrome and Severe Acute Respiratory Syndrome: Current Therapeutic Options and Potential Targets for Novel Therapies. Drugs 2017; 77:1935-1966. [PMID: 29143192 PMCID: PMC5733787 DOI: 10.1007/s40265-017-0830-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
No specific antivirals are currently available for two emerging infectious diseases, Middle East respiratory syndrome (MERS) and severe acute respiratory syndrome (SARS). A literature search was performed covering pathogenesis, clinical features and therapeutics, clinically developed drugs for repurposing and novel drug targets. This review presents current knowledge on the epidemiology, pathogenesis and clinical features of the SARS and MERS coronaviruses. The rationale for and outcomes with treatments used for SARS and MERS is discussed. The main focus of the review is on drug development and the potential that drugs approved for other indications provide for repurposing. The drugs we discuss belong to a wide range of different drug classes, such as cancer therapeutics, antipsychotics, and antimalarials. In addition to their activity against MERS and SARS coronaviruses, many of these approved drugs have broad-spectrum potential and have already been in clinical use for treating other viral infections. A wealth of knowledge is available for these drugs. However, the information in this review is not meant to guide clinical decisions, and any therapeutic described here should only be used in context of a clinical trial. Potential targets for novel antivirals and antibodies are discussed as well as lessons learned from treatment development for other RNA viruses. The article concludes with a discussion of the gaps in our knowledge and areas for future research on emerging coronaviruses.
Collapse
Affiliation(s)
- Julie Dyall
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA.
| | - Robin Gross
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Jason Kindrachuk
- Department of Medical Microbiology, University of Manitoba, Winnipeg, MN, Canada
| | - Reed F Johnson
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | | | - Lisa E Hensley
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Matthew B Frieman
- Department of Microbiology and Immunology, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Peter B Jahrling
- Integrated Research Facility, Division of Clinical Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
- Emerging Viral Pathogens Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| |
Collapse
|
29
|
Hu Y, Sneyd H, Dekant R, Wang J. Influenza A Virus Nucleoprotein: A Highly Conserved Multi-Functional Viral Protein as a Hot Antiviral Drug Target. Curr Top Med Chem 2017; 17:2271-2285. [PMID: 28240183 DOI: 10.2174/1568026617666170224122508] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2015] [Revised: 07/13/2016] [Accepted: 07/16/2016] [Indexed: 01/25/2023]
Abstract
Prevention and treatment of influenza virus infection is an ongoing unmet medical need. Each year, thousands of deaths and millions of hospitalizations are attributed to influenza virus infection, which poses a tremendous health and economic burden to the society. Aside from the annual influenza season, influenza viruses also lead to occasional influenza pandemics as a result of emerging or re-emerging influenza strains. Influenza viruses are RNA viruses that exist in quasispecies, meaning that they have a very diverse genetic background. Such a feature creates a grand challenge in devising therapeutic intervention strategies to inhibit influenza virus replication, as a single agent might not be able to inhibit all influenza virus strains. Both classes of currently approved anti-influenza drugs have limitations: the M2 channel blockers amantadine and rimantadine are no longer recommended for use in the U.S. due to predominant drug resistance, and resistance to the neuraminidase inhibitor oseltamivir is continuously on the rise. In pursuing the next generation of antiviral drugs with broad-spectrum activity and higher genetic barrier of drug resistance, the influenza virus nucleoprotein (NP) stands out as a high-profile drug target. This review summarizes recent developments in designing inhibitors targeting influenza NP and their mechanisms of action.
