1
|
Lichtenegger S, Sonnleitner ST, Saiger S, Zauner A, Hardt M, Kleinhappl B, Wagner GE, Steinmetz I. Anti-RBD immunoglobulin levels and their predictive value for SARS-CoV-2 neutralization. Microbiol Spectr 2025; 13:e0214824. [PMID: 40197084 PMCID: PMC12054179 DOI: 10.1128/spectrum.02148-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
A large number of studies have demonstrated that anti-receptor-binding domain (RBD)-binding antibody titers correlate with SARS-CoV-2 neutralization and protection from the disease. Unlike live virus neutralization assays, antibody-binding assays are easier to perform, require lower biosafety levels, and have therefore served as a substitute for virus neutralization assays throughout the SARS-CoV-2 pandemic. Although anti-RBD antibodies are usually neutralizing, there is evidence that they can also be non-neutralizing. Moreover, different immunization regimens can vary in the induction of SARS-CoV-2-neutralizing antibodies. In this study, we hypothesized that sera from individuals with different immunization and infection histories, but with the same amount of anti-RBD total immunoglobulin, differ in their neutralizing potency. A total of 27 sera from SARS-CoV-2 convalescent, vaccinated individuals, and 27 vaccinated-only individuals were investigated by using a widely used antibody-binding assay (Elecsys anti-SARS-CoV-2 S enzyme-linked immunosorbent assay [ELISA]) and a live virus neutralization assay. As expected, anti-RBD immunoglobulin units correlated with virus neutralization capacity within the vaccine and hybrid immunized group. However, sera from both groups with matched anti-RBD units varied significantly in their neutralization potential. In detail, our data indicate a significantly higher neutralization potency of hybrid immunity compared to vaccinated-only sera with similar anti-RBD immunoglobulin levels. Our study highlights the need for cautious interpretation of quantitative antibody data from anti-RBD ELISAs, especially when comparing differently immunized groups. In other words, very similar anti-RBD levels can show very different functional activity. This finding has implications for determining possible future correlates of protection. IMPORTANCE Throughout the SARS-CoV-2 pandemic, neutralizing antibody levels have been central to predict a protective immune response. Anti-receptor-binding domain (RBD) enzyme-linked immunosorbent assays (ELISAs) correlate with neutralization assays and are due to the integration of simple performance with timely results used as surrogate assays. However, previous studies determining correlation used homogeneous cohorts. We reevaluated the correlation of a frequently used anti-RBD ELISA and a live virus neutralization assay using a heterogeneous cohort consisting of a vaccinated group without prior SARS-CoV-2 infection and a vaccinated convalescent group. The neutralizing capacity of sera with matched anti-RBD units significantly differed between groups, decreasing the correlation of the assays. Our findings highlight the necessity of considering the immunization context when interpreting serological tests and suggest that different immunization groups may require distinct protective thresholds. Considering the immunization history, we can develop more accurate predictions of immunity not only for SARS-CoV-2 but also for future challenges.
Collapse
Affiliation(s)
- Sabine Lichtenegger
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Sissy Therese Sonnleitner
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Sabine Saiger
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Andrea Zauner
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Melina Hardt
- Diagnostic & Research Institute of Pathology, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Barbara Kleinhappl
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Gabriel E. Wagner
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| | - Ivo Steinmetz
- Diagnostic & Research Institute of Hygiene, Microbiology and Environmental Medicine, Diagnostic and Research Center for Molecular Biomedicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
2
|
Vukovikj M, Melidou A, Nannapaneni P, Normark T, Kraus A, Broberg EK. Impact of SARS-CoV-2 variant mutations on susceptibility to monoclonal antibodies and antiviral drugs: a non-systematic review, April 2022 to October 2024. Euro Surveill 2025; 30:2400252. [PMID: 40084420 PMCID: PMC11912142 DOI: 10.2807/1560-7917.es.2025.30.10.2400252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 12/10/2024] [Indexed: 03/16/2025] Open
Abstract
BackgroundMonoclonal antibodies (mAbs) and antiviral drugs have emerged as additional tools for treatment of COVID-19.AimWe aimed to review data on susceptibility of 14 SARS-CoV-2 variants to mAbs and antiviral drugs authorised in the European Union/European Economic Area (EU/EEA) countries.MethodsWe constructed a literature review compiling 298 publications from four databases: PubMed, Science Direct, LitCovid and BioRxiv/MedRxiv preprint servers. We included publications on nirmatrelvir and ritonavir, remdesivir and tixagevimab and cilgavimab, regdanvimab, casirivimab and imdevimab, and sotrovimab approved by the European Medicines Agency (EMA) by 1 October 2024.ResultsThe mutations identified in the open reading frame (ORF)1ab, specifically nsp5:H172Y, nsp5:H172Y and Q189E, nsp5:L50F and E166V and nsp5:L50F, E166A and L167V, led to a decrease in susceptibility to nirmatrelvir and ritonavir, ranging from moderate (25-99) to high reductions (> 100). Casirivimab and imdevimab exhibited highly reduced neutralisation capacity across all Omicron sub-lineages. Sub-lineages BA.1, BA.2 and BA.5 had decreased susceptibility to regdanvimab, while sotrovimab showed decreased efficacy for BA.2, BA.4, BQ.1.1 and BA.2.86. Tixagevimab and cilgavimab exhibited highly reduced neutralisation activity against BQ.1, BQ.1.1, XBB, XBB.1.5 and BA.2.86 sub-lineages.ConclusionsThe emergence of new variants, some with altered antigenic characteristics, may lead to resistance against mAbs and/or antiviral drugs and evasion of immunity induced naturally or by vaccination. This summary of mutations, combination of mutations and SARS-CoV-2 variants linked to reduced susceptibility to mAbs and antiviral drugs, should aid the selection of appropriate treatment strategies and/or phasing out therapies that have lost their effectiveness.
Collapse
Affiliation(s)
- Maja Vukovikj
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - Angeliki Melidou
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | | | - Tanja Normark
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - Annette Kraus
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| | - Eeva K Broberg
- European Centre for Disease Prevention and Control (ECDC), Stockholm, Sweden
| |
Collapse
|
3
|
Guan M, Zuo Y, Yuan Y, Zhu H, Zheng H. High-dose Vitamin C intake and COVID-19 related symptoms during the SARS-CoV-2 pandemic. Am J Med Sci 2024; 368:265-268. [PMID: 38788926 DOI: 10.1016/j.amjms.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 04/25/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024]
Affiliation(s)
- Mingcheng Guan
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Yibo Zuo
- Department / Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, PR China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, PR China
| | - Yukang Yuan
- Department / Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, PR China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, PR China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou, PR China
| | - Hui Zheng
- Department / Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, PR China; International Institute of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, PR China.
| |
Collapse
|
4
|
Cui Z, Wang H, Zou H, Li L, Zhang Y, Chen W. Efficacy and safety of casirivimab and imdevimab for preventing and treating COVID-19: a systematic review and meta-analysis. J Thorac Dis 2024; 16:3606-3622. [PMID: 38983147 PMCID: PMC11228754 DOI: 10.21037/jtd-23-1604] [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: 10/17/2023] [Accepted: 04/26/2024] [Indexed: 07/11/2024]
Abstract
Background The ongoing global epidemic of coronavirus disease 2019 (COVID-19) has created a serious public health problem. The selection of safe and effective therapeutic agents is of paramount importance. This systematic review aims to evaluate the efficacy and safety of the combination of casirivimab and imdevimab in the treatment of global cases of COVID-19. Methods To identify randomized controlled trials (RCTs) investigating the combined administration of casirivimab and imdevimab for COVID-19 management, a comprehensive search was conducted across multiple databases including PubMed, Web of Science, Embase, and the Cochrane Library from their inception to September 10, 2022. Data on the efficacy and safety of casirivimab and imdevimab were extracted. Subgroup analyses and sensitivity analyses were performed. Results A total of 851 articles were searched. Twelve studies were finally included in the meta-analysis, with 27,179 participants. Dichotomous and continuous variables were presented as odds ratios (ORs) and weighted mean differences (WMDs) with their 95% confidence intervals (CIs), respectively. Compared to placebo or alternative medications, the combination of casirivimab and imdevimab reduced viral load (WMD: -0.73, 95% CI: -1.09 to -0.38, P<0.01), all-cause mortality (OR =0.90, 95% CI: 0.82-0.99, P=0.03), the incidence of any serious adverse events (OR =0.80, 95% CI: 0.67-0.95, P=0.01), the incidence of Grade 3 or more severe adverse events (OR =0.76, 95% CI: 0.62-0.92, P=0.01), the likelihood of contracting COVID-19, the incidence of hospitalization, emergency room visits, and mortality (OR =0.54, 95% CI: 0.32-0.93, P=0.03). Conclusions The monoclonal antibody combination of casirivimab and imdevimab is effective in treating patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as they can reduce viral load, all-cause mortality, infection rates, and the incidence of clinical outcomes of special interest after treatment, while maintaining a favorable safety profile.
Collapse
Affiliation(s)
- Zhifang Cui
- Department of Respiratory Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Hongwu Wang
- Department of Respiratory Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Heng Zou
- Department of Respiratory Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Li
- Department of Respiratory Medicine, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ye Zhang
- Department of General Medicine, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Wenyu Chen
- Department of Respiratory Medicine, Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
5
|
Esmaeili S, Owens K, Wagoner J, Polyak SJ, White JM, Schiffer JT. A unifying model to explain frequent SARS-CoV-2 rebound after nirmatrelvir treatment and limited prophylactic efficacy. Nat Commun 2024; 15:5478. [PMID: 38942778 PMCID: PMC11213957 DOI: 10.1038/s41467-024-49458-9] [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: 02/06/2024] [Accepted: 06/04/2024] [Indexed: 06/30/2024] Open
Abstract
In a pivotal trial (EPIC-HR), a 5-day course of oral ritonavir-boosted nirmatrelvir, given early during symptomatic SARS-CoV-2 infection (within three days of symptoms onset), decreased hospitalization and death by 89.1% and nasal viral load by 0.87 log relative to placebo in high-risk individuals. Yet, nirmatrelvir/ritonavir failed as post-exposure prophylaxis in a trial, and frequent viral rebound has been observed in subsequent cohorts. We develop a mathematical model capturing viral-immune dynamics and nirmatrelvir pharmacokinetics that recapitulates viral loads from this and another clinical trial (PLATCOV). Our results suggest that nirmatrelvir's in vivo potency is significantly lower than in vitro assays predict. According to our model, a maximally potent agent would reduce the viral load by approximately 3.5 logs relative to placebo at 5 days. The model identifies that earlier initiation and shorter treatment duration are key predictors of post-treatment rebound. Extension of treatment to 10 days for Omicron variant infection in vaccinated individuals, rather than increasing dose or dosing frequency, is predicted to lower the incidence of viral rebound significantly.
Collapse
Affiliation(s)
- Shadisadat Esmaeili
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| | - Katherine Owens
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jessica Wagoner
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Stephen J Polyak
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA, USA
| | - Judith M White
- Department of Cell Biology, University of Virginia, Charlottesville, VA, USA
| | - Joshua T Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
6
|
Sam KS, Khosla P, Taneja V, Dessai R. Casirivimab-imdevimab monoclonal antibody treatment for an immunocompromised patient with persistent SARS-CoV-2 infection: a case report. COMMUNICATIONS MEDICINE 2024; 4:103. [PMID: 38824216 PMCID: PMC11144204 DOI: 10.1038/s43856-024-00523-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/08/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Persistent acute SARS-CoV- 2 infection is characterised by the persistence of symptoms of a COVID-19 illness and a persistently positive SARS-CoV-2 RT PCR result. It is often seen in immunocompromised individuals. Currently, there are no approved treatment regimens or guidelines for management. METHODS Our patient is a middle-aged male who had received chemotherapy prior to the onset of his SARS- CoV-2 infection and subsequently was diagnosed with a persistent and acute SARS- CoV-2 infection after presenting to us with a pyrexia of unknown origin. He was treated on an off-label basis following informed consent with casirivimab-imdevimab monoclonal antibody therapy, comprising two IgG1 neutralising human monoclonal antibodies at a dose of 600 mg each. RESULTS Our patient had significant clinical improvement on treatment with the monoclonal antibody therapy and it was well tolerated without any adverse effects. He is currently doing well during follow up. CONCLUSIONS Casirivimab-imdevimab monoclonal antibody therapy could be beneficial for people with persistent acute SARS-CoV-2 infection. Therapy is warranted on a case-to-case basis. This highlights the need to identify immunocompromised individuals who are at risk of developing persistent acute SARS-CoV-2 infection to enable their prompt treatment.
Collapse
Affiliation(s)
| | - Pooja Khosla
- Department of internal Medicine, Sir Ganga Ram Hospital, New Delhi, India
| | - Vinus Taneja
- Department of internal Medicine, Sir Ganga Ram Hospital, New Delhi, India.
| | - Rishikesh Dessai
- Department of internal Medicine, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
7
|
Mayer BT, Zhang L, deCamp AC, Yu C, Sato A, Angier H, Seaton KE, Yates N, Ledgerwood JE, Mayer K, Caskey M, Nussenzweig M, Stephenson K, Julg B, Barouch DH, Sobieszczyk ME, Edupuganti S, Kelley CF, McElrath MJ, Gelderblom HC, Pensiero M, McDermott A, Gama L, Koup RA, Gilbert PB, Cohen MS, Corey L, Hyrien O, Tomaras GD, Huang Y. Impact of LS Mutation on Pharmacokinetics of Preventive HIV Broadly Neutralizing Monoclonal Antibodies: A Cross-Protocol Analysis of 16 Clinical Trials in People without HIV. Pharmaceutics 2024; 16:594. [PMID: 38794258 PMCID: PMC11125931 DOI: 10.3390/pharmaceutics16050594] [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: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 05/26/2024] Open
Abstract
Monoclonal antibodies are commonly engineered with an introduction of Met428Leu and Asn434Ser, known as the LS mutation, in the fragment crystallizable region to improve pharmacokinetic profiles. The LS mutation delays antibody clearance by enhancing binding affinity to the neonatal fragment crystallizable receptor found on endothelial cells. To characterize the LS mutation for monoclonal antibodies targeting HIV, we compared pharmacokinetic parameters between parental versus LS variants for five pairs of anti-HIV immunoglobin G1 monoclonal antibodies (VRC01/LS/VRC07-523LS, 3BNC117/LS, PGDM1400/LS PGT121/LS, 10-1074/LS), analyzing data from 16 clinical trials of 583 participants without HIV. We described serum concentrations of these monoclonal antibodies following intravenous or subcutaneous administration by an open two-compartment disposition, with first-order elimination from the central compartment using non-linear mixed effects pharmacokinetic models. We compared estimated pharmacokinetic parameters using the targeted maximum likelihood estimation method, accounting for participant differences. We observed lower clearance rate, central volume, and peripheral volume of distribution for all LS variants compared to parental monoclonal antibodies. LS monoclonal antibodies showed several improvements in pharmacokinetic parameters, including increases in the elimination half-life by 2.7- to 4.1-fold, the dose-normalized area-under-the-curve by 4.1- to 9.5-fold, and the predicted concentration at 4 weeks post-administration by 3.4- to 7.6-fold. Results suggest a favorable pharmacokinetic profile of LS variants regardless of HIV epitope specificity. Insights support lower dosages and/or less frequent dosing of LS variants to achieve similar levels of antibody exposure in future clinical applications.
Collapse
Affiliation(s)
- Bryan T. Mayer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Lily Zhang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Allan C. deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Chenchen Yu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Alicia Sato
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Heather Angier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Kelly E. Seaton
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Nicole Yates
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Julie E. Ledgerwood
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | | | - Marina Caskey
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; (M.C.); (M.N.)
| | - Michel Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10065, USA; (M.C.); (M.N.)
| | - Kathryn Stephenson
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA 02139, USA; (K.S.); (B.J.)
| | - Boris Julg
- Ragon Institute of Mass General, MIT and Harvard, Cambridge, MA 02139, USA; (K.S.); (B.J.)
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA;
| | | | - Srilatha Edupuganti
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.E.); (C.F.K.)
| | - Colleen F. Kelley
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA; (S.E.); (C.F.K.)
| | - M. Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Huub C. Gelderblom
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Michael Pensiero
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Adrian McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Lucio Gama
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Richard A. Koup
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, Bethesda, MD 20892, USA (M.P.); (A.M.); (L.G.); (R.A.K.)
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Myron S. Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA;
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Departments of Medicine and Laboratory Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
| | - Georgia D. Tomaras
- Duke University Medical Center, Durham, NC 27705, USA; (K.E.S.); (N.Y.); (G.D.T.)
| | - Yunda Huang
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; (L.Z.); (A.C.d.); (C.Y.); (A.S.); (H.A.); (M.J.M.); (H.C.G.); (P.B.G.); (L.C.); (O.H.)
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
8
|
Ghasemiyeh P, Mohammadi-Samani S. Lessons we learned during the past four challenging years in the COVID-19 era: pharmacotherapy, long COVID complications, and vaccine development. Virol J 2024; 21:98. [PMID: 38671455 PMCID: PMC11055380 DOI: 10.1186/s12985-024-02370-6] [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: 01/30/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
About four years have passed since the detection of the first cases of COVID-19 in China. During this lethal pandemic, millions of people have lost their lives around the world. Since the first waves of COVID-19 infection, various pharmacotherapeutic agents have been examined in the management of COVID-19. Despite all these efforts in pharmacotherapy, drug repurposing, and design and development of new drugs, multiple organ involvement and various complications occurred during COVID-19. Some of these complications became chronic and long-lasting which led to the "long COVID" syndrome appearance. Therefore, the best way to eradicate this pandemic is prophylaxis through mass vaccination. In this regard, various vaccine platforms including inactivated vaccines, nucleic acid-based vaccines (mRNA and DNA vaccines), adenovirus-vectored vaccines, and protein-based subunit vaccines have been designed and developed to prevent or reduce COVID-19 infection, hospitalization, and mortality rates. In this focused review, at first, the most commonly reported clinical presentations of COVID-19 during these four years have been summarized. In addition, different therapeutic regimens and their latest status in COVID-19 management have been listed. Furthermore, the "long COVID" and related signs, symptoms, and complications have been mentioned. At the end, the effectiveness of available COVID-19 vaccines with different platforms against early SARS-CoV-2 variants and currently circulating variants of interest (VOI) and the necessity of booster vaccine shots have been summarized and discussed in more detail.
Collapse
Affiliation(s)
- Parisa Ghasemiyeh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Pharmacy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soliman Mohammadi-Samani
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
9
|
Iketani S, Ho DD. SARS-CoV-2 resistance to monoclonal antibodies and small-molecule drugs. Cell Chem Biol 2024; 31:632-657. [PMID: 38640902 PMCID: PMC11084874 DOI: 10.1016/j.chembiol.2024.03.008] [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: 09/07/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Over four years have passed since the beginning of the COVID-19 pandemic. The scientific response has been rapid and effective, with many therapeutic monoclonal antibodies and small molecules developed for clinical use. However, given the ability for viruses to become resistant to antivirals, it is perhaps no surprise that the field has identified resistance to nearly all of these compounds. Here, we provide a comprehensive review of the resistance profile for each of these therapeutics. We hope that this resource provides an atlas for mutations to be aware of for each agent, particularly as a springboard for considerations for the next generation of antivirals. Finally, we discuss the outlook and thoughts for moving forward in how we continue to manage this, and the next, pandemic.
Collapse
Affiliation(s)
- Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
10
|
Esmaeili S, Owens K, Wagoner J, Polyak SJ, White JM, Schiffer JT. A unifying model to explain high nirmatrelvir therapeutic efficacy against SARS-CoV-2, despite low post-exposure prophylaxis efficacy and frequent viral rebound. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.23.23294505. [PMID: 38352583 PMCID: PMC10862980 DOI: 10.1101/2023.08.23.23294505] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
In a pivotal trial (EPIC-HR), a 5-day course of oral ritonavir-boosted nirmatrelvir, given early during symptomatic SARS-CoV-2 infection (within three days of symptoms onset), decreased hospitalization and death by 89.1% and nasal viral load by 0.87 log relative to placebo in high-risk individuals. Yet, nirmatrelvir/ritonavir failed as post-exposure prophylaxis in a trial, and frequent viral rebound has been observed in subsequent cohorts. We developed a mathematical model capturing viral-immune dynamics and nirmatrelvir pharmacokinetics that recapitulated viral loads from this and another clinical trial (PLATCOV). Our results suggest that nirmatrelvir's in vivo potency is significantly lower than in vitro assays predict. According to our model, a maximally potent agent would reduce the viral load by approximately 3.5 logs relative to placebo at 5 days. The model identifies that earlier initiation and shorter treatment duration are key predictors of post-treatment rebound. Extension of treatment to 10 days for Omicron variant infection in vaccinated individuals, rather than increasing dose or dosing frequency, is predicted to lower the incidence of viral rebound significantly.
Collapse
Affiliation(s)
- Shadisadat Esmaeili
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, USA
| | - Katherine Owens
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, USA
| | - Jessica Wagoner
- Department of Medicine, University of Washington; Seattle, WA, USA
| | | | - Judith M. White
- Department of Cell Biology, University of Virginia; Charlottesville, VA, USA
| | - Joshua T. Schiffer
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center; Seattle, WA, USA
- Department of Medicine, University of Washington; Seattle, WA, USA
| |
Collapse
|
11
|
Huygens S, Preijers T, Swaneveld FH, Kleine Budde I, GeurtsvanKessel CH, Koch BCP, Rijnders BJA. Dosing of Convalescent Plasma and Hyperimmune Anti-SARS-CoV-2 Immunoglobulins: A Phase I/II Dose-Finding Study. Clin Pharmacokinet 2024; 63:497-509. [PMID: 38427270 PMCID: PMC11052786 DOI: 10.1007/s40262-024-01351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND AND OBJECTIVE During the COVID-19 pandemic, trials on convalescent plasma (ConvP) were performed without preceding dose-finding studies. This study aimed to assess potential protective dosing regimens by constructing a population pharmacokinetic (popPK) model describing anti-SARS-CoV-2 antibody titers following the administration of ConvP or hyperimmune globulins (COVIg). METHODS Immunocompromised patients, testing negative for anti-SARS-CoV-2 spike antibodies despite vaccination, received a range of anti-SARS-CoV-2 antibodies in the form of COVIg or ConvP infusion. The popPK analysis was performed using NONMEM v7.4. Monte Carlo simulations were performed to assess potential COVIg and ConvP dosing regimens for prevention of COVID-19. RESULTS Forty-four patients were enrolled, and data from 42 were used for constructing the popPK model. A two-compartment elimination model with mixed residual error best described the Nab-titers after administration. Inter-individual variation was associated to CL (44.3%), V1 (27.3%), and V2 (29.2%). Lean body weight and type of treatment (ConvP/COVIg) were associated with V1 and V2, respectively. Median elimination half-life was 20 days (interquartile range: 17-25 days). Simulations demonstrated that even monthly infusions of 600 mL of the ConvP or COVIg used in this trial would not achieve potentially protective serum antibody titers for > 90% of the time. However, as a result of hybrid immunity and/or repeated vaccination, plasma donors with extremely high antibody titers are now readily available, and a > 90% target attainment should be possible. CONCLUSION The results of this study may inform future intervention studies on the prophylactic and therapeutic use of antiviral antibodies in the form of ConvP or COVIg. CLINICAL TRIAL REGISTRATION NUMBER NL9379 (The Netherlands Trial Register).
Collapse
Affiliation(s)
- Sammy Huygens
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Tim Preijers
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Francis H Swaneveld
- Unit of Transfusion Medicine, Sanquin Blood Supply Foundation, 1066 CX, Amsterdam, The Netherlands
| | - Ilona Kleine Budde
- Clinical Operations, Prothya Biosolutions, 1066 CX, Amsterdam, The Netherlands
| | - Corine H GeurtsvanKessel
- Department of Viroscience, Erasmus University Medical Center Rotterdam, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Zhang H, Zhou J, Chen H, Mao J, Tang Y, Yan W, Zhang T, Li C, Chen S, Li G, Zhang G, Ding Y, Liu L. Phase I study, and dosing regimen selection for a pivotal COVID-19 trial of GST-HG171. Antimicrob Agents Chemother 2024; 68:e0111523. [PMID: 38099673 PMCID: PMC10777829 DOI: 10.1128/aac.01115-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/28/2023] [Indexed: 01/11/2024] Open
Abstract
This study is aimed to evaluate the safety, tolerability, and pharmacokinetics (PK), as well as to select an appropriate dosing regimen for the pivotal clinical trial of GST-HG171, an orally bioavailable, potent, and selective 3CL protease inhibitor by a randomized, double-blind, and placebo-controlled phase I trial in healthy subjects. We conducted a Ph1 study involving 78 healthy subjects to assess the safety, tolerability, and PK of single ascending doses (150-900 mg) as well as multiple ascending doses (MADs) (150 and 300 mg) of GST-HG171. Additionally, we examined the food effect and drug-drug interaction of GST-HG171 in combination with ritonavir through a MAD regimen of GST-HG171/ritonavir (BID or TID) for 5 days. Throughout the course of these studies, no serious AEs or deaths occurred, and no AEs necessitated study discontinuation. We observed that food had no significant impact on the exposure of GST-HG171. However, the presence of ritonavir substantially increased the exposure of GST-HG171, which facilitated the selection of the GST-HG171/ritonavir dose and regimen (150/100 mg BID) for subsequent phase II/III trials. The selected dose regimen was achieved through concentrations continuously at 6.2-9.9-fold above the levels required for protein-binding adjusted 50% inhibition (IC50) of viral replication in vitro. The combination of 150 mg GST-HG171/100 mg ritonavir demonstrated favorable safety and tolerability profiles. The PK data obtained from GST-HG171/ritonavir administration guided the selection of appropriate dose for a pivotal phase II/III trial currently in progress. (This study has been registered at ClinicalTrials.gov under identifier NCT05668897).
Collapse
Affiliation(s)
- Hong Zhang
- 1 Phase I Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Zhou
- 1 Phase I Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hong Chen
- 1 Phase I Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - John Mao
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Yanan Tang
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Wenhao Yan
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Tianxiang Zhang
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Chuanjing Li
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Shikui Chen
- Fujian Cosunter Pharmaceutical Co., Ltd., Fuzhou, Fujian, China
| | - Guoping Li
- Fujian Cosunter Pharmaceutical Co., Ltd., Fuzhou, Fujian, China
| | - George Zhang
- Fujian Akeylink Biotechnology Co., Ltd., Fuzhou, Fujian, China
| | - Yanhua Ding
- 1 Phase I Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Li Liu
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
13
|
Krammer F. The role of vaccines in the COVID-19 pandemic: what have we learned? Semin Immunopathol 2024; 45:451-468. [PMID: 37436465 PMCID: PMC11136744 DOI: 10.1007/s00281-023-00996-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/24/2023] [Indexed: 07/13/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged late in 2019 and caused the coronavirus disease 2019 (COVID-19) pandemic that has so far claimed approximately 20 million lives. Vaccines were developed quickly, became available in the end of 2020, and had a tremendous impact on protection from SARS-CoV-2 mortality but with emerging variants the impact on morbidity was diminished. Here I review what we learned from COVID-19 from a vaccinologist's perspective.
Collapse
Affiliation(s)
- Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Wicaksono IA, Suhandi C, Elamin KM, Wathoni N. Efficacy and safety of casirivimab-imdevimab combination on COVID-19 patients: A systematic review and meta-analysis randomized controlled trial. Heliyon 2023; 9:e22839. [PMID: 38058433 PMCID: PMC10696184 DOI: 10.1016/j.heliyon.2023.e22839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 11/12/2023] [Accepted: 11/20/2023] [Indexed: 12/08/2023] Open
Abstract
Background The advantages and disadvantages of casirivimab-imdevimab for coronavirus disease 2019 are not well understood. We conducted a systematic review and meta-analysis of relevant literature to determine the therapeutic effectiveness and potential side effects of casirivimab-imdevimab in COVID-19 patients. Methods Databases were searched from the time of their commencement until February 28th, 2023. The primary results evaluated were the death rate at 28 days, progression of current clinical symptoms within 28 days, viral load, discharge from hospital, and any adverse events. Also, we contrasted the effects of the casirivimab-imdevimab treatment with placebo or standard of care. The protocol registration for this systematic review and meta-analysis was recorded in the PROSPERO database (CRD42023412835). Results A total of eight studies were included, comprising 19,819 patients, and conducted a qualitative assessment of their risk of bias using the Cochrane risk of bias tool. Casirivimab-imdevimab effectively reduced the mortality rate (OR = 0.62; 95 % CI of 0.40-0.98; p = 0.04; I2 = 30 %) and reduced the progression of clinical symptoms (OR = 0.86; 95 % CI of 0.79-0.93; p = 0.0003; I2 = 57 %). Casirivimab-imdevimab also improved viral load clearance and hospital discharge. Additionally, the trials' findings demonstrated a slight decrease in the likelihood of adverse events occurring with the use of casirivimab-imdevimab. Conclusion Our research suggests that casirivimab-imdevimab may be a valuable, safe, and effective anti-SARS-CoV-2 regimen.
Collapse
Affiliation(s)
- Imam Adi Wicaksono
- Department of Pharmacology and Clinical Pharmacy, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
| | - Cecep Suhandi
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
| | - Khaled M. Elamin
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Universitas Padjadjaran, Sumedang 45363, West Java, Indonesia
| |
Collapse
|
15
|
Xu M, O’Brien MP, Hooper AT, Forleo-Neto E, Isa F, Hou P, Chan KC, Cohen MS, Marovich MA, Hamilton JD, Hirshberg B, Herman GA, Musser BJ. Nasopharyngeal Viral Load Is the Major Driver of Incident Antibody Immune Response to SARS-CoV-2 Infection. Open Forum Infect Dis 2023; 10:ofad598. [PMID: 38111750 PMCID: PMC10727195 DOI: 10.1093/ofid/ofad598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/29/2023] [Indexed: 12/20/2023] Open
Abstract
Background Virologic determinants of seroconversion to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection were defined in a post hoc analysis of prospectively studied vaccine- and infection-naïve individuals at high risk for coronavirus disease 2019 (COVID-19). Methods This phase 3 COVID-19 prevention trial (NCT04452318) with casirivimab and imdevimab was conducted in July 2020-February 2021, before widespread vaccine availability. Placebo-treated participants who were uninfected (SARS-CoV-2 quantitative reverse transcription polymerase chain reaction [RT-qPCR] negative) and seronegative were assessed weekly for 28 days (efficacy assessment period [EAP]) for COVID-19 symptoms and SARS-CoV-2 infection by RT-qPCR of nasopharyngeal swab samples and for serostatus by antinucleocapsid immunoglobulin (Ig) G. Regression-based modeling, including causal mediation analysis, estimated the effects of viral load on seroconversion. Results Of 157/1069 (14.7%) uninfected and seronegative (for antispike IgG, antispike IgA, and antinucleocapsid IgG) participants who became infected during the EAP, 105 (65%) seroconverted. The mean (SD) maximum viral load of seroconverters was 7.23 (1.68) log10 copies/mL vs 4.8 (2.2) log10 copies/mL in those who remained seronegative; viral loads of ∼6.0 log10 copies/mL better predicted seroconversion. The mean of the maximum viral load was 7.11 log10 copies/mL in symptomatic participants vs 5.58 log10 copies/mL in asymptomatic participants. The mean duration of detectable viral load was longer in seroconverted vs seronegative participants: 3.24 vs 1.63 weeks. Conclusions Maximum SARS-CoV-2 viral load is a major driver of seroconversion and symptomatic COVID-19, with high viral loads (∼6.0 log10 copies/mL) better predicting seroconversion. Serology underestimates infection rates, incidence, and prevalence of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Meng Xu
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | | | | | | | - Flonza Isa
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Peijie Hou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Kuo-Chen Chan
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Myron S Cohen
- University of North Carolina Chapel Hill School of Medicine, Institute for Global Health and Infectious Diseases, Chapel Hill, North Carolina, USA
| | - Mary A Marovich
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | - Boaz Hirshberg
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Gary A Herman
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| | - Bret J Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, NewYork, USA
| |
Collapse
|
16
|
Lee JY, Bu SH, Song E, Cho S, Yu S, Kim J, Kym S, Seo KW, Kwon KT, Kim JY, Kim S, Ahn K, Jung N, Lee Y, Jung Y, Hwang C, Park SW. Safety and Effectiveness of Regdanvimab for COVID-19 Treatment: A Phase 4 Post-marketing Surveillance Study Conducted in South Korea. Infect Dis Ther 2023; 12:2417-2435. [PMID: 37833467 PMCID: PMC10600078 DOI: 10.1007/s40121-023-00859-1] [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: 06/26/2023] [Accepted: 08/15/2023] [Indexed: 10/15/2023] Open
Abstract
INTRODUCTION Regdanvimab, a neutralising monoclonal antibody (mAb) against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), received approval for the treatment of coronavirus disease 2019 (COVID-19) in South Korea in 2021. The Ministry of Food and Drug Safety in South Korea mandate that new medications be re-examined for safety and effectiveness post-approval in at least 3000 individuals. This post-marketing surveillance (PMS) study was used to evaluate the safety and effectiveness of regdanvimab in real-world clinical care. METHODS This prospective, multicentre, phase 4 PMS study was conducted between February 2021 and March 2022 in South Korea. Eligible patients were aged ≥ 18 years with confirmed mild COVID-19 at high risk of disease progression or moderate COVID-19. Patients were hospitalised and treated with regdanvimab (40 mg/kg, day 1) and then monitored until discharge, with a follow-up call on day 28. Adverse events (AEs) were documented, and the COVID-19 disease progression rate was used to measure effectiveness. RESULTS Of the 3123 patients with COVID-19 infection identified, 3036 were eligible for inclusion. Approximately 80% and 5% of the eligible patients were diagnosed with COVID-19 during the delta- and omicron-dominant periods, respectively. Median (range) age was 57 (18-95) years, and 50.6% of patients were male. COVID-19 severity was assessed before treatment, and high-risk mild and moderate COVID-19 was diagnosed in 1030 (33.9%) and 2006 (66.1%) patients, respectively. AEs and adverse drug reactions (ADRs) were experienced by 684 (22.5%) and 363 (12.0%) patients, respectively. The most common ADR was increased liver function test (n = 62, 2.0%). Nine (0.3%) patients discontinued regdanvimab due to ADRs. Overall, 378 (12.5%) patients experienced disease progression after regdanvimab infusion, with extended hospitalisation/re-admission (n = 300, 9.9%) as the most common reason. Supplemental oxygen was required by 282 (9.3%) patients. Ten (0.3%) patients required intensive care monitoring and 3 (0.1%) died due to COVID-19. CONCLUSION This large-scale PMS study demonstrated that regdanvimab was effective against COVID-19 progression and had an acceptable safety profile when used in real-world clinical practice.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Keimyung University Daegu Dongsan Hospital, Daegu, Republic of Korea
| | - Seon Hee Bu
- Seoul Metropolitan City Bukbu Hospital, Seoul, Republic of Korea
| | - EunHyang Song
- Seoul Metropolitan City Seobuk Hospital, Seoul, Republic of Korea
| | | | - Sungbong Yu
- Bagae General Hospital, Pyeongtaek, Republic of Korea
| | - Jungok Kim
- Chungnam National University Sejong Hospital, Sejong, Republic of Korea
| | - Sungmin Kym
- Chungnam National University Sejong Hospital, Sejong, Republic of Korea
| | - Kwang Won Seo
- Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Republic of Korea
| | - Ki Tae Kwon
- School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jin Yong Kim
- Incheon Medical Centre, Incheon, Republic of Korea
| | | | | | | | - Yeonmi Lee
- Celltrion, Inc., Incheon, Republic of Korea
| | | | | | - Sang Won Park
- Department of Internal Medicine, Seoul National University Boramae Medical Centre, 20 Boramae-ro 5-gil, Dongjak-gu, Seoul, 07061, Republic of Korea.
| |
Collapse
|
17
|
Murayama A, Kugo H, Saito Y, Saito H, Tanimoto T, Ozaki A. A 9-Year Investigation of Healthcare Industry Payments to Pulmonologists in the United States. Ann Am Thorac Soc 2023; 20:1283-1292. [PMID: 36961514 DOI: 10.1513/annalsats.202209-827oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 03/24/2023] [Indexed: 03/25/2023] Open
Abstract
Rationale: The healthcare industry sometimes makes payments to physicians for nonresearch and research purposes in the United States. Objectives: We aimed to evaluate the trends in nonresearch and research industry payments to pulmonologists since the inception of the Open Payments database in 2013. Methods: Using the Open Payments database between August 2013 and December 2021, this population-based observational cohort study examined nonresearch and research payments made by the healthcare industry to pulmonologists registered in the National Plan and Provider Enumeration System in the United States. We performed descriptive analyses on payment data and generalized estimating equations for payment trends. Results: Of 12,488 active pulmonologists, 11,074 (88.7%) accepted a total of 2,246,412 payments totaling $1,053,344,669. Total payments were $253,405,965 (24.1%) in nonresearch, $17,382,904 (1.7%) in direct research, and $782,555,800 (74.3%) in associated research payments between 2013 and 2021. Median per-physician payments (interquartile range) were $2,342 ($496 to $8,299) for nonresearch, $4,688 ($1,435 to $21,803) for direct research, and $95,927 ($20,300 to $344,995) for associated research payments. The top 1%, 5%, and 10% of pulmonologists accepted 37.3%, 71.9%, and 83.7% of the total nonresearch payments. The per-physician nonresearch payments increased by 2.9% (95% confidence interval [CI], 1.2 to 4.7; P = 0.001) annually between 2014 and 2019 and decreased by 50.2% (95% CI, -55.3 to -44.6; P < 0.001) in 2020, whereas there was no yearly change in research payments. Conclusions: Nearly 90% of pulmonologists received nonresearch and research payments from the healthcare industry in the United States. Nonresearch payments have been increasing since the inception of the Open Payments database.
Collapse
Affiliation(s)
- Anju Murayama
- School of Medicine, Tohoku University, Sendai, Japan
| | - Hinari Kugo
- School of Medicine, Tohoku University, Sendai, Japan
| | - Yoshika Saito
- Medical Governance Research Institute, Minato-ku, Japan
- Faculty of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroaki Saito
- Medical Governance Research Institute, Minato-ku, Japan
- Department of Internal Medicine, Soma Central Hospital, Soma, Japan
| | - Tetsuya Tanimoto
- Medical Governance Research Institute, Minato-ku, Japan
- Department of Internal Medicine, Navitas Clinic Tachikawa, Tachikawa, Japan; and
| | - Akihiko Ozaki
- Medical Governance Research Institute, Minato-ku, Japan
- Department of Breast and Thyroid Surgery, Jyoban Hospital, Iwaki, Japan
| |
Collapse
|
18
|
Stadler E, Burgess MT, Schlub TE, Khan SR, Chai KL, McQuilten ZK, Wood EM, Polizzotto MN, Kent SJ, Cromer D, Davenport MP, Khoury DS. Monoclonal antibody levels and protection from COVID-19. Nat Commun 2023; 14:4545. [PMID: 37507368 PMCID: PMC10382502 DOI: 10.1038/s41467-023-40204-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Multiple monoclonal antibodies have been shown to be effective for both prophylaxis and therapy for SARS-CoV-2 infection. Here we aggregate data from randomized controlled trials assessing the use of monoclonal antibodies (mAb) in preventing symptomatic SARS-CoV-2 infection. We use data on the in vivo concentration of mAb and the associated protection from COVID-19 over time to model the dose-response relationship of mAb for prophylaxis. We estimate that 50% protection from COVID-19 is achieved with a mAb concentration of 96-fold of the in vitro IC50 (95% CI: 32-285). This relationship provides a tool for predicting the prophylactic efficacy of new mAb and against SARS-CoV-2 variants. Finally, we compare the relationship between neutralization titer and protection from COVID-19 after either mAb treatment or vaccination. We find no significant difference between the 50% protective titer for mAb and vaccination, although sample sizes limited the power to detect a difference.
Collapse
Affiliation(s)
- Eva Stadler
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Martin T Burgess
- School of Mathematics and Statistics, University of New South Wales, Sydney, NSW, Australia
| | - Timothy E Schlub
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Shanchita R Khan
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Zoe K McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Clinical Haematology, Monash Health, Clayton, VIC, Australia
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
- Department of Clinical Haematology, Monash Health, Clayton, VIC, Australia
| | - Mark N Polizzotto
- Clinical Hub for Interventional Research, College of Health and Medicine, The Australian National University, Canberra, ACT, Australia
- Department of Clinical Haematology, Canberra Region Cancer Centre, The Canberra Hospital, Canberra, ACT, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
19
|
Jancy SV, Lupitha SS, Chandrasekharan A, Varadarajan SN, Nelson-Sathi S, Prasad R, Jones S, Easwaran S, Darvin P, Sivasailam A, Santhoshkumar TR. A high-throughput screening system for SARS-CoV-2 entry inhibition, syncytia formation and cell toxicity. Biol Proced Online 2023; 25:22. [PMID: 37495994 PMCID: PMC10373420 DOI: 10.1186/s12575-023-00214-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023] Open
Abstract
BACKGROUND The entry of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) into the host cell is mediated through the binding of the SARS-CoV-2 Spike protein via the receptor binding domain (RBD) to human angiotensin-converting enzyme 2 (hACE2). Identifying compounds that inhibit Spike-ACE2 binding would be a promising and safe antiviral approach against COVID-19. METHODS In this study, we used a BSL-2 compatible replication-competent vesicular stomatitis virus (VSV) expressing Spike protein of SARS-CoV-2 with eGFP reporter system (VSV-eGFP-SARS-CoV-2) in a recombinant permissive cell system for high-throughput screening of viral entry blockers. The SARS-CoV-2 permissive reporter system encompasses cells that stably express hACE2-tagged cerulean and H2B tagged with mCherry, as a marker of nuclear condensation, which also enables imaging of fused cells among infected EGFP positive cells and could provide real-time information on syncytia formation. RESULTS A limited high-throughput screening identified six natural products that markedly inhibited VSV-eGFP-SARS-CoV-2 with minimum toxicity. Further studies of Spike-S1 binding using the permissive cells showed Scillaren A and 17-Aminodemethoxygeldanamycin could inhibit S1 binding to ACE2 among the six leads. A real-time imaging revealed delayed inhibition of syncytia by Scillaren A, Proscillaridin, Acetoxycycloheximide and complete inhibition by Didemnin B indicating that the assay is a reliable platform for any image-based drug screening. CONCLUSION A BSL-2 compatible assay system that is equivalent to the infectious SARS-CoV-2 is a promising tool for high-throughput screening of large compound libraries for viral entry inhibitors against SARS-CoV-2 along with toxicity and effects on syncytia. Studies using clinical isolates of SARS-CoV-2 are warranted to confirm the antiviral potency of the leads and the utility of the screening system.
Collapse
Affiliation(s)
- Shine Varghese Jancy
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Santhik Subhasingh Lupitha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Aneesh Chandrasekharan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Shankara Narayanan Varadarajan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Shijulal Nelson-Sathi
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Roshny Prasad
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Sara Jones
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Sreekumar Easwaran
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Pramod Darvin
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Aswathy Sivasailam
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India
| | - Thankayyan Retnabai Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud P.O., Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
20
|
Ison MG, Weinstein DF, Dobryanska M, Holmes A, Phelan AM, Li Y, Gupta D, Narayan K, Tosh K, Hershberger E, Connolly LE, Yalcin I, Campanaro E, Hawn P, Schmidt P, for the EVADE Study Group. Prevention of COVID-19 Following a Single Intramuscular Administration of Adintrevimab: Results From a Phase 2/3 Randomized, Double-Blind, Placebo-Controlled Trial (EVADE). Open Forum Infect Dis 2023; 10:ofad314. [PMID: 37496612 PMCID: PMC10368201 DOI: 10.1093/ofid/ofad314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/09/2023] [Indexed: 07/28/2023] Open
Abstract
Background The prevention of coronavirus disease 2019 (COVID-19) in vulnerable populations is a global health priority. EVADE was a phase 2/3 multicenter, double-blