1
|
Woodall M, Ellis S, Zhang S, Kembou-Ringert J, Kite KA, Buggiotti L, Jacobs AI, Agyeman AA, Masonou T, Palor M, McHugh TD, Breuer J, Standing JF, Smith CM. Efficient in vitro assay for evaluating drug efficacy and synergy against emerging SARS-CoV-2 strains. Antimicrob Agents Chemother 2025; 69:e0123324. [PMID: 39688407 PMCID: PMC11823597 DOI: 10.1128/aac.01233-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/23/2024] [Indexed: 12/18/2024] Open
Abstract
Novel and repurposed antiviral drugs are available for the treatment of coronavirus disease 2019 (COVID-19). However, antiviral combinations may be more potent and lead to faster viral clearance, but the methods for screening antiviral combinations against respiratory viruses are not well established and labor-intensive. Here, we describe a time-efficient (72-96 h) and simple in vitro drug-sensitivity assay for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) using standard 96-well plates. We employ different synergy models (zero interaction potency, highest single agent, Loewe, Bliss) to determine the efficacy of antiviral therapies and synergistic combinations against ancestral and emerging clinical SARS-CoV-2 strains. We found that monotherapy of remdesivir, nirmatrelvir, and active metabolite of molnupiravir (EIDD-1931) demonstrated baseline EC50s within clinically achievable levels of 4.34 mg/L (CI: 3.74-4.94 mg/L), 1.25 mg/L (CI: 1.10-1.45 mg/L), and 0.25 mg/L (CI: 0.20-0.30 mg/L), respectively, against the ancestral SARS-CoV-2 strain. However, their efficacy varied against newer Omicron variants BA.1.1.15 and BA.2, particularly with the protease inhibitor nirmatrelvir. We also found that remdesivir and nirmatrelvir have a consistent, strong synergistic effect (Bliss synergy score >10) at clinically relevant drug concentrations (nirmatrelvir 0.25-1 mg/L with remdesivir 1-4 mg/L) across all SARS-CoV-2 strains tested. This method offers a practical tool that streamlines the identification of effective combination therapies and the detection of antiviral resistance. Our findings support the use of antiviral drug combinations targeting multiple viral components to enhance COVID-19 treatment efficacy, particularly in the context of emerging viral strains.
Collapse
Affiliation(s)
| | - Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Shengyuan Zhang
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Kerry-Anne Kite
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Laura Buggiotti
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Amy I. Jacobs
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | | | - Tereza Masonou
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Machaela Palor
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Timothy D. McHugh
- UCL Centre for Clinical Microbiology, Royal Free Campus, London, United Kingdom
| | - Judith Breuer
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Joseph F. Standing
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Pharmacy, Great Ormond Street Hospital for Children, London, United Kingdom
| | - Claire M. Smith
- UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| |
Collapse
|
2
|
Arrousse N, Berdimurodov E, Bogacheva M, Zakham F, Esslali S, EL Kadiri S, Taleb M, Vapalahti O. N-alkylation of amines for the synthesis of potential antiviral agents: A structural modification approach. Heliyon 2024; 10:e38587. [PMID: 39397970 PMCID: PMC11470537 DOI: 10.1016/j.heliyon.2024.e38587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
The threat of emerging viral outbreaks has increased the need for fast and effective development of therapeutics against emerging pathogens. One approach is to modify the structure of existing therapeutic agents to achieve the desired antiviral properties. Here, we attempted to synthesize a new antiviral compound by modifying the structure of chloroquine using the N-alkylation of the primary amine (N1,N1-diethylpentane-1,4-diamine) that is used in chloroquine synthesis. Chloroquine is commonly used to treat malaria. Like chloroquine, chloroquine is used for treating conditions such as rheumatoid arthritis, lupus, and malaria. For instance, in malaria treatment, it targets and inhibits the growth of the malaria parasite, aiding in its elimination from the body. The synthesized compounds MP1, C1, and TT1 were further tested in vitro against the B.1 lineage of SARS-CoV-2. One of the compounds, MP1, demonstrated minor effectiveness, with an IC50 of XX at only a high concentration (at a concentration of 60 μM) and decreased both the number of SARS-CoV-2 copies and the amount of infectious virus. Although the synthesized compounds failed to markedly inhibit SARS-CoV-2, this could be a pontial mechanism for manipulating the drug structure against other pathogens. MP1, TT1, C1, and chloroquine diphosphate were used as ligands for molecular docking to determine the principal interactions between these compounds and the active site of the protein downloaded from the Protein Data Bank (PDB ID: 6lzg). Finally, ADMET assays were performed on the synthesized compounds to determine their pharmacokinetics and bioavailability.