Collapse
Affiliation(s)
- Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ, United States
| | - Hannah Sneyd
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ, United States
| | - Raphael Dekant
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, the University of Arizona, Tucson, AZ, United States
| |
Collapse
|
30
|
Wu X, Wu X, Sun Q, Zhang C, Yang S, Li L, Jia Z. Progress of small molecular inhibitors in the development of anti-influenza virus agents. Am J Cancer Res 2017; 7:826-845. [PMID: 28382157 PMCID: PMC5381247 DOI: 10.7150/thno.17071] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/18/2016] [Indexed: 02/05/2023] Open
Abstract
The influenza pandemic is a major threat to human health, and highly aggressive strains such as H1N1, H5N1 and H7N9 have emphasized the need for therapeutic strategies to combat these pathogens. Influenza anti-viral agents, especially active small molecular inhibitors play important roles in controlling pandemics while vaccines are developed. Currently, only a few drugs, which function as influenza neuraminidase (NA) inhibitors and M2 ion channel protein inhibitors, are approved in clinical. However, the acquired resistance against current anti-influenza drugs and the emerging mutations of influenza virus itself remain the major challenging unmet medical needs for influenza treatment. It is highly desirable to identify novel anti-influenza agents. This paper reviews the progress of small molecular inhibitors act as antiviral agents, which include hemagglutinin (HA) inhibitors, RNA-dependent RNA polymerase (RdRp) inhibitors, NA inhibitors and M2 ion channel protein inhibitors etc. Moreover, we also summarize new, recently reported potential targets and discuss strategies for the development of new anti-influenza virus drugs.
Collapse
|
31
|
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: 241] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
32
|
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: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
33
|
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: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar 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
|
34
|
Wu L, Dai J, Zhao X, Chen Y, Wang G, Li K. Chloroquine enhances replication of influenza A virus A/WSN/33 (H1N1) in dose-, time-, and MOI-dependent manners in human lung epithelial cells A549. J Med Virol 2015; 87:1096-103. [PMID: 25715935 DOI: 10.1002/jmv.24135] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2014] [Indexed: 02/05/2023]
Abstract
Anti-malaria drug, chloroquine, has been reported to be effective against influenza A virus (IAV) in vitro and used in in-vivo experiments and clinical trial for prevention or treatment of influenza. In this study, it has been shown by immunofluorescence, hemagglutination, and plaque assays that chloroquine enhanced A/WSN/33 (H1N1) replication with pronounced cytopathic effect in dose-, time-, and MOI-dependent manners in human lung epithelial cells A549. Time-of-addition assay showed that inhibitory effect on virus replication by chloroquine pre-treatment was indistinctive, and virus productions were enhanced when the drug was applied after viral adsorption. The effectiveness of chloroquine as an anti-influenza drug is questioned, and caution in its use is recommended.
Collapse
Affiliation(s)
- Liqi Wu
- Department of Microbiology and Immunology, Shantou University Medical College, Shantou, China; The Key Immunopathology Laboratory of Guangdong Province, Shantou University Medical College, Shantou, China
| | | | | | | | | | | |
Collapse
|
35
|
Dalhoff A. Antiviral, antifungal, and antiparasitic activities of fluoroquinolones optimized for treatment of bacterial infections: a puzzling paradox or a logical consequence of their mode of action? Eur J Clin Microbiol Infect Dis 2015; 34:661-8. [PMID: 25515946 PMCID: PMC7087824 DOI: 10.1007/s10096-014-2296-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/02/2014] [Indexed: 12/19/2022]
Abstract
This review summarizes evidence that commercially available fluoroquinolones used for the treatment of bacterial infections are active against other non-bacterial infectious agents as well. Any of these fluoroquinolones exerts, in parallel to its antibacterial action, antiviral, antifungal, and antiparasitic actions at clinically achievable concentrations. This broad range of anti-infective activities is due to one common mode of action, i.e., the inhibition of type II topoisomerases or inhibition of viral helicases, thus maintaining the selective toxicity of fluoroquinolones inhibiting microbial topoisomerases at low concentrations but mammalian topoisomerases at much higher concentrations. Evidence suggests that standard doses of the fluoroquinolones studied are clinically effective against viral and parasitic infections, whereas higher doses administered topically were active against Candida spp. causing ophthalmological infections. Well-designed clinical studies should be performed to substantiate these findings.
Collapse
Affiliation(s)
- A Dalhoff
- Institute for Infection Medicine, University Medical Center Schleswig-Holstein, Brunswiker Str. 4, 24105, Kiel, Germany,
| |
Collapse
|
36
|
Ferraris O, Moroso M, Pernet O, Emonet S, Ferrier Rembert A, Paranhos-Baccalà G, Peyrefitte CN. Evaluation of Crimean-Congo hemorrhagic fever virus in vitro inhibition by chloroquine and chlorpromazine, two FDA approved molecules. Antiviral Res 2015; 118:75-81. [PMID: 25796972 PMCID: PMC7113845 DOI: 10.1016/j.antiviral.2015.03.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 03/12/2015] [Accepted: 03/13/2015] [Indexed: 12/20/2022]
Abstract
We analyzed the anti-CCHFV potential of the chloroquine and chlorpromazine molecules. They inhibited CCHFV in Vero and Huh7 cells: selectivity index ranging from 3 to 35. We analyzed the anti-CCHV potential of the combinations. Combinations of ribavirin and chloroquine or chlorpromazine were synergistic.
Crimean-Congo hemorrhagic virus (CCHFV) causes hemorrhagic fever with high case mortality rates and is endemic in south-eastern Europe, Africa, and Asia. The limited catalog of specific treatment, highlight the necessity to look for additional therapeutic solutions. Previous experiments suggested that CCHFV enters the cells via a clathrin dependent pathway. Therefore, we have evaluated the potential anti-CCHFV activity of several molecules targeting this entry possibility. We identified two molecules chloroquine and chlorpromazine. Neutralization and virus yield reduction assays were tested in Vero E6 and Huh7 cells on two different CCHFV strains. Several combinations, including ribavirin, were assayed to test a potential synergistic effect. The two molecules inhibited CCHFV, and depending on the virus and the cell lines, the 50% inhibitory concentration (IC50) values for chloroquine and chlorpromazine ranged from 28 to 43 and 10.8–15.7 μM, respectively. Time-of-addition studies demonstrated that these molecules had a direct effect on CCHFV infectivity and spread. The antiviral activity of the two molecules was still effective even when added up to 6 h post-infection and up to 24 h. The selectivity index ranging from 3 to 35 lead us to evaluate combinations with ribavirin. Combinations of ribavirin and chloroquine or chlorpromazine were synergistic against CCHFV. Though the low chlorpromazine selectivity index suggests the need for a chemical improvement, our present study highlights chloroquine as the main drug having the potential for drug repurposing.
Collapse
Affiliation(s)
- O Ferraris
- Institut de Recherche Biomédicale des Armées, Unité de Virologie, Lyon, France
| | - M Moroso
- Fondation Mérieux, Laboratoire des Pathogènes Émergents, Lyon, France
| | - O Pernet
- Unité de Virologie Humaine - INSERM U758, Lyon, France
| | - S Emonet
- Institut de Recherche Biomédicale des Armées, Unité de Virologie, Lyon, France
| | - A Ferrier Rembert
- Institut de Recherche Biomédicale des Armées, Unité de Virologie, Lyon, France
| | | | - C N Peyrefitte
- Institut de Recherche Biomédicale des Armées, Unité de Virologie, Lyon, France; Fondation Mérieux, Laboratoire des Pathogènes Émergents, Lyon, France
| |
Collapse
|
37
|
GASPARINI R, AMICIZIA D, LAI P, BRAGAZZI N, PANATTO D. Compounds with anti-influenza activity: present and future of strategies for the optimal treatment and management of influenza. Part II: Future compounds against influenza virus. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2014; 55:109-29. [PMID: 26137785 PMCID: PMC4718316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
In the first part of this overview, we described the life cycle of the influenza virus and the pharmacological action of the currently available drugs. This second part provides an overview of the molecular mechanisms and targets of still-experimental drugs for the treatment and management of influenza. Briefly, we can distinguish between compounds with anti-influenza activity that target influenza virus proteins or genes, and molecules that target host components that are essential for viral replication and propagation. These latter compounds have been developed quite recently. Among the first group, we will focus especially on hemagglutinin, M2 channel and neuraminidase inhibitors. The second group of compounds may pave the way for personalized treatment and influenza management. Combination therapies are also discussed. In recent decades, few antiviral molecules against influenza virus infections have been available; this has conditioned their use during human and animal outbreaks. Indeed, during seasonal and pandemic outbreaks, antiviral drugs have usually been administered in mono-therapy and, sometimes, in an uncontrolled manner to farm animals. This has led to the emergence of viral strains displaying resistance, especially to compounds of the amantadane family. For this reason, it is particularly important to develop new antiviral drugs against influenza viruses. Indeed, although vaccination is the most powerful means of mitigating the effects of influenza epidemics, antiviral drugs can be very useful, particularly in delaying the spread of new pandemic viruses, thereby enabling manufacturers to prepare large quantities of pandemic vaccine. In addition, antiviral drugs are particularly valuable in complicated cases of influenza, especially in hospitalized patients. To write this overview, we mined various databases, including Embase, PubChem, DrugBank and Chemical Abstracts Service, and patent repositories.
