1
|
Boulon R, Mazeaud C, Farahani MD, Broquière M, Iddir M, Charpentier T, Anton A, Ayotte Y, Woo S, Lamarre A, Chatel-Chaix L, LaPlante SR. Repurposing Drugs and Synergistic Combinations as Potential Therapies for Inhibiting SARS-CoV-2 and Coronavirus Replication. ACS Pharmacol Transl Sci 2024; 7:4043-4055. [PMID: 39698276 PMCID: PMC11650740 DOI: 10.1021/acsptsci.4c00512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/20/2024]
Abstract
Drug repurposing can serve an important role in rapidly discovering medicament options for emerging microbial pandemics. In this study, a pragmatic approach is demonstrated for screening and testing drug combinations as potential broad-spectrum therapies against SARS-CoV-2 and other betacoronaviruses. Rapid cell-based phenotypic small molecule screens were executed using related common-cold-causing HCoV-OC43 betacoronavirus to identify replication inhibitors from a library of drugs approved by regulatory agencies for other indications. Given the best inhibitors, an expedient checkerboard strategy then served to identify synergistic drug combinations. These combinations were then validated using more challenging assays involving SARS-CoV-2 and variants. Promising drug combinations against multiple viral variants were discovered and involved Tilorone with Nelfinavir or Molnupiravir.
Collapse
Affiliation(s)
- Richard Boulon
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Clément Mazeaud
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Majid D. Farahani
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Mathilde Broquière
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Mustapha Iddir
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Tania Charpentier
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Anaïs Anton
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Yann Ayotte
- NMX
Research and Solutions|Accelerating drug discovery, 500 boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Simon Woo
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
- NMX
Research and Solutions|Accelerating drug discovery, 500 boulevard Cartier Ouest, Laval, Quebec H7V 5B7, Canada
| | - Alain Lamarre
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Laurent Chatel-Chaix
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| | - Steven R. LaPlante
- Institut
National de la Recherche Scientifique−Centre Armand-Frappier
Santé Biotechnologie, 531 boulevard des Prairies, Laval, Québec H7V
1B7, Canada
| |
Collapse
|
2
|
Bay AC, Clausen MR, Røge BT, Sydenham TV, Steinke K, Pedersen RM, Bang LL, Andersen TE, Jensen A, Madsen LW. Antiviral combination treatment of SARS-CoV-2 after repeated treatment failures of remdesivir monotherapy: A case report. IDCases 2024; 38:e02118. [PMID: 39634632 PMCID: PMC11615917 DOI: 10.1016/j.idcr.2024.e02118] [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: 02/23/2024] [Revised: 09/30/2024] [Accepted: 11/12/2024] [Indexed: 12/07/2024] Open
Abstract
Immunocompromised patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can have a longer duration of viral shedding and persistence of symptoms. The optimal treatment strategy for these patients remains to be established. This case describes a male in his late sixties with follicular lymphoma and persistent symptoms of infection with SARS-CoV-2 variant BA.2 who was treated with remdesivir five times over a period of six months. The clinical effect of remdesivir treatment decreased over time, and further viral sequencing revealed the emergence of mutations across the SARS-CoV-2 genome. Due to the lack of other treatment options, the patient was treated with a combination of remdesivir and molnupiravir for 10 days, and epcoritamab was discontinued, which led to the cessation of symptoms. This case illustrates the risk of a diminished effect of remdesivir with prolonged use and the need for treatment guidelines for immunocompromised patients with persistent COVID-19.
Collapse
Affiliation(s)
| | | | | | - Thomas V. Sydenham
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Kat Steinke
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Rune Micha Pedersen
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Line L. Bang
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Thomas E. Andersen
- Department of Clinical Microbiology, Odense University Hospital, Odense, Denmark
- Research Unit for Clinical Microbiology, University of Southern Denmark, Odense, Denmark
| | - Anders Jensen
- Department of Clinical Microbiology, Vejle Hospital, Vejle, Denmark
| | - Lone W. Madsen
- Department of Internal Medicine, Kolding Hospital, Kolding, Denmark
- Department of Infectious Diseases, Odense University Hospital, Odense, Denmark
- Department of Regional Health Research, University of Southern Denmark, Denmark
| |
Collapse
|
3
|
Aboras SI, Korany MA, Ebied SA, Haggag RS, Hamdy MMA. Hyphenating sustainability with chemometrics in chromatographic analysis of COVID combo therapy, nirmatrelvir and Molnupiravir, in presence of their overlapping degradation products; blue-green dual evaluation tools. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1247:124304. [PMID: 39270420 DOI: 10.1016/j.jchromb.2024.124304] [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: 07/28/2024] [Revised: 08/31/2024] [Accepted: 09/07/2024] [Indexed: 09/15/2024]
Abstract
Our manuscript managed to hyphenate the novelty and sustainability in one method. A novel combination of molnupiravir (MNP) and nirmatrelvir (NTV) was found to be a potential symbiotic therapy against SARS-CoV-2. Yet, there is no analytical method published for either determination or stability investigation of this combination simultaneously. So, the proposed HPLC technique focused on determination of MNP and NTV in presence of their degradation products. The sustainability was achieved in our method by using chemometrics tools to quantify NTV in presence of its co-eluted degradation product (NTV-D) without excessive time and solvent usage for separation (run time 5 min.). Moreover, the linearity parameters of both MNP and NTV, including correlation coefficient, LOD and LOQ, have been enhanced significantly after chemometrics treatment through convolution of the resultant derivative curves using trigonometric Fourier function. For example, LOQ of MNP decreased from 3.53 to 0.31 µg/mL and for NTV, LOQ decreased from 4.98 to 2.10 µg/mL after chemometrics treatment. The stability results of the proposed method indicates that no interaction or change in stability behavior of both drugs when co-administered with each other. Thus, this can be used as an empirical basis to initiate clinical trials of this combination for the treatment of COVID-19 patients. Additionally, in order to determine the impact of chemometric methods in minimizing analysis time and reducing solvent, energy, and waste consumption, our chemometric methodology is evaluated in terms of greenness and blueness (dichromic assessment) using AGREE and BAGI, respectively. Besides, the method sustainability using Hexagon was evaluated.