Collapse
Affiliation(s)
- Nadia Arrousse
- School of Science and Engineering, Al Akhawayn University in Ifrane, Hassan II avenue, 53000 Ifrane, Morocco
- Laboratory of Engineering, Electrochemistry, Modelling and Environment (LIEME), Faculty of Sciences, University Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Elyor Berdimurodov
- Faculty of Chemistry, National University of Uzbekistan, Tashkent, 100034, Uzbekistan
- Physics and Chemistry, Western Caspian University, AZ-1001, Baku, Azerbaijan
- University of Tashkent for Applied Sciences, Str. Gavhar 1, 100149, Tashkent, Uzbekistan
- Physics and Chemistry, Tashkent Institute of Irrigation and Agricultural Mechanization Engineers” National Research University, 100000, Tashkent, Uzbekistan
| | - Mariia Bogacheva
- Viral Zone Research Unit, Faculty of Medicine, University of Helsinki, Finland
| | - Fathiah Zakham
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Soukaina Esslali
- Laboratory of Chemistry-Biology Applied to the Environment, Chemistry Department, Faculty of Sciences, Moulay-Ismail University, B.P. 11201, Zitoune, Meknes, Morocco
| | - Sghir EL Kadiri
- Laboratory of Applied Chemistry & Environment (LCAE), Faculty of Science, Mohammed First University, Oujda, Morocco
| | - Mustapha Taleb
- Laboratory of Engineering, Electrochemistry, Modelling and Environment (LIEME), Faculty of Sciences, University Sidi Mohamed Ben Abdellah, Fez, Morocco
| | - Olli Vapalahti
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
3
|
Garcia MC, Tsang KL, Lohit S, Deng J, Schneider T, Matos Silva J, Mbuagbaw L, Holbrook A. Hydroxychloroquine-Chloroquine, QT-Prolongation, and Major Adverse Cardiac Events: A Meta-analysis and Scoping Review. Ann Pharmacother 2024; 58:742-755. [PMID: 37881891 PMCID: PMC11151715 DOI: 10.1177/10600280231204969] [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] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVES We aimed to evaluate the high-quality literature on the frequency and nature of major adverse cardiac events (MACE) associated with either hydroxychloroquine (HCQ) or chloroquine (CQ). DATA SOURCES We searched Medline, Embase, International Pharmaceutical Abstracts, and Cochrane Central from 1996 onward using search strategies created in collaboration with medical science librarians. STUDY SELECTION AND DATA EXTRACTION Randomized controlled trials (RCTs) published in English language from January 1996 to September 2022, involving adult patients at least 18 years of age, were selected. Outcomes of interest were death, arrhythmias, syncope, and seizures. Random-effects meta-analyses were performed with a Treatment Arm Continuity Correction for single and double zero event studies. DATA SYNTHESIS By study drug, there were 31 HCQ RCTs (n = 6677), 9 CQ RCTs (n = 622), and 1 combined HCQ-CQ trial (n = 105). Mortality was the most commonly reported MACE at 220 of 255 events (86.3%), with no reports of torsades de pointes or sudden cardiac death. There was no increased risk of MACE with exposure to HCQ-CQ compared with control (risk ratio [RR] = 0.90, 95% CI = 0.69-1.17, I2 = 0%). RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE These findings have important implications with respect to patient reassurance and updated guidance for prescribing practices of these medications. CONCLUSIONS Despite listing as QT-prolonging meds, HCQ-CQ did not increase the risk of MACE.