Collapse
Affiliation(s)
- R. GASPARINI
- Correspondence: R. Gasparini, Department of Health Sciences of Genoa University, via Pastore 1, 16132 Genoa, Italy - E-mail:
| | | | | | | | | |
Collapse
|
38
|
Borges MC, Castro LA, Fonseca BALD. Chloroquine use improves dengue-related symptoms. Mem Inst Oswaldo Cruz 2014; 108:596-9. [PMID: 23903975 DOI: 10.1590/s0074-02762013000500010] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/06/2013] [Indexed: 01/27/2023] Open
Abstract
Dengue is the most important arboviral disease in the world. As chloroquine, an antimalarial agent, has shown some antiviral effects, this study evaluated its effect in patients with dengue. A randomised, double-blind study was performed by administering chloroquine or placebo for three days to 129 patients with dengue-related symptoms. Of these patients, 37 were confirmed as having dengue and completed the study; in total, 19 dengue patients received chloroquine and 18 received placebo. There was no significant difference in the duration of the disease or the degree and days of fever. However, 12 patients (63%) with confirmed dengue reported a substantial decrease in pain intensity and a great improvement in their ability to perform daily activities (p = 0.0004) while on the medication and the symptoms returned immediately after these patients stopped taking the medication. The same effect was not observed in patients with diseases other than dengue. Therefore, this study shows that patients with dengue treated with chloroquine had an improvement in their quality of life and were able to resume their daily activities. However, as chloroquine did not alter the duration of the disease or the intensity and days of fever, further studies are necessary to confirm the clinical effects and to assess the side effects of chloroquine in dengue patients.
Collapse
Affiliation(s)
- Marcos Carvalho Borges
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | | | | |
Collapse
|
39
|
Marois I, Cloutier A, Meunier I, Weingartl HM, Cantin AM, Richter MV. Inhibition of influenza virus replication by targeting broad host cell pathways. PLoS One 2014; 9:e110631. [PMID: 25333287 PMCID: PMC4204995 DOI: 10.1371/journal.pone.0110631] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/24/2014] [Indexed: 12/19/2022] Open
Abstract
Antivirals that are currently used to treat influenza virus infections target components of the virus which can mutate rapidly. Consequently, there has been an increase in the number of resistant strains to one or many antivirals in recent years. Here we compared the antiviral effects of lysosomotropic alkalinizing agents (LAAs) and calcium modulators (CMs), which interfere with crucial events in the influenza virus replication cycle, against avian, swine, and human viruses of different subtypes in MDCK cells. We observed that treatment with LAAs, CMs, or a combination of both, significantly inhibited viral replication. Moreover, the drugs were effective even when they were administered 8 h after infection. Finally, analysis of the expression of viral acidic polymerase (PA) revealed that both drugs classes interfered with early events in the viral replication cycle. This study demonstrates that targeting broad host cellular pathways can be an efficient strategy to inhibit influenza replication. Furthermore, it provides an interesting avenue for drug development where resistance by the virus might be reduced since the virus is not targeted directly.