Collapse
Affiliation(s)
- Sara I Aboras
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Mohamed A Korany
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Shaza A Ebied
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza complex 21648, Alexandria, Egypt
| | - Rim S Haggag
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza complex 21648, Alexandria, Egypt
| | - Mohamed M A Hamdy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Beside Green Plaza complex 21648, Alexandria, Egypt
| |
Collapse
|
4
|
Chen Z, Cui Q, Ran Y, Achi JG, Chen Z, Rong L, Du R. A BSL-2 compliant mouse model of SARS-CoV-2 infection for efficient and convenient antiviral evaluation. J Virol 2024; 98:e0050424. [PMID: 38899934 PMCID: PMC11265351 DOI: 10.1128/jvi.00504-24] [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: 03/15/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Animal models of authentic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection require operation in biosafety level 3 (BSL-3) containment. In the present study, we established a mouse model employing a single-cycle infectious virus replicon particle (VRP) system of SARS-CoV-2 that can be safely handled in BSL-2 laboratories. The VRP [ΔS-VRP(G)-Luc] contains a SARS-CoV-2 genome in which the spike gene was replaced by a firefly luciferase (Fluc) reporter gene (Rep-Luci), and incorporates the vesicular stomatitis virus glycoprotein on the surface. Intranasal inoculation of ΔS-VRP(G)-Luc can successfully transduce the Rep-Luci genome into mouse lungs, initiating self-replication of Rep-Luci and, accordingly, inducing acute lung injury mimicking the authentic SARS-CoV-2 pathology. In addition, the reporter Fluc expression can be monitored using a bioluminescence imaging approach, allowing a rapid and convenient determination of viral replication in ΔS-VRP(G)-Luc-infected mouse lungs. Upon treatment with an approved anti-SARS-CoV-2 drug, VV116, the viral replication in infected mouse lungs was significantly reduced, suggesting that the animal model is feasible for antiviral evaluation. In summary, we have developed a BSL-2-compliant mouse model of SARS-CoV-2 infection, providing an advanced approach to study aspects of the viral pathogenesis, viral-host interactions, as well as the efficacy of antiviral therapeutics in the future.IMPORTANCESevere acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly contagious and pathogenic in humans; thus, research on authentic SARS-CoV-2 has been restricted to biosafety level 3 (BSL-3) laboratories. However, due to the scarcity of BSL-3 facilities and trained personnel, the participation of a broad scientific community in SARS-CoV-2 research had been greatly limited, hindering the advancement of our understanding on the basic virology as well as the urgently necessitated drug development. Previously, our colleagues Jin et al. had generated a SARS-CoV-2 replicon by replacing the essential spike gene in the viral genome with a Fluc reporter (Rep-Luci), which can be safely operated under BSL-2 conditions. By incorporating the Rep-Luci into viral replicon particles carrying vesicular stomatitis virus glycoprotein on their surface, and via intranasal inoculation, we successfully transduced the Rep-Luci into mouse lungs, developing a mouse model mimicking SARS-CoV-2 infection. Our model can serve as a useful platform for SARS-CoV-2 pathological studies and antiviral evaluation under BSL2 containment.
Collapse
Affiliation(s)
- Zinuo Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| | - Yan Ran
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jazmin Galvan Achi
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Zhaoyu Chen
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Ruikun Du
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Qingdao, China
| |
Collapse
|
5
|
Kinsella PM, Moso MA, Morrissey CO, Dendle C, Guy S, Bond K, Sasadeusz J, Slavin MA. Antiviral therapies for the management of persistent coronavirus disease 2019 in immunocompromised hosts: A narrative review. Transpl Infect Dis 2024; 26:e14301. [PMID: 38809102 DOI: 10.1111/tid.14301] [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: 12/10/2023] [Revised: 04/11/2024] [Accepted: 05/08/2024] [Indexed: 05/30/2024]
Abstract
Antiviral agents with activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have played a critical role in disease management; however, little is known regarding the efficacy of these medications in the treatment of SARS-CoV-2 infection in immunocompromised patients, particularly in the management of persistent SARS-CoV-2 positivity. This narrative review discusses the management of persistent coronavirus disease 2019 in immunocompromised hosts, with a focus on antiviral therapies. We identified 84 cases from the literature describing a variety of approaches, including prolonged antiviral therapy (n = 11), combination antivirals (n = 13), and mixed therapy with antiviral and antibody treatments (n = 60). A high proportion had an underlying haematologic malignancy (n = 67, 80%), and were in receipt of anti-CD20 agents (n = 51, 60%). Success was reported in 70 cases (83%) which varied according to the therapy type. Combination therapies with antivirals may be an effective approach for individuals with persistent SARS-CoV-2 positivity, particularly those that incorporate treatments aimed at increasing neutralizing antibody levels. Any novel approaches taken to this difficult management dilemma should be mindful of the emergence of antiviral resistance.
Collapse
Affiliation(s)
- Paul M Kinsella
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Michael A Moso
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | | | - Claire Dendle
- Monash Infectious Diseases, Monash Health, Melbourne, Australia
- School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Stephen Guy
- Department of Infectious Diseases, Eastern Health, Melbourne, Australia
- Eastern Health Clinical School, Monash University, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology, Royal Melbourne Hospital, Melbourne, Australia
- Victorian Infectious Diseases Reference Laboratory (VIDRL) at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Microbiology and Immunology, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Joseph Sasadeusz
- Department of Infectious Diseases, University of Melbourne at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Monica A Slavin
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at the Doherty Institute of Infection and Immunity, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
6
|
Orth HM, Flasshove C, Berger M, Hattenhauer T, Biederbick KD, Mispelbaum R, Klein U, Stemler J, Fisahn M, Doleschall AD, Baermann BN, Koenigshausen E, Tselikmann O, Killer A, de Angelis C, Gliga S, Stegbauer J, Spuck N, Silling G, Rockstroh JK, Strassburg CP, Brossart P, Panse JP, Jensen BEO, Luedde T, Boesecke C, Heine A, Cornely OA, Monin MB. Early combination therapy of COVID-19 in high-risk patients. Infection 2024; 52:877-889. [PMID: 38017344 PMCID: PMC11142969 DOI: 10.1007/s15010-023-02125-5] [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: 08/13/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
PURPOSE Prolonged shedding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been observed in immunocompromised hosts. Early monotherapy with direct-acting antivirals or monoclonal antibodies, as recommended by the international guidelines, does not prevent this with certainty. Dual therapies may therefore have a synergistic effect. METHODS This retrospective, multicentre study compared treatment strategies for corona virus disease-19 (COVID-19) with combinations of nirmatrelvir/ritonavir, remdesivir, molnupiravir, and/ or mABs during the Omicron surge. Co-primary endpoints were prolonged viral shedding (≥ 106 copies/ml at day 21 after treatment initiation) and days with SARS-CoV-2 viral load ≥ 106 copies/ml. Therapeutic strategies and risk groups were compared using odds ratios and Fisher's tests or Kaplan-Meier analysis and long-rank tests. Multivariable regression analysis was performed. RESULTS 144 patients were included with a median duration of SARS-CoV-2 viral load ≥ 106 copies/ml of 8.0 days (IQR 6.0-15.3). Underlying haematological malignancies (HM) (p = 0.03) and treatment initiation later than five days after diagnosis (p < 0.01) were significantly associated with longer viral shedding. Prolonged viral shedding was observed in 14.6% (n = 21/144), particularly in patients with underlying HM (OR 3.5; 95% CI 1.2-9.9; p = 0.02). Clinical courses of COVID-19 were mild to moderate with only few adverse effects potentially related to combination treatment. CONCLUSION Early combination treatment of COVID-19 effectively prevented prolonged viral shedding in 85.6% of cases. Considering the rapid viral clearance rates and low toxicity, individualized dual therapy approaches may be beneficial in high-risk patients.
Collapse
Affiliation(s)
- Hans Martin Orth
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Charlotte Flasshove
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Moritz Berger
- Institute for Medical Biometry, Informatics and Epidemiology, Bonn University Hospital, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Tessa Hattenhauer
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Kaja D Biederbick
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Rebekka Mispelbaum
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Uwe Klein
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jannik Stemler
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department I of Internal Medicine, European Diamond Excellence Centre for Medical Mycology (ECMM), University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Institute of Translational Research, Cologne Excellence Cluster On Cellular Stress Responses, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Kerpener Str. 62, 50937, Cologne, Germany
| | - Matthis Fisahn
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department I of Internal Medicine, European Diamond Excellence Centre for Medical Mycology (ECMM), University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Institute of Translational Research, Cologne Excellence Cluster On Cellular Stress Responses, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Kerpener Str. 62, 50937, Cologne, Germany
| | - Anna D Doleschall
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Ben-Niklas Baermann
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Hematology, Oncology, and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Eva Koenigshausen
- Department of Nephrology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Olga Tselikmann
- Department of Nephrology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Alexander Killer
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Clara de Angelis
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Smaranda Gliga
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Johannes Stegbauer
- Department of Nephrology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Nikolai Spuck
- Institute for Medical Biometry, Informatics and Epidemiology, Bonn University Hospital, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Gerda Silling
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Jürgen K Rockstroh
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Christian P Strassburg
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Peter Brossart
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Jens P Panse
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Hemostaseology and Stem Cell Transplantation, University Hospital RWTH Aachen, Pauwelsstraße 30, 52074, Aachen, Germany
| | - Björn-Erik Ole Jensen
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Tom Luedde
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Christoph Boesecke
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Annkristin Heine
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Oliver A Cornely
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany
- Department I of Internal Medicine, European Diamond Excellence Centre for Medical Mycology (ECMM), University of Cologne, Faculty of Medicine, and University Hospital of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- Institute of Translational Research, Cologne Excellence Cluster On Cellular Stress Responses, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Kerpener Str. 62, 50937, Cologne, Germany
| | - Malte B Monin
- Centre for Integrated Oncology (CIO), Aachen, Bonn, Cologne, Düsseldorf, (ABCD), Aachen, Germany.
- Department of Internal Medicine I, University Hospital Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- German Centre for Infection Research (DZIF), Partner-Site Cologne-Bonn, Venusberg-Campus 1, 53127, Bonn, Germany.
- Johanniter-Kliniken Bonn GmbH, Johanniter-Krankenhaus Bonn, Bonn, Germany.
| |
Collapse
|
7
|
Petit PR, Touret F, Driouich JS, Cochin M, Luciani L, Bernadin O, Laprie C, Piorkowski G, Fraisse L, Sjö P, Mowbray CE, Escudié F, Scandale I, Chatelain E, de Lamballerie X, Solas C, Nougairède A. Further preclinical characterization of molnupiravir against SARS-CoV-2: Antiviral activity determinants and viral genome alteration patterns. Heliyon 2024; 10:e30862. [PMID: 38803975 PMCID: PMC11128822 DOI: 10.1016/j.heliyon.2024.e30862] [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: 01/09/2024] [Revised: 04/12/2024] [Accepted: 05/07/2024] [Indexed: 05/29/2024] Open
Abstract
The SARS-CoV-2 pandemic has highlighted the need for broad-spectrum antiviral drugs to respond promptly to viral emergence. We conducted a preclinical study of molnupiravir (MOV) against SARS-CoV-2 to fully characterise its antiviral properties and mode of action. The antiviral activity of different concentrations of MOV was evaluated ex vivo on human airway epithelium (HAE) and in vivo in a hamster model at three escalating doses (150, 300 and 400 mg/kg/day) according to three different regimens (preventive, pre-emptive and curative). We assessed viral loads and infectious titres at the apical pole of HAE and in hamster lungs, and MOV trough concentration in plasma and lungs. To explore the mode of action of the MOV, the entire genomes of the collected viruses were deep-sequenced. MOV effectively reduced viral titres in HAE and in the lungs of treated animals. Early treatment after infection was a key factor in efficacy, probably associated with high lung concentrations of MOV, suggesting good accumulation in the lung. MOV induced genomic alteration in viral genomes with an increase in the number of minority variants, and predominant G to A transitions. The observed reduction in viral replication and its mechanism of action leading to lethal mutagenesis, supported by clinical trials showing antiviral action in humans, provide a convincing basis for further research as an additional means in the fight against COVID-19 and other RNA viruses.