Collapse
Affiliation(s)
- Michael Cristian Garcia
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Kai La Tsang
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
| | - Simran Lohit
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jiawen Deng
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Tyler Schneider
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Jessyca Matos Silva
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - Lawrence Mbuagbaw
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- Biostatistics Unit, Father Sean O’Sullivan Research Centre, St. Joseph’s Healthcare, Hamilton, ON, Canada
| | - Anne Holbrook
- Clinical Pharmacology & Toxicology Research Group, St. Joseph’s Healthcare, Hamilton, ON, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
- Division of Clinical Pharmacology & Toxicology, Department of Medicine, St. Joseph’s Healthcare, Hamilton, ON, Canada
| |
Collapse
|
4
|
Generalova LV, Laryushkin DP, Leneva IA, Ivanina AV, Trunova GV, Dolinnyi SV, Generalov EA. Evaluation of the Polysaccharide "Immeran" Activity in Syrian hamsters' Model of SARS-CoV-2. Viruses 2024; 16:423. [PMID: 38543788 PMCID: PMC10976179 DOI: 10.3390/v16030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/04/2024] [Accepted: 03/07/2024] [Indexed: 05/23/2024] Open
Abstract
COVID-19 is a highly contagious respiratory disease with a high number of lethal cases in humans, which causes the need to search for new therapeutic agents. Polysaccharides could be one of the prospective types of molecules with a large variety of biological activities, especially antiviral. The aim of this work was to study the specific antiviral activity of the drug "Immeran" on a model of a new coronavirus infection SARS-CoV-2 in hamsters. Based on the second experiment, intraperitoneal treatment with the drug according to a treatment regimen in doses of 500 and 1000 μg/kg (administration after an hour, then once a day every other day, a total of 3 administrations) was effective, reliably suppressing the replication of the virus in the lungs and, at a dose of 1000 μg/kg, prevented weight loss in animals. In all cases, the treatment stimulated the formation of virus-neutralizing antibodies to the SARS-CoV-2 virus, which suggests that the drug possesses adjuvant properties.
Collapse
Affiliation(s)
- Liubov Viktorovna Generalova
- Faculty of Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (L.V.G.); (S.V.D.)
| | - Denis Pavlovich Laryushkin
- Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, Institute of Cell Biophysics of the Russian Academy of Sciences, 142290 Pushchino, Russia;
| | - Irina Anatolievna Leneva
- Mechnikov Research Institute of Vaccines and Sera, Department of Virology, 105064 Moscow, Russia; (I.A.L.); (A.V.I.)
| | - Anna Valerievna Ivanina
- Mechnikov Research Institute of Vaccines and Sera, Department of Virology, 105064 Moscow, Russia; (I.A.L.); (A.V.I.)
| | - Galina Vladimirovna Trunova
- Federal State Budgetary Institution National Medical Research Radiological Center (FSBI NMRRC) of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow, Oncology Research Institute, 125284 Moscow, Russia;
| | - Sergei Vladimirovich Dolinnyi
- Faculty of Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (L.V.G.); (S.V.D.)
| | | |
Collapse
|
5
|
Li X, Le Y, Li Y, Chen S, Guo L, Fu X, Manjanatha MG, Mei N. Evaluation of weak genotoxicity of hydroxychloroquine in human TK6 cells. Toxicol Lett 2024; 393:84-95. [PMID: 38311193 PMCID: PMC11369915 DOI: 10.1016/j.toxlet.2024.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
Hydroxychloroquine (HCQ), a derivative of chloroquine (CQ), is an antimalarial and antirheumatic drug. Since there is limited data available on the genotoxicity of HCQ, in the current study, we used a battery of in vitro assays to systematically examine the genotoxicity of HCQ in human lymphoblastoid TK6 cells. We first showed that HCQ is not mutagenic in TK6 cells up to 80 μM with or without exogenous metabolic activation. Subsequently, we found that short-term (3-4 h) HCQ treatment did not cause DNA strand breakage as measured by the comet assay and the phosphorylation of histone H2A.X (γH2A.X), and did not induce chromosomal damage as determined by the micronucleus (MN) assay. However, after 24-h treatment, both CQ and HCQ induced comparable and weak DNA damage and MN formation in TK6 cells; upregulated p53 and p53-mediated DNA damage responsive genes; and triggered apoptosis and mitochondrial damage that may partially contribute to the observed MN formation. Using a benchmark dose (BMD) modeling analysis, the lower 95% confidence limit of BMD50 values (BMDL50) for MN induction in TK6 cells were about 19.7 μM for CQ and 16.3 μM for HCQ. These results provide additional information for quantitative genotoxic risk assessment of these drugs.