Collapse
Affiliation(s)
- Isabelle Marois
- Department of Medicine, Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Alexandre Cloutier
- Department of Medicine, Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Isabelle Meunier
- Department of Medicine, Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Hana M. Weingartl
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Manitoba, Canada, and Department of Medical Microbiology, College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - André M. Cantin
- Department of Medicine, Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
| | - Martin V. Richter
- Department of Medicine, Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Centre de Recherche du CHUS, Sherbrooke, Québec, Canada
- * E-mail:
| |
Collapse
|
40
|
Si Y, Li J, Niu Y, Liu X, Ren L, Guo L, Cheng M, Zhou H, Wang J, Jin Q, Yang W. Entry properties and entry inhibitors of a human H7N9 influenza virus. PLoS One 2014; 9:e107235. [PMID: 25222852 PMCID: PMC4164620 DOI: 10.1371/journal.pone.0107235] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2014] [Accepted: 08/08/2014] [Indexed: 11/18/2022] Open
Abstract
The recently identified human infections with a novel avian influenza H7N9 virus in China raise important questions regarding possible risk to humans. However, the entry properties and tropism of this H7N9 virus were poorly understood. Moreover, neuraminidase inhibitor resistant H7N9 isolates were recently observed in two patients and correlated with poor clinical outcomes. In this study, we aimed to elucidate the entry properties of H7N9 virus, design and evaluate inhibitors for H7N9 virus entry. We optimized and developed an H7N9-pseudotyped particle system (H7N9pp) that could be neutralized by anti-H7 antibodies and closely mimicked the entry process of the H7N9 virus. Avian, human and mouse-derived cultured cells showed high, moderate and low permissiveness to H7N9pp, respectively. Based on influenza virus membrane fusion mechanisms, a potent anti-H7N9 peptide (P155-185-chol) corresponding to the C-terminal ectodomain of the H7N9 hemagglutinin protein was successfully identified. P155-185-chol demonstrated H7N9pp-specific inhibition of infection with IC50 of 0.19 µM. Importantly, P155-185-chol showed significant suppression of A/Anhui/1/2013 H7N9 live virus propagation in MDCK cells and additive effects with NA inhibitors Oseltamivir and Zanamivir. These findings expand our knowledge of the entry properties of the novel H7N9 viruses, and they highlight the potential for developing a new class of inhibitors targeting viral entry for use in the next pandemic.
Collapse
Affiliation(s)
- Youhui Si
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianguo Li
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuqiang Niu
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xiuying Liu
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Lili Ren
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Li Guo
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Cheng
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hongli Zhou
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianwei Wang
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (WY); (QJ); (JW)
| | - Qi Jin
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (WY); (QJ); (JW)
| | - Wei Yang
- Ministry of Health Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- * E-mail: (WY); (QJ); (JW)
| |
Collapse
|
41
|
El-Khoury V, Pierson S, Szwarcbart E, Brons NHC, Roland O, Cherrier-De Wilde S, Plawny L, Van Dyck E, Berchem G. Disruption of autophagy by the histone deacetylase inhibitor MGCD0103 and its therapeutic implication in B-cell chronic lymphocytic leukemia. Leukemia 2014; 28:1636-46. [PMID: 24418989 PMCID: PMC4131250 DOI: 10.1038/leu.2014.19] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 12/04/2013] [Accepted: 12/31/2013] [Indexed: 12/19/2022]
Abstract
Evading apoptosis is a hallmark of B-cell chronic lymphocytic leukemia (CLL) cells and an obstacle to current chemotherapeutic approaches. Inhibiting histone deacetylase (HDAC) has emerged as a promising strategy to induce cell death in malignant cells. We have previously reported that the HDAC inhibitor MGCD0103 induces CLL cell death by activating the intrinsic pathway of apoptosis. Here, we show that MGCD0103 decreases the autophagic flux in primary CLL cells. Activation of the PI3K/AKT/mTOR pathway, together with the activation of caspases, and to a minor extent CAPN1, resulting in cleavage of autophagy components, were involved in MGCD0103-mediated inhibition of autophagy. In addition, MGCD0103 directly modulated the expression of critical autophagy genes at the transcriptional level that may contribute to autophagy impairment. Besides, we demonstrate that autophagy is a pro-survival mechanism in CLL whose disruption potentiates cell death induced by anticancer molecules including HDAC and cyclin-dependent kinase inhibitors. In particular, our data highlight the therapeutic potential of MGCD0103 as not only an inducer of apoptosis but also an autophagy suppressor in both combination regimens with molecules like flavopiridol, known to induce protective autophagy in CLL cells, or as an alternative to circumvent undesired immunomodulatory effects seen in the clinic with conventional autophagy inhibitors.
Collapse
Affiliation(s)
- V El-Khoury
- Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - S Pierson
- Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - E Szwarcbart
- Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - N H C Brons
- Flow Cytometry Core Facility, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - O Roland
- Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | | | - L Plawny
- Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| | - E Van Dyck
- Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg
| | - G Berchem
- 1] Laboratory of Experimental Hemato-Oncology, Centre de Recherche Public de la Santé, Luxembourg, Luxembourg [2] Centre Hospitalier de Luxembourg, Luxembourg, Luxembourg
| |
Collapse
|
42
|
Design of inhibitors of influenza virus membrane fusion: Synthesis, structure–activity relationship and in vitro antiviral activity of a novel indole series. Antiviral Res 2013; 99:125-35. [DOI: 10.1016/j.antiviral.2013.05.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 05/01/2013] [Accepted: 05/13/2013] [Indexed: 12/21/2022]
|
43
|
Abstract
Dengue is the most important arboviral disease in the world. As chloroquine, an antimalarial agent, has shown some antiviral effects, this study evaluated its effect in patients with dengue. A randomised, double-blind study was performed by administering chloroquine or placebo for three days to 129 patients with dengue-related symptoms. Of these patients, 37 were confirmed as having dengue and completed the study; in total, 19 dengue patients received chloroquine and 18 received placebo. There was no significant difference in the duration of the disease or the degree and days of fever. However, 12 patients (63%) with confirmed dengue reported a substantial decrease in pain intensity and a great improvement in their ability to perform daily activities (p = 0.0004) while on the medication and the symptoms returned immediately after these patients stopped taking the medication. The same effect was not observed in patients with diseases other than dengue. Therefore, this study shows that patients with dengue treated with chloroquine had an improvement in their quality of life and were able to resume their daily activities. However, as chloroquine did not alter the duration of the disease or the intensity and days of fever, further studies are necessary to confirm the clinical effects and to assess the side effects of chloroquine in dengue patients.
Collapse
Affiliation(s)
- Marcos Carvalho Borges
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | - Luiza Antunes Castro
- Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo,
Ribeirão Preto, SP, Brasil
| | | |
Collapse
|
44
|
Chloroquine: modes of action of an undervalued drug. Immunol Lett 2013; 153:50-7. [PMID: 23891850 DOI: 10.1016/j.imlet.2013.07.004] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 07/10/2013] [Accepted: 07/15/2013] [Indexed: 12/30/2022]
Abstract
For more than two decades, chloroquine (CQ) was largely and deliberately used as first choice drug for malaria treatment. However, worldwide increasing cases of resistant strains of Plasmodium have hampered its use. Nevertheless, CQ has recently been tested as adjunct therapy in several inflammatory situations, such as rheumatoid arthritis and transplantation procedures, presenting intriguing and promising results. In this review, we discuss recent findings and CQ mechanisms of action vis-à-vis its use as a broad adjunct therapy.
Collapse
|
45
|
Madrid PB, Chopra S, Manger ID, Gilfillan L, Keepers TR, Shurtleff AC, Green CE, Iyer LV, Dilks HH, Davey RA, Kolokoltsov AA, Carrion R, Patterson JL, Bavari S, Panchal RG, Warren TK, Wells JB, Moos WH, Burke RL, Tanga MJ. A systematic screen of FDA-approved drugs for inhibitors of biological threat agents. PLoS One 2013; 8:e60579. [PMID: 23577127 PMCID: PMC3618516 DOI: 10.1371/journal.pone.0060579] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 02/28/2013] [Indexed: 12/15/2022] Open
Abstract
Background The rapid development of effective medical countermeasures against potential biological threat agents is vital. Repurposing existing drugs that may have unanticipated activities as potential countermeasures is one way to meet this important goal, since currently approved drugs already have well-established safety and pharmacokinetic profiles in patients, as well as manufacturing and distribution networks. Therefore, approved drugs could rapidly be made available for a new indication in an emergency. Methodology/Principal Findings A large systematic effort to determine whether existing drugs can be used against high containment bacterial and viral pathogens is described. We assembled and screened 1012 FDA-approved drugs for off-label broad-spectrum efficacy against Bacillus anthracis; Francisella tularensis; Coxiella burnetii; and Ebola, Marburg, and Lassa fever viruses using in vitro cell culture assays. We found a variety of hits against two or more of these biological threat pathogens, which were validated in secondary assays. As expected, antibiotic compounds were highly active against bacterial agents, but we did not identify any non-antibiotic compounds with broad-spectrum antibacterial activity. Lomefloxacin and erythromycin were found to be the most potent compounds in vivo protecting mice against Bacillus anthracis challenge. While multiple virus-specific inhibitors were identified, the most noteworthy antiviral compound identified was chloroquine, which disrupted entry and replication of two or more viruses in vitro and protected mice against Ebola virus challenge in vivo. Conclusions/Significance The feasibility of repurposing existing drugs to face novel threats is demonstrated and this represents the first effort to apply this approach to high containment bacteria and viruses.