Collapse
Affiliation(s)
- Paul-Rémi Petit
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Jean-Sélim Driouich
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Maxime Cochin
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Léa Luciani
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Ornéllie Bernadin
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | | | - Géraldine Piorkowski
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Laurent Fraisse
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Peter Sjö
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | | | - Fanny Escudié
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Ivan Scandale
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Eric Chatelain
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| | - Caroline Solas
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
- Laboratoire de Pharmacocinétique et Toxicologie, Hôpital La Timone, APHM, Marseille, France
| | - Antoine Nougairède
- Unité des Virus Émergents (UVE: Aix-Marseille Univ, Università di Corsica, IRD 190, Inserm 1207, IRBA), France
| |
Collapse
|
8
|
Gidari A, Sabbatini S, Bastianelli S, Pierucci S, Busti C, Svizzeretto E, Tommasi A, Pallotto C, Schiaroli E, Francisci D. Tixagevimab/Cilgavimab: Still a Valid Prophylaxis against COVID-19 New Variants? Viruses 2024; 16:354. [PMID: 38543720 PMCID: PMC10975286 DOI: 10.3390/v16030354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND this study aims to evaluate the efficacy of tixagevimab/cilgavimab (Evusheld™) against various SARS-CoV-2 variants, including newer Omicron sublineages, in an immunocompromised cohort and in vitro. STUDY DESIGN Conducted in Italy, this research involves immunocompromised patients who received Evusheld. It evaluates serum neutralization activity against different SARS-CoV-2 strains (20A.EU1, BA.5, BQ.1, XBB.1.5, XBB.1.16, and EG.5) before (T0), after 14 (T1), and after 30 (T2) days from the tixagevimab/cilgavimab injection. Furthermore, the in vitro activity of Evusheld against SARS-CoV-2 VOCs was evaluated. RESULTS The cohort was composed of 72 immunocompromised patients. The serum neutralizing activity of tixagevimab/cilgavimab-treated patients was notably lower against newer variants such as BQ.1, XBB.1.5, XBB.1.16, and EG.5. Then, the in vitro study detailed specific EC50 values to quantify the activity of tixagevimab/cilgavimab against various SARS-CoV-2 VOCs. Newer variants like BQ.1 and XBB.1.5 exhibited notably lower neutralization, underscoring the challenges in effectively countering the evolving virus. Interestingly, tixagevimab/cilgavimab maintained reduced but still valid activity against EG.5 with an EC50 of 189 ng/mL and Cmax/EC90 of 110.7. CONCLUSIONS Tixagevimab/cilgavimab efficacy wanes against novel subvariants. This underscores the critical need for ongoing adaptation and vigilance in prophylactic strategies to effectively counter the dynamic and unpredictable nature of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Anna Gidari
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, 06123 Perugia, Italy;
| | - Sabrina Bastianelli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Sara Pierucci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Chiara Busti
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Elisabetta Svizzeretto
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Andrea Tommasi
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Carlo Pallotto
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Elisabetta Schiaroli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| | - Daniela Francisci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, 06132 Perugia, Italy; (S.B.); (S.P.); (C.B.); (E.S.); (A.T.); (C.P.); (E.S.); (D.F.)
| |
Collapse
|
9
|
Khurshid R, Schulz JM, Hu J, Snowden TS, Reynolds RC, Schürer SC. Targeted degrader technologies as prospective SARS-CoV-2 therapies. Drug Discov Today 2024; 29:103847. [PMID: 38029836 PMCID: PMC10836335 DOI: 10.1016/j.drudis.2023.103847] [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: 09/30/2023] [Revised: 11/10/2023] [Accepted: 11/23/2023] [Indexed: 12/01/2023]
Abstract
COVID-19 remains a severe public health threat despite the WHO declaring an end to the public health emergency in May 2023. Continual development of SARS-CoV-2 variants with resistance to vaccine-induced or natural immunity necessitates constant vigilance as well as new vaccines and therapeutics. Targeted protein degradation (TPD) remains relatively untapped in antiviral drug discovery and holds the promise of attenuating viral resistance development. From a unique structural design perspective, this review covers antiviral degrader merits and challenges by highlighting key coronavirus protein targets and their co-crystal structures, specifically illustrating how TPD strategies can refine existing SARS-CoV-2 3CL protease inhibitors to potentially produce superior protease-degrading agents.
Collapse
Affiliation(s)
- Rabia Khurshid
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Joseph M Schulz
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Jiaming Hu
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Timothy S Snowden
- The University of Alabama, Department of Chemistry and Biochemistry and Center for Convergent Bioscience and Medicine, 250 Hackberry Lane, Tuscaloosa, AL 35487-0336, USA
| | - Robert C Reynolds
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35205, USA
| | - Stephan C Schürer
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA; Institute for Data Science & Computing, University of Miami, Coral Gables, FL 33146, USA.
| |
Collapse
|
10
|
Bianconi E, Gidari A, Souma M, Sabbatini S, Grifagni D, Bigiotti C, Schiaroli E, Comez L, Paciaroni A, Cantini F, Francisci D, Macchiarulo A. The hope and hype of ellagic acid and urolithins as ligands of SARS-CoV-2 Nsp5 and inhibitors of viral replication. J Enzyme Inhib Med Chem 2023; 38:2251721. [PMID: 37638806 PMCID: PMC10464554 DOI: 10.1080/14756366.2023.2251721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/29/2023] Open
Abstract
Non-structural protein 5 (Nsp5) is a cysteine protease that plays a key role in SARS-CoV-2 replication, suppressing host protein synthesis and promoting immune evasion. The investigation of natural products as a potential strategy for Nsp5 inhibition is gaining attention as a means of developing antiviral agents. In this work, we have investigated the physicochemical properties and structure-activity relationships of ellagic acid and its gut metabolites, urolithins A-D, as ligands of Nsp5. Results allow us to identify urolithin D as promising ligand of Nsp5, with a dissociation constant in the nanomolar range of potency. Although urolithin D is able to bind to the catalytic cleft of Nsp5, the appraisal of its viral replication inhibition against SARS-CoV-2 in Vero E6 assay highlights a lack of activity. While these results are discussed in the framework of the available literature reporting conflicting data on polyphenol antiviral activity, they provide new clues for natural products as potential viral protease inhibitors.
Collapse
Affiliation(s)
- Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Anna Gidari
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Maria Souma
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Medical Microbiology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Deborah Grifagni
- Centre for Magnetic Resonance, University of Florence, Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Carlo Bigiotti
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Elisabetta Schiaroli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Lucia Comez
- Istituto Officina dei Materiali-IOM, National Research Council-CNR, Perugia, Italy
| | | | - Francesca Cantini
- Centre for Magnetic Resonance, University of Florence, Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, Sesto Fiorentino, Italy
| | - Daniela Francisci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, University of Perugia, Perugia, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| |
Collapse
|
11
|
Su WC, Chen ZY, Chang YS, Jeng KS, Le UNP, Chou YC, Kuo LL, Melano I, Jesse, Wang WJ, Song YC, Li SR, Hung MC, Lai MMC, Lin CW. Functional assessments of SARS-CoV-2 single-round infectious particles with variant-specific spike proteins on infectivity, drug sensitivity, and antibody neutralization. Antiviral Res 2023; 220:105744. [PMID: 37944823 DOI: 10.1016/j.antiviral.2023.105744] [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: 07/29/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
Working with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is restricted to biosafety level III (BSL-3) laboratory. The study used a trans-complementation system consisting of virus-like particles (VLPs) and DNA-launched replicons to generate SARS-CoV-2 single-round infectious particles (SRIPs) with variant-specific spike (S) proteins. S gene of Wuhan-Hu-1 strain (SWH1) or Omicron BA.1 variant (SBA.1), along with the envelope (E) and membrane (M) genes, were cloned into a tricistronic vector, co-expressed in the cells to produce variant-specific S-VLPs. Additionally, the replicon of the WH1-like strain without S, E, M and accessory genes, was engineered under the control by a CMV promoter to produce self-replicating RNAs within VLP-producing cells, led to create SWH1- and SBA.1-based SARS-CoV-2 SRIPs. The SBA.1-based SRIP showed lower virus yield, replication, N protein expression, fusogenicity, and infectivity compared to SWH1-based SRIPs. SBA.1-based SRIP also exhibited intermediate resistance to neutralizing antibodies produced by SWH1-based vaccines, but were effective at infecting cells with low ACE2 expression. Importantly, both S-based SRIPs responded similarly to remdesivir and GC376, with EC50 values ranging from 0.17 to 1.46 μM, respectively. The study demonstrated that this trans-complementation system is a reliable and efficient tool for generating SARS-CoV-2 SRIPs with variant-specific S proteins. SARS-CoV-2 SRIPs, mimicking authentic live viruses, facilitate comprehensive analysis of variant-specific virological characteristics, including antibody neutralization, and drug sensitivity in non-BSL-3 laboratories.