Collapse
Affiliation(s)
- Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| | - Yuan Le
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Yuxi Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Xin Fu
- Division of Pharmacology Toxicology Review, Office of Safety and Clinical Evaluation, Center for Drug Evaluation and Research, Silver Spring, MD 20993, USA
| | - Mugimane G Manjanatha
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA.
| |
Collapse
|
6
|
Gerhart J, Cox DS, Singh RSP, Chan PLS, Rao R, Allen R, Shi H, Masters JC, Damle B. A Comprehensive Review of the Clinical Pharmacokinetics, Pharmacodynamics, and Drug Interactions of Nirmatrelvir/Ritonavir. Clin Pharmacokinet 2024; 63:27-42. [PMID: 38177893 PMCID: PMC10786959 DOI: 10.1007/s40262-023-01339-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2023] [Indexed: 01/06/2024]
Abstract
Nirmatrelvir is a potent and selective inhibitor of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) main protease that is used as an oral antiviral coronavirus disease 2019 (COVID-19) treatment. To sustain unbound systemic trough concentrations above the antiviral in vitro 90% effective concentration value (EC90), nirmatrelvir is coadministered with 100 mg of ritonavir, a pharmacokinetic enhancer. Ritonavir inhibits nirmatrelvir's cytochrome P450 (CYP) 3A4-mediated metabolism which results in renal elimination becoming the primary route of nirmatrelvir elimination when dosed concomitantly. Nirmatrelvir exhibits absorption-limited nonlinear pharmacokinetics. When coadministered with ritonavir in patients with mild-to-moderate COVID-19, nirmatrelvir reaches a maximum concentration of 3.43 µg/mL (11.7× EC90) in approximately 3 h on day 5 of dosing, with a geometric mean day 5 trough concentration of 1.57 µg/mL (5.4× EC90). Drug interactions with nirmatrelvir/ritonavir (PAXLOVIDTM) are primarily attributed to ritonavir-mediated CYP3A4 inhibition, and to a lesser extent CYP2D6 and P-glycoprotein inhibition. Population pharmacokinetics and quantitative systems pharmacology modeling support twice daily dosing of 300 mg/100 mg nirmatrelvir/ritonavir for 5 days, with a reduced 150 mg/100 mg dose for patients with moderate renal impairment. Rapid clinical development of nirmatrelvir/ritonavir in response to the emerging COVID-19 pandemic was enabled by innovations in clinical pharmacology research, including an adaptive phase 1 trial design allowing direct to pivotal phase 3 development, fluorine nuclear magnetic resonance spectroscopy to delineate absorption, distribution, metabolism, and excretion profiles, and innovative applications of model-informed drug development to accelerate development.
Collapse
Affiliation(s)
- Jacqueline Gerhart
- Pfizer Inc, Research and Development, 500 Arcola Road, Collegeville, PA, 19424, USA.
| | - Donna S Cox
- Pfizer Inc, Research and Development, 500 Arcola Road, Collegeville, PA, 19424, USA
| | | | | | - Rohit Rao
- Pfizer Inc, Research and Development, Cambridge, MA, USA
| | - Richard Allen
- Pfizer Inc, Research and Development, Cambridge, MA, USA
| | - Haihong Shi
- Pfizer Inc, Research and Development, Groton, CT, USA
| | | | - Bharat Damle
- Pfizer Inc, Research and Development, New York, NY, USA
| |
Collapse
|
7
|
Alfaqeeh M, Zakiyah N, Suwantika AA, Shabrina Z. Evaluation of Global Post-Outbreak COVID-19 Treatment Interventions: A Systematic Review and Bibliometric Analysis of Randomized Controlled Trials. J Multidiscip Healthc 2023; 16:4193-4209. [PMID: 38152831 PMCID: PMC10752030 DOI: 10.2147/jmdh.s448786] [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: 11/08/2023] [Accepted: 12/18/2023] [Indexed: 12/29/2023] Open
Abstract
Purpose The outbreak of COVID-19 has led to a global pandemic with millions of cases and deaths. Many randomized controlled trials (RCTs) were conducted to establish effective therapies. However, the methodological quality of these trials is paramount, as it directly impacts the reliability of results. This systematic review and bibliometric analysis aim to assess the methodological approach, execution diversity, global trends, and distribution of COVID-19 treatment RCTs post-outbreak, covering the period from the second wave and onward up to the present. Methods We utilize articles from three electronic databases published from September 1, 2020, to April 1, 2023. Inclusion and exclusion criteria were applied to identify relevant RCTs. Data extraction involved the collection of various study details. Risk of Bias (RoB) 2 tool assessed methodological quality, while implementation variability was evaluated against registration information. Bibliometric analysis, including keyword co-occurrence and country distribution, used VOSviewer and Tableau software. Results Initially, 501 studies were identified, but only 22 met the inclusion criteria, of which 19 had registration information. The methodological quality assessment revealed deficiencies in five main domains: randomization process (36%), deviations from intended interventions (9%), missing outcome data (4%), measurement of the outcome (18%), and selection of reported results (4%). An analysis of alignment between research protocols and registration data revealed common deviations in eight critical aspects. Bibliometric findings showcased global collaboration in COVID-19 treatment RCTs, with Iran and Brazil prominently contributing, while keyword co-occurrence analysis illuminated prominent research trends and terms in study titles and abstracts. Conclusion This study offers valuable insights into the evaluation of COVID-19 treatment RCTs. The scarcity of high-quality RCTs highlights the importance of enhancing trial rigor and transparency in global health emergencies.
Collapse
Affiliation(s)
- Mohammed Alfaqeeh
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Neily Zakiyah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Auliya A Suwantika
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Center for Health Technology Assessment, Universitas Padjadjaran, Bandung, Indonesia
| | - Zahratu Shabrina
- Department of Geography, King’s College London, London, UK
- Regional Innovation, Graduate School, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
8
|
Barbosa AN, Chebabo A, Starling C, Pérez C, Cunha CA, de Luna D, Nunes EP, Zambrano G, Ferreira JC, Croda J, Falavigna M, Gomes-da-Silva MM, Thormann M, Cimerman S, Parahiba SM, Tanni S, Bernardo WM, Rodriguez-Morales AJ. Pan-American Guidelines for the treatment of SARS-CoV-2/COVID-19: a joint evidence-based guideline of the Brazilian Society of Infectious Diseases (SBI) and the Pan-American Association of Infectious Diseases (API). Ann Clin Microbiol Antimicrob 2023; 22:67. [PMID: 37550690 PMCID: PMC10408214 DOI: 10.1186/s12941-023-00623-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/28/2023] [Indexed: 08/09/2023] Open
Abstract
BACKGROUND Since the beginning of the COVID-19 pandemic, therapeutic options for treating COVID-19 have been investigated at different stages of clinical manifestations. Considering the particular impact of COVID-19 in the Americas, this document aims to present recommendations for the pharmacological treatment of COVID-19 specific to this population. METHODS Fifteen experts, members of the Brazilian Society of Infectious Diseases (SBI) and the Pan-American Association of Infectious Diseases (API) make up the panel responsible for developing this guideline. Questions were formulated regarding prophylaxis and treatment of COVID-19 in outpatient and inpatient settings. The outcomes considered in decision-making were mortality, hospitalisation, need for mechanical ventilation, symptomatic COVID-19 episodes, and adverse events. In addition, a systematic review of randomised controlled trials was conducted. The quality of evidence assessment and guideline development process followed the GRADE system. RESULTS Nine technologies were evaluated, and ten recommendations were made, including the use of tixagevimab + cilgavimab in the prophylaxis of COVID-19, tixagevimab + cilgavimab, molnupiravir, nirmatrelvir + ritonavir, and remdesivir in the treatment of outpatients, and remdesivir, baricitinib, and tocilizumab in the treatment of hospitalised patients with severe COVID-19. The use of hydroxychloroquine or chloroquine and ivermectin was discouraged. CONCLUSION This guideline provides recommendations for treating patients in the Americas following the principles of evidence-based medicine. The recommendations present a set of drugs that have proven effective in the prophylaxis and treatment of COVID-19, emphasising the strong recommendation for the use of nirmatrelvir/ritonavir in outpatients as the lack of benefit from the use of hydroxychloroquine and ivermectin.
Collapse
Affiliation(s)
- Alexandre Naime Barbosa
- Infectious Diseases Department - Botucatu School of Medicine - UNESP, Av. Prof. Mário R. G. Montenegro, s/n, Botucatu, SP, CEP 18.618-687, Brazil.
- Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubiao Jr, s/n, Botucatu, SP, CEP 18618-970, Brazil.
| | - Alberto Chebabo
- Universidade Federal do Rio de Janeiro, Avenida Professor Rodolpho Paulo Rocco, 255, 50. Andar, Rio de Janeiro, RJ, CEP 21941-913, Brazil
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
| | - Carlos Starling
- Sociedade Mineira de Infectologia - SMI, Avenida João Pinheiro, 161, Belo Horizonte, MG, CEP 30130-180, Brazil
| | - Clevy Pérez
- Universidad Autónoma de Santo Domingo (UASD), Avenida Simón Bolívar, 902, Santo Domingo, 10108, República Dominicana
| | - Clóvis Arns Cunha
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
- Universidade Federal do Paraná, Rua XV de Novembro, 1299, Curitiba, PR, CEP 80060-000, Brazil
| | - David de Luna
- Comisión Nacional de Arbitraje Médico, C Mitla, 250, Ciudad de México, 03020, México
| | - Estevão Portela Nunes
- Instituto Nacional de Infectologia (INI), Fiocruz, Avenida Brasil, 4365, Rio de Janeiro, RJ, CEP 21040-360, Brazil
| | - Gabriela Zambrano
- Faculty of Medicine, Department of Infectious Diseases, Universidad Central del Ecuador, Quito, Ecuador
- Pontificia Universidad Católica del Ecuador, Facultad de Medicina, Posgrado de Medicina Interna, Quito, Ecuador
| | - Juliana Carvalho Ferreira
- Divisão de Pneumologia, Instituto do Coração, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, Universidade de São Paulo, Avenida Dr. Enéas Carvalho de Aguiar, 44, São Paulo, SP, CEP 05403-900, Brazil
- Intensive Care Unit, AC Camargo Cancer Center, Rua Prof. Antônio Prudente, 211, São Paulo, SP, CEP 01509-001, Brazil
| | - Julio Croda
- Oswaldo Cruz Foundation, Avenida Costa e Silva, s/n, Cidade Universitária, Campo Grande, MS, CEP 79070-900, Brazil
| | - Maicon Falavigna
- HTAnalyze Consulting and Training, Rua João Abbott, 109, Porto Alegre, RS, CEP 90460-150, Brazil
| | - Monica Maria Gomes-da-Silva
- Infectious Disease Control Service, Clinical Hospital, Universidade Federal Do Paraná, Rua General Carneiro, 181, Curitiba, PR, CEP 80060-900, Brazil
| | - Monica Thormann
- Hospital Salvador Bienvenido Gautier, Calle Alexander Fleming, 177, Santo Domingo, 10514, Dominican Republic
| | - Sergio Cimerman
- Brazilian Society for Infectious Diseases, Rua Teixeira da Silva, 660, São Paulo, SP, CEP 04002-033, Brazil
- Institute of Infectious Diseases Emilio Ribas, Avenida Dr. Arnaldo, 165, São Paulo, SP, CEP 05402-000, Brazil
| | - Suena Medeiros Parahiba
- HTAnalyze Consulting and Training, Rua João Abbott, 109, Porto Alegre, RS, CEP 90460-150, Brazil
| | - Suzana Tanni
- Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubiao Jr, s/n, Botucatu, SP, CEP 18618-970, Brazil
| | - Wanderley Marques Bernardo
- Medical Education Development Center (CEDEM) of Medical Faculty of São Paulo University (FMUSP), São Paulo, SP, Brazil
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de Las Américas-Institución Universitaria Visión de Las Américas, 660003, Pereira, Risaralda, Colombia.
- Clinical Epidemiology and Biostatistics, Faculty of Health Sciences, Universidad Científica del Sur, Lima, 4861, Peru.
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Beirut, Lebanon.
- Latin American Network of Coronavirus Disease 2019 - COVID-19 Research (LANCOVID-19), Pereira, Risaralda, Colombia.
| |
Collapse
|
9
|
Basit SA, Qureshi R, Musleh S, Guler R, Rahman MS, Biswas KH, Alam T. COVID-19Base v3: Update of the knowledgebase for drugs and biomedical entities linked to COVID-19. Front Public Health 2023; 11:1125917. [PMID: 36950105 PMCID: PMC10025554 DOI: 10.3389/fpubh.2023.1125917] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
COVID-19 has taken a huge toll on our lives over the last 3 years. Global initiatives put forward by all stakeholders are still in place to combat this pandemic and help us learn lessons for future ones. While the vaccine rollout was not able to curb the spread of the disease for all strains, the research community is still trying to develop effective therapeutics for COVID-19. Although Paxlovid and remdesivir have been approved by the FDA against COVID-19, they are not free of side effects. Therefore, the search for a therapeutic solution with high efficacy continues in the research community. To support this effort, in this latest version (v3) of COVID-19Base, we have summarized the biomedical entities linked to COVID-19 that have been highlighted in the scientific literature after the vaccine rollout. Eight different topic-specific dictionaries, i.e., gene, miRNA, lncRNA, PDB entries, disease, alternative medicines registered under clinical trials, drugs, and the side effects of drugs, were used to build this knowledgebase. We have introduced a BLSTM-based deep-learning model to predict the drug-disease associations that outperforms the existing model for the same purpose proposed in the earlier version of COVID-19Base. For the very first time, we have incorporated disease-gene, disease-miRNA, disease-lncRNA, and drug-PDB associations covering the largest number of biomedical entities related to COVID-19. We have provided examples of and insights into different biomedical entities covered in COVID-19Base to support the research community by incorporating all of these entities under a single platform to provide evidence-based support from the literature. COVID-19Base v3 can be accessed from: https://covidbase-v3.vercel.app/. The GitHub repository for the source code and data dictionaries is available to the community from: https://github.com/91Abdullah/covidbasev3.0.
Collapse
Affiliation(s)
- Syed Abdullah Basit
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| | - Rizwan Qureshi
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| | - Saleh Musleh
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| | - Reto Guler
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, University of Cape Town, Cape Town, South Africa
- Department of Pathology, Division of Immunology and South African Medical Research Council (SAMRC) Immunology of Infectious Diseases, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Diseases and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - M. Sohel Rahman
- Department of Computer Science and Engineering, Bangladesh University of Engineering and Technology, Dhaka, Bangladesh
| | - Kabir H. Biswas
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Tanvir Alam
- College of Science and Engineering, Hamad Bin Khalifa University, Doha, Qatar
| |
Collapse
|
10
|
Haddad F, Dokmak G, Karaman R. A Comprehensive Review on the Efficacy of Several Pharmacologic Agents for the Treatment of COVID-19. Life (Basel) 2022; 12:1758. [PMID: 36362912 PMCID: PMC9692303 DOI: 10.3390/life12111758] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/15/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
SARS-CoV-2, the coronavirus disease-2019 (COVID-19), and the cause of the pandemic is extremely contagious among people and has spread around the world. Antivirals, immunomodulators, and other medications, such as antibiotics, stem cells, and plasma therapy, have all been utilized in the treatment of COVID-19. To better understand the clinical efficacy of these agents and to aid in the selection of effective COVID-19 therapies in various countries, this study reviewed the effectiveness of the various pharmacologic agents that have been used for COVID-19 therapy globally by summarizing the clinical outcomes that have been obtained from the clinical trials published on each drug related to COVID-19 infection. The Food and Drug Administration (FDA) has authorized the use of remdesivir, paxlovid, molnupiravir, baricitinib, tixagevimab-cilgavimab, and bebtelovimab for the management of COVID-19. On the other hand, most research advises against using chloroquine and hydroxychloroquine to treat COVID-19 patients because they are not beneficial. Although the FDA has given emergency use authorization for some monoclonal antibodies, including bamlanivimab, etesevimab, casirivimab, and imdevimab for managing COVID-19, they are not currently approved for use because the Omicron variant has significantly reduced their in vitro susceptibility. In this study, we also included a wide range of alternative therapy strategies that effectively treat COVID-19 patients, although further randomized studies are necessary to support and assess their applicability.
Collapse
Affiliation(s)
- Fatma Haddad
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 9103401, Palestine
- Faculty of Life Sciences, University of Bradford, Bradford BD7 1DP, UK
| | - Ghadeer Dokmak
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 9103401, Palestine
| | - Rafik Karaman
- Pharmaceutical Sciences Department, Faculty of Pharmacy, Al-Quds University, Jerusalem 9103401, Palestine
- Department of Sciences, University of Basilicata, 85100 Potenza, Italy
| |
Collapse
|
11
|
Plebani R, Bai H, Si L, Li J, Zhang C, Romano M. 3D Lung Tissue Models for Studies on SARS-CoV-2 Pathophysiology and Therapeutics. Int J Mol Sci 2022; 23:ijms231710071. [PMID: 36077471 PMCID: PMC9456220 DOI: 10.3390/ijms231710071] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 08/30/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19), has provoked more than six million deaths worldwide and continues to pose a major threat to global health. Enormous efforts have been made by researchers around the world to elucidate COVID-19 pathophysiology, design efficacious therapy and develop new vaccines to control the pandemic. To this end, experimental models are essential. While animal models and conventional cell cultures have been widely utilized during these research endeavors, they often do not adequately reflect the human responses to SARS-CoV-2 infection. Therefore, models that emulate with high fidelity the SARS-CoV-2 infection in human organs are needed for discovering new antiviral drugs and vaccines against COVID-19. Three-dimensional (3D) cell cultures, such as lung organoids and bioengineered organs-on-chips, are emerging as crucial tools for research on respiratory diseases. The lung airway, small airway and alveolus organ chips have been successfully used for studies on lung response to infection by various pathogens, including corona and influenza A viruses. In this review, we provide an overview of these new tools and their use in studies on COVID-19 pathogenesis and drug testing. We also discuss the limitations of the existing models and indicate some improvements for their use in research against COVID-19 as well as future emerging epidemics.
Collapse
Affiliation(s)
- Roberto Plebani
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence:
| | - Haiqing Bai
- Xellar Biosystems Inc., Cambridge, MA 02138, USA
| | - Longlong Si
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Li
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Chunhe Zhang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mario Romano
- Center on Advanced Studies and Technology (CAST), Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
12
|
Schwartz IS, Boulware DR, Lee TC. Hydroxychloroquine for COVID19: The curtains close on a comedy of errors. LANCET REGIONAL HEALTH. AMERICAS 2022; 11:100268. [PMID: 35531052 PMCID: PMC9069223 DOI: 10.1016/j.lana.2022.100268] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Affiliation(s)
| | - David R Boulware
- Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota, Minneapolis, USA
| | - Todd C Lee
- Division of Infectious Diseases, Department of Medicine, McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
13
|
Calvo-Alvarez E, Dolci M, Perego F, Signorini L, Parapini S, D’Alessandro S, Denti L, Basilico N, Taramelli D, Ferrante P, Delbue S. Antiparasitic Drugs against SARS-CoV-2: A Comprehensive Literature Survey. Microorganisms 2022; 10:1284. [PMID: 35889004 PMCID: PMC9320270 DOI: 10.3390/microorganisms10071284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [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
|