Collapse
Affiliation(s)
- Peter B Madrid
- Center for Infectious Disease and Biodefense Research, SRI International, Menlo Park, California, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wang J, Ma C, Wang J, Jo H, Canturk B, Fiorin G, Pinto LH, Lamb RA, Klein ML, DeGrado WF. Discovery of novel dual inhibitors of the wild-type and the most prevalent drug-resistant mutant, S31N, of the M2 proton channel from influenza A virus. J Med Chem 2013; 56:2804-12. [PMID: 23437766 DOI: 10.1021/jm301538e] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Anti-influenza drugs, amantadine and rimantadine, targeting the M2 channel from influenza A virus are no longer effective because of widespread drug resistance. S31N is the predominant and amantadine-resistant M2 mutant, present in almost all of the circulating influenza A strains as well as in the pandemic 2009 H1N1 and the highly pathogenic H5N1 flu strains. Thus, there is an urgent need to develop second-generation M2 inhibitors targeting the S31N mutant. However, the S31N mutant presents a huge challenge to drug discovery, and it has been considered undruggable for several decades. Using structural information, classical medicinal chemistry approaches, and M2-specific biological testing, we discovered benzyl-substituted amantadine derivatives with activity against both S31N and WT, among which 4-(adamantan-1-ylaminomethyl)-benzene-1,3-diol (44) is the most potent dual inhibitor. These inhibitors demonstrate that S31N is a druggable target and provide a new starting point to design novel M2 inhibitors that address the problem of drug-resistant influenza A infections.
Collapse
Affiliation(s)
- Jizhou Wang
- Influmedix Inc, 170 North Radnor-Chester Road, Suite 300, Radnor, Pennsylvania 19087, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zuo Z, de Abin M, Chander Y, Kuehn TH, Goyal SM, Pui DYH. Comparison of spike and aerosol challenge tests for the recovery of viable influenza virus from non-woven fabrics. Influenza Other Respir Viruses 2013; 7:637-44. [PMID: 23398976 PMCID: PMC5781197 DOI: 10.1111/irv.12095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/31/2012] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND To experimentally determine the survival kinetics of influenza virus on personal protective equipment (PPE) and to evaluate the risk of virus transfer from PPE, it is important to compare the effects on virus recovery of the method used to contaminate the PPE with virus and the type of eluent used to recover it. METHODS Avian influenza virus (AIV) was applied as a liquid suspension (spike test) and as an aerosol to three types of non-woven fabrics [polypropylene (PP), polyester (PET), and polyamide (Nylon)] that are commonly used in the manufacture of PPE. This was followed by virus recovery using eight different eluents (phosphate-buffered saline, minimum essential medium, and 1.5% or 3.0% beef extract at pH 7, 8, or 9). RESULTS For spike tests, no statistically significant difference was found in virus recovery using any of the eluents tested. Hydrophobic surfaces (PP and PET) yielded higher spiked virus recovery than hydrophilic Nylon. From all materials, the virus recovery was much lower in aerosol challenge tests than in spike tests. CONCLUSIONS Significant differences were found in the recovery of viable AIV from non-woven fabrics between spike and aerosol challenge tests. The findings of this study demonstrate the need for realistic aerosol challenge tests rather than liquid spike tests in studies of virus survival on surfaces where airborne transmission of influenza virus may get involved.
Collapse
Affiliation(s)
- Zhili Zuo
- Department of Mechanical Engineering, College of Science and Engineering, University of Minnesota, Minneapolis, MN 55455, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Enhancement of T cell-mediated immune responses to whole inactivated influenza virus by chloroquine treatment in vivo. Vaccine 2013; 31:1717-24. [PMID: 23380456 DOI: 10.1016/j.vaccine.2013.01.037] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2012] [Revised: 12/16/2012] [Accepted: 01/21/2013] [Indexed: 11/21/2022]
Abstract
Current influenza vaccines induce poor cross-reactive CD8+ T cell responses. Cellular immunity is generally specific for epitopes that are remarkably conserved among different subtypes, suggesting that strategies to improve the cross-presentation of viral antigens by dendritic cells (DC) could elicit a broadly protective immune response. Previous studies have shown that limited proteolysis within the endocytic pathway can favorably influence antigen processing and thus immune responses. Herein, we demonstrate that chloroquine improves the cross-presentation of non-replicating influenza virus in vitro and T cell responses in mice following a single administration of inactivated HI-X31 virus. CD8+ T cells were also recruited to lymph nodes draining the site of infection and able to reduce viral load following pulmonary challenge with the heterologous PR8 virus. These findings may have implications for vaccination strategies aimed at improving the cross-presentation capacity of DCs and thus the size of effector and memory CD8+ T cells against influenza vaccines.
Collapse
|
49
|
Farias KJS, Machado PRL, da Fonseca BAL. Chloroquine inhibits dengue virus type 2 replication in Vero cells but not in C6/36 cells. ScientificWorldJournal 2013; 2013:282734. [PMID: 23431254 PMCID: PMC3572654 DOI: 10.1155/2013/282734] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 12/23/2012] [Indexed: 11/17/2022] Open
Abstract
Dengue viruses are the most important arthropod-borne viruses in terms of morbidity and mortality in the world. Since there is no dengue vaccine available for human use, we have set out to investigate the use of chloroquine as an antiviral drug against dengue. Chloroquine, an amine acidotropic drug known to affect intracellular exocytic pathways by increasing endosomal pH, was used in the in vitro treatment of Vero and C6/36 cells infected with dengue virus type 2 (DENV-2). Real-time RT-PCR and plaque assays were used to quantify the DENV-2 load in infected Vero and C6/36 cells after chloroquine treatment. Our results showed that a dose of 50 μg/ml of chloroquine was not toxic to the cells and induced a statistically significant inhibition of virus production in infected Vero cells when compared to untreated cells. In C6/36 cells, chloroquine does not induce a statistically significant difference in viral replication when compared to untreated cells, showing that this virus uses an unlikely pathway of penetration in these cells, and results were also confirmed by the plaque assay (PFU). These data suggest that the inhibition of virus infection induced by chloroquine is due to interference with acidic vesicles in mammalian cells.
Collapse
Affiliation(s)
- Kleber Juvenal Silva Farias
- Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, 14049-900 Ribeirão Preto, SP, Brazil.
| | | | | |
Collapse
|
50
|
Protein glycosylation in infectious disease pathobiology and treatment. Open Life Sci 2011; 6:802. [PMID: 32215117 PMCID: PMC7088636 DOI: 10.2478/s11535-011-0050-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2011] [Accepted: 05/22/2011] [Indexed: 02/07/2023] Open
Abstract
A host of bacteria and viruses are dependent on O-linked and N-linked glycosylation to perform vital biological functions. Pathogens often have integral proteins that participate in host-cell interactions such as receptor binding and fusion with host membrane. Fusion proteins from a broad range of disparate viruses, such as paramyxovirus, HIV, ebola, and the influenza viruses share a variety of common features that are augmented by glycosylation. Each of these viruses contain multiple glycosylation sites that must be processed and modified by the host post-translational machinery to be fusogenically active. In most viruses, glycosylation plays a role in biogenesis, stability, antigenicity and infectivity. In bacteria, glycosylation events play an important role in the formation of flagellin and pili and are vitally important to adherence, attachment, infectivity and immune evasion. With the importance of glycosylation to pathogen survival, it is clear that a better understanding of the processes is needed to understand the pathogen requirement for glycosylation and to capitalize on this requirement for the development of novel therapeutics.
Collapse
|