Collapse
Affiliation(s)
- Wen-Chi Su
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; International Master's Program of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan; Drug Development Center, China Medical University, Taichung, 404333, Taiwan
| | - Zan-Yu Chen
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404333, Taiwan
| | - Young-Sheng Chang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404333, Taiwan
| | - King-Song Jeng
- Department of Medical Research, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Uyen Nguyen Phuong Le
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404333, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 404333, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, 115201, Taiwan
| | - Li-Lan Kuo
- International Master's Program of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan
| | - Ivonne Melano
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan
| | - Jesse
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan
| | - Wei-Jan Wang
- Department of Biological Science and Technology, China Medical University, Taichung, 404333, Taiwan
| | - Ying-Chyi Song
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Sin-Rong Li
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404333, Taiwan; Department of Laboratory Medicine, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; Research Center for Cancer Biology, China Medical University, Taichung, 404327, Taiwan; Center for Molecular Medicine, China Medical University Hospital, Taichung, 404327, Taiwan
| | - Michael M C Lai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; Institute of Molecular Biology, Academia Sinica, Taipei, 115201, Taiwan.
| | - Cheng-Wen Lin
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404333, Taiwan; Drug Development Center, China Medical University, Taichung, 404333, Taiwan; Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, 404333, Taiwan; Department of Biological Science and Technology, China Medical University, Taichung, 404333, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Wufeng, Taichung, 413305, Taiwan.
| |
Collapse
|
12
|
Valipour M, Irannejad H, Keyvani H. An Overview on Anti-COVID-19 Drug Achievements and Challenges Ahead. ACS Pharmacol Transl Sci 2023; 6:1248-1265. [PMID: 37705590 PMCID: PMC10496143 DOI: 10.1021/acsptsci.3c00121] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Indexed: 09/15/2023]
Abstract
The appearance of several coronavirus pandemics/epidemics during the last two decades (SARS-CoV-1 in 2002, MERS-CoV in 2012, and SARS-CoV-2 in 2019) indicates that humanity will face increasing challenges from coronaviruses in the future. The emergence of new strains with similar transmission characteristics as SARS-CoV-2 and mortality rates similar to SARS-CoV-1 (∼10% mortality) or MERS-CoV (∼35% mortality) in the future is a terrifying possibility. Therefore, getting enough preparations to face such risks is an inevitable necessity. The present study aims to review the drug achievements and challenges in the fight against SARS-CoV-2 with a combined perspective derived from pharmacology, pharmacotherapy, and medicinal chemistry insights. Appreciating all the efforts made during the past few years, there is strong evidence that the desired results have not yet been achieved and research in this area should still be pursued seriously. By expressing some pessimistic possibilities and concluding that the drug discovery and pharmacotherapy of COVID-19 have not been successful so far, this short essay tries to draw the attention of responsible authorities to be more prepared against future coronavirus epidemics/pandemics.
Collapse
Affiliation(s)
- Mehdi Valipour
- Razi
Drug Research Center, Iran University of
Medical Sciences, Tehran 1134845764, Iran
| | - Hamid Irannejad
- Department
of Medicinal Chemistry, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari 48157-33971, Iran
| | - Hossein Keyvani
- Department
of Virology, School of Medicine, Iran University
of Medical Sciences, Tehran 1134845764, Iran
| |
Collapse
|
13
|
Yan D, Yan B. Viral target and metabolism-based rationale for combined use of recently authorized small molecule COVID-19 medicines: Molnupiravir, nirmatrelvir, and remdesivir. Fundam Clin Pharmacol 2023; 37:726-738. [PMID: 36931725 PMCID: PMC10505250 DOI: 10.1111/fcp.12889] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 02/12/2023] [Accepted: 02/27/2023] [Indexed: 03/19/2023]
Abstract
The COVID-19 pandemic remains a major health concern worldwide, and SARS-CoV-2 is continuously evolving. There is an urgent need to identify new antiviral drugs and develop novel therapeutic strategies. Combined use of newly authorized COVID-19 medicines including molnupiravir, nirmatrelvir, and remdesivir has been actively pursued. Mechanistically, nirmatrelvir inhibits SARS-CoV-2 replication by targeting the viral main protease (Mpro ), a critical enzyme in the processing of the immediately translated coronavirus polyproteins for viral replication. Molnupiravir and remdesivir, on the other hand, inhibit SARS-CoV-2 replication by targeting RNA-dependent RNA-polymerase (RdRp), which is directly responsible for genome replication and production of subgenomic RNAs. Molnupiravir targets RdRp and induces severe viral RNA mutations (genome), commonly referred to as error catastrophe. Remdesivir, in contrast, targets RdRp and causes chain termination and arrests RNA synthesis of the viral genome. In addition, all three medicines undergo extensive metabolism with strong therapeutic significance. Molnupiravir is hydrolytically activated by carboxylesterase-2 (CES2), nirmatrelvir is inactivated by cytochrome P450-based oxidation (e.g., CYP3A4), and remdesivir is hydrolytically activated by CES1 but covalently inhibits CES2. Additionally, remdesivir and nirmatrelvir are oxidized by the same CYP enzymes. The distinct mechanisms of action provide strong rationale for their combined use. On the other hand, these drugs undergo extensive metabolism that determines their therapeutic potential. This review discusses how metabolism pathways and enzymes involved should be carefully considered during their combined use for therapeutic synergy.
Collapse
Affiliation(s)
- Daisy Yan
- Department of Dermatology, Boston University School of Medicine 609 Albany Street Boston, MA 02118
| | - Bingfang Yan
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229
| |
Collapse
|
14
|
Mikulska M, Sepulcri C, Dentone C, Magne F, Balletto E, Baldi F, Labate L, Russo C, Mirabella M, Magnasco L, Di Grazia C, Ghiggi C, Raiola AM, Giacobbe DR, Vena A, Beltramini S, Bruzzone B, Lemoli RM, Angelucci E, Bassetti M. Triple Combination Therapy With 2 Antivirals and Monoclonal Antibodies for Persistent or Relapsed Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Immunocompromised Patients. Clin Infect Dis 2023; 77:280-286. [PMID: 36976301 DOI: 10.1093/cid/ciad181] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Severely immunocompromised patients are at risk for prolonged or relapsed Coronavirus Disease 2019 (COVID-19), leading to increased morbidity and mortality. We aimed to evaluate efficacy and safety of combination treatment in immunocompromised COVID-19 patients. METHODS We included all immunocompromised patients with prolonged/relapsed COVID-19 treated with combination therapy with 2 antivirals (remdesivir plus nirmatrelvir/ritonavir, or molnupiravir in case of renal failure) plus, if available, anti-spike monoclonal antibodies (mAbs), between February and October 2022. The main outcomes were virological response at day 14 (negative Severe Acute Respiratory Syndrome Coronavirus 2 [SARS-CoV-2] swab) and virological and clinical response (alive, asymptomatic, with negative SARS-CoV-2 swab) at day 30 and the last follow-up. RESULTS Overall, 22 patients (Omicron variant in 17/18) were included: 18 received full combination of 2 antivirals and mAbs and 4 received 2 antivirals only; in 20 of 22 (91%) patients, 2 antivirals were nirmatrelvir/ritonavir plus remdesivir. Nineteen (86%) patients had hematological malignancy, and 15 (68%) had received anti-CD20 therapy. All were symptomatic; 8 (36%) required oxygen. Four patients received a second course of combination treatment. The response rate at day 14, day 30, and last follow-up was 75% (15/20 evaluable), 73% (16/22), and 82% (18/22), respectively. Day 14 and 30 response rates were significantly higher when combination therapy included mAbs. Higher number of vaccine doses was associated with better final outcome. Two patients (9%) developed severe side effects (bradycardia leading to remdesivir discontinuation and myocardial infarction). CONCLUSIONS Combination therapy including 2 antivirals (mainly remdesivir and nirmatrelvir/ritonavir) and mAbs was associated with high rate of virological and clinical response in immunocompromised patients with prolonged/relapsed COVID-19.
Collapse
Affiliation(s)
- Malgorzata Mikulska
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Sepulcri
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Dentone
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Magne
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Elisa Balletto
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Federico Baldi
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Laura Labate
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Russo
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Michele Mirabella
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Laura Magnasco
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Carmen Di Grazia
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Chiara Ghiggi
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Anna Maria Raiola
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Daniele Roberto Giacobbe
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Antonio Vena
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Bianca Bruzzone
- Department of Health Sciences, Hygiene Unit, Ospedale Policlinico San Martino, University of Genoa, Genova, Italy
| | - Roberto M Lemoli
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, Clinic of Hematology, University of Genoa, Genova, Italy
| | - Emanuele Angelucci
- Ematologia e Terapie Cellulari, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Matteo Bassetti
- Division of Infectious Diseases, Department of Health Sciences, University of Genoa, Genoa, Italy
- Division of Infectious Diseases, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
15
|
Rosenke K, Lewis MC, Feldmann F, Bohrnsen E, Schwarz B, Okumura A, Bohler WF, Callison J, Shaia C, Bosio CM, Lovaglio J, Saturday G, Jarvis MA, Feldmann H. Combined molnupiravir-nirmatrelvir treatment improves the inhibitory effect on SARS-CoV-2 in macaques. JCI Insight 2023; 8:166485. [PMID: 36574296 PMCID: PMC9977490 DOI: 10.1172/jci.insight.166485] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/21/2022] [Indexed: 12/28/2022] Open
Abstract
The periodic emergence of SARS-CoV-2 variants of concern (VOCs) with unpredictable clinical severity and ability to escape preexisting immunity emphasizes the continued need for antiviral interventions. Two small molecule inhibitors, molnupiravir (MK-4482), a nucleoside analog, and nirmatrelvir (PF-07321332), a 3C-like protease inhibitor, have recently been approved as monotherapy for use in high-risk patients with COVID-19. As preclinical data are only available for rodent and ferret models, here we assessed the efficacy of MK-4482 and PF-07321332 alone and in combination against infection with the SARS-CoV-2 Delta VOC in the rhesus macaque COVID-19 model. Macaques were infected with the SARS-CoV-2 Delta variant and treated with vehicle, MK-4482, PF-07321332, or a combination of MK-4482 and PF-07321332. Clinical exams were performed at 1, 2, and 4 days postinfection to assess disease and virological parameters. Notably, use of MK-4482 and PF-07321332 in combination improved the individual inhibitory effect of both drugs, resulting in milder disease progression, stronger reduction of virus shedding from mucosal tissues of the upper respiratory tract, stronger reduction of viral replication in the lower respiratory tract, and reduced lung pathology. Our data strongly indicate superiority of combined MK-4482 and PF-07321332 treatment of SARS-CoV-2 infections as demonstrated in the closest COVID-19 surrogate model of human infection.
Collapse
Affiliation(s)
| | | | | | - Eric Bohrnsen
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | - Benjamin Schwarz
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | | | | | | | | | - Catharine M Bosio
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Hamilton, Montana, USA
| | | | | | - Michael A Jarvis
- Laboratory of Virology.,University of Plymouth, Plymouth, Devon, United Kingdom.,The Vaccine Group Ltd, Plymouth, Devon, United Kingdom
| | | |
Collapse
|
16
|
Focosi D, McConnell S, Shoham S, Casadevall A, Maggi F, Antonelli G. Nirmatrelvir and COVID-19: development, pharmacokinetics, clinical efficacy, resistance, relapse, and pharmacoeconomics. Int J Antimicrob Agents 2023; 61:106708. [PMID: 36603694 DOI: 10.1016/j.ijantimicag.2022.106708] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023]
Abstract
Nirmatrelvir/ritonavir (N/R) is one of the most effective antiviral drugs against SARS-CoV-2. The preclinical development, pharmacodynamics and pharmacokinetics of N/R are reviewed herein. Randomized clinical trials have been conducted exclusively with pre-Omicron variants of concern, but in vitro studies show that efficacy against all Omicron sublineages is preserved, as confirmed by post-marketing observational studies. Nevertheless, investigations of large viral genome repositories have shown that mutation in the main protease causing resistance to N/R are increasingly frequent. In addition, virological and clinical rebounds after N/R discontinuation have been reported in immunocompetent patients. This finding is of concern when translated to immunocompromised patients, in whom N/R efficacy has not been formally investigated in clinical trials. Economical sustainability and perspectives for this therapeutic arena are discussed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy.
| | - Scott McConnell
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD, USA
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD, USA
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, MD, USA
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases "Spallanzani", Rome, Italy
| | - Guido Antonelli
- Department of Molecular Medicine, Sapienza University of Rome, and Sapienza University Hospital "Policlinico Umberto I", Rome, Italy
| |
Collapse
|
17
|
Anjani, Kumar S, Rathi B, Poonam. Recent updates on the biological efficacy of approved drugs and potent synthetic compounds against SARS-CoV-2. RSC Adv 2023; 13:3677-3687. [PMID: 36756584 PMCID: PMC9890797 DOI: 10.1039/d2ra06834f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/16/2023] [Indexed: 01/27/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), also known as COVID-19, has triggered a global pandemic that has prompted severe public health concerns. Researchers worldwide are continuously trying to find options that could be effective against COVID-19. The main focus of research during the initial phase of the pandemic was to use the already approved drugs as supportive care, and efforts were made to find new therapeutic options. Nirmatrelvir (PF-07321332), a Pfizer chemical, recently received approval for usage in conjunction with ritonavir. This mini-review summarises the biological effectiveness of vital synthetic compounds and FDA-approved medications against SARS-CoV-2. Understanding how functional groups are included in the creation of synthetic compounds could help enhance the biological activity profile of those compounds to increase their efficacy against SARS-CoV-2. This opened the way for researchers to explore opportunities to develop better therapeutics by investigating synthetic analogs.
Collapse
Affiliation(s)
- Anjani
- Department of Chemistry, Chaudhary Charan Singh Haryana Agricultural University Hisar Haryana-125004 India
| | - Sumit Kumar
- Department of Chemistry, Miranda House, University of Delhi Delhi-110007 India
| | - Brijesh Rathi
- Laboratory for Translational Chemistry and Drug Discovery, Hansraj College, University of Delhi Delhi-110007 India
- Delhi School of Public Health, Institute of Eminence, University of Delhi Delhi-110007 India
| | - Poonam
- Department of Chemistry, Miranda House, University of Delhi Delhi-110007 India
- Delhi School of Public Health, Institute of Eminence, University of Delhi Delhi-110007 India
| |
Collapse
|
18
|
Erdik B. Antiviral treatment of COVID-19 is associated with lack of immune response. J Med Virol 2023; 95:e28233. [PMID: 36257836 PMCID: PMC9874897 DOI: 10.1002/jmv.28233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/13/2022] [Accepted: 10/15/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Baran Erdik
- CCM HealthBoard of AdvisorsBrooklynNew YorkUSA
| |
Collapse
|
19
|
Alteri C, Fox V, Scutari R, Burastero GJ, Volpi S, Faltoni M, Fini V, Granaglia A, Esperti S, Gallerani A, Costabile V, Fontana B, Franceschini E, Meschiari M, Campana A, Bernardi S, Villani A, Bernaschi P, Russo C, Guaraldi G, Mussini C, Perno CF. A proof-of-concept study on the genomic evolution of Sars-Cov-2 in molnupiravir-treated, paxlovid-treated and drug-naïve patients. Commun Biol 2022; 5:1376. [PMID: 36522489 PMCID: PMC9753865 DOI: 10.1038/s42003-022-04322-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/30/2022] [Indexed: 12/16/2022] Open
Abstract
Little is known about SARS-CoV-2 evolution under Molnupiravir and Paxlovid, the only antivirals approved for COVID-19 treatment. By investigating SARS-CoV-2 variability in 8 Molnupiravir-treated, 7 Paxlovid-treated and 5 drug-naïve individuals at 4 time-points (Days 0-2-5-7), a higher genetic distance is found under Molnupiravir pressure compared to Paxlovid and no-drug pressure (nucleotide-substitutions/site mean±Standard error: 18.7 × 10-4 ± 2.1 × 10-4 vs. 3.3 × 10-4 ± 0.8 × 10-4 vs. 3.1 × 10-4 ± 0.8 × 10-4, P = 0.0003), peaking between Day 2 and 5. Molnupiravir drives the emergence of more G-A and C-T transitions than other mutations (P = 0.031). SARS-CoV-2 selective evolution under Molnupiravir pressure does not differ from that under Paxlovid or no-drug pressure, except for orf8 (dN > dS, P = 0.001); few amino acid mutations are enriched at specific sites. No RNA-dependent RNA polymerase (RdRp) or main proteases (Mpro) mutations conferring resistance to Molnupiravir or Paxlovid are found. This proof-of-concept study defines the SARS-CoV-2 within-host evolution during antiviral treatment, confirming higher in vivo variability induced by Molnupiravir compared to Paxlovid and drug-naive, albeit not resulting in apparent mutation selection.
Collapse
Affiliation(s)
- Claudia Alteri
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Valeria Fox
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Rossana Scutari
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giulia Jole Burastero
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara Volpi
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Faltoni
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Vanessa Fini
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Annarita Granaglia
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Sara Esperti
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Altea Gallerani
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentino Costabile
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Beatrice Fontana
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | - Andrea Campana
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Stefania Bernardi
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Alberto Villani
- Academic Department of Pediatrics, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Paola Bernaschi
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Cristina Russo
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy
| | - Giovanni Guaraldi
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Department of Infectious Diseases, University of Modena and Reggio Emilia, Modena, Italy
| | - Carlo Federico Perno
- Multimodal Research Area, Microbiology and Diagnostics of Immunology Unit, Bambino Gesù Children Hospital IRCCS, Rome, Italy.
| |
Collapse
|
20
|
Wagoner J, Herring S, Hsiang TY, Ianevski A, Biering SB, Xu S, Hoffmann M, Pöhlmann S, Gale M, Aittokallio T, Schiffer JT, White JM, Polyak SJ. Combinations of Host- and Virus-Targeting Antiviral Drugs Confer Synergistic Suppression of SARS-CoV-2. Microbiol Spectr 2022; 10:e0333122. [PMID: 36190406 PMCID: PMC9718484 DOI: 10.1128/spectrum.03331-22] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 02/08/2023] Open
Abstract
Three directly acting antivirals (DAAs) demonstrated substantial reduction in COVID-19 hospitalizations and deaths in clinical trials. However, these agents did not completely prevent severe illness and are associated with cases of rebound illness and viral shedding. Combination regimens can enhance antiviral potency, reduce the emergence of drug-resistant variants, and lower the dose of each component in the combination. Concurrently targeting virus entry and virus replication offers opportunities to discover synergistic drug combinations. While combination antiviral drug treatments are standard for chronic RNA virus infections, no antiviral combination therapy has been approved for SARS-CoV-2. Here, we demonstrate that combining host-targeting antivirals (HTAs) that target TMPRSS2 and hence SARS-CoV-2 entry, with the DAA molnupiravir, which targets SARS-CoV-2 replication, synergistically suppresses SARS-CoV-2 infection in Calu-3 lung epithelial cells. Strong synergy was observed when molnupiravir, an oral drug, was combined with three TMPRSS2 (HTA) oral or inhaled inhibitors: camostat, avoralstat, or nafamostat. The combination of camostat plus molnupiravir was also effective against the beta and delta variants of concern. The pyrimidine biosynthesis inhibitor brequinar combined with molnupiravir also conferred robust synergistic inhibition. These HTA+DAA combinations had similar potency to the synergistic all-DAA combination of molnupiravir plus nirmatrelvir, the protease inhibitor found in paxlovid. Pharmacodynamic modeling allowed estimates of antiviral potency at all possible concentrations of each agent within plausible therapeutic ranges, suggesting possible in vivo efficacy. The triple combination of camostat, brequinar, and molnupiravir further increased antiviral potency. These findings support the development of HTA+DAA combinations for pandemic response and preparedness. IMPORTANCE Imagine a future viral pandemic where if you test positive for the new virus, you can quickly take some medicines at home for a few days so that you do not get too sick. To date, only single drugs have been approved for outpatient use against SARS-CoV-2, and we are learning that these have some limitations and may succumb to drug resistance. Here, we show that combinations of two oral drugs are better than the single ones in blocking SARS-CoV-2, and we use mathematical modeling to show that these drug combinations are likely to work in people. We also show that a combination of three oral drugs works even better at eradicating the virus. Our findings therefore bode well for the development of oral drug cocktails for at home use at the first sign of an infection by a coronavirus or other emerging viral pathogens.
Collapse
Affiliation(s)
- Jessica Wagoner
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Shawn Herring
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Tien-Ying Hsiang
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Aleksandr Ianevski
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Scott B. Biering
- Division of Infectious Diseases and Vaccinology, School of Public Health, University of California—Berkeley, Berkeley, California, USA
| | - Shuang Xu
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University of Göttingen, Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Leibniz Institute for Primate Research, Göttingen, Germany
- Faculty of Biology and Psychology, University of Göttingen, Göttingen, Germany
| | - Michael Gale
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology, University of Oslo and Oslo University Hospital, Oslo, Norway
| | - Joshua T. Schiffer
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
- Division of Allergy and Infectious Disease, University of Washington, Seattle, Washington, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Judith M. White
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Microbiology, University of Virginia, Charlottesville, Virginia, USA
| | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
21
|
Rosenke K, Lewis MC, Feldmann F, Bohrnsen E, Schwarz B, Okumura A, Bohler WF, Callison J, Shaia C, Bosio CM, Lovaglio J, Saturday G, Jarvis MA, Feldmann H. Combined Molnupiravir and Nirmatrelvir Treatment Improves the Inhibitory Effect on SARS-CoV-2 in Rhesus Macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.09.03.506479. [PMID: 36263071 PMCID: PMC9580379 DOI: 10.1101/2022.09.03.506479] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The periodic emergence of SARS-CoV-2 variants of concern (VOCs) with unpredictable clinical severity and ability to escape preexisting immunity emphasizes the continued need for antiviral interventions. Two small molecule inhibitors, molnupiravir (MK-4482), a nucleoside analog, and nirmatrelvir (PF-07321332), a 3C-like protease inhibitor, have each recently been approved as monotherapy for use in high risk COVID-19 patients. As preclinical data are only available for rodent and ferret models, we originally assessed the efficacy of MK-4482 and PF-07321332 alone and then in combination Against infection with the SARS-CoV-2 Delta VOC in the rhesus macaque COVID-19 model. Notably, use of MK-4482 and PF-07321332 in combination improved the individual inhibitory effect of both drugs. Combined treatment resulted in milder disease progression, stronger reduction of virus shedding from mucosal tissues of the upper respiratory tract, stronger reduction of viral replication in the lower respiratory tract, and reduced lung pathology. Our data strongly indicate superiority of combined MK-4482 and PF-07321332 treatment of SARS-CoV-2 infections as demonstrated here in the closest COVID-19 surrogate model. One Sentence Summary The combination of molnupiravir and nirmatrelvir inhibits SARS-CoV-2 replication and shedding more effectively than individual treatments in the rhesus macaque model.
Collapse
Affiliation(s)
- Kyle Rosenke
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Matt C. Lewis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Friederike Feldmann
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Eric Bohrnsen
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Benjamin Schwarz
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Atsushi Okumura
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - W. Forrest Bohler
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Julie Callison
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Catharine M. Bosio
- Laboratory of Bacteriology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Michael A. Jarvis
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
- University of Plymouth, Plymouth, Devon, UK
- The Vaccine Group Ltd, Plymouth, Devon, UK
| | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| |
Collapse
|
22
|
Gidari A, Schiaroli E, Sabbatini S, Bastianelli S, Pierucci S, Busti C, Francisci D. Impact of SARS-CoV-2 Omicron Variants on Serum Neutralization in a Cohort of Healthcare Workers Vaccinated with BNT162b2. J Infect 2022; 85:e125-e126. [PMID: 36031155 PMCID: PMC9419996 DOI: 10.1016/j.jinf.2022.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/19/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Anna Gidari
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy.
| | - Elisabetta Schiaroli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Samuele Sabbatini
- Department of Medicine and Surgery, Medical Microbiology Section, University of Perugia, Perugia, Italy
| | - Sabrina Bastianelli
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Sara Pierucci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Chiara Busti
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| | - Daniela Francisci
- Department of Medicine and Surgery, Clinic of Infectious Diseases, "Santa Maria della Misericordia" Hospital, University of Perugia, Perugia, Italy
| |
Collapse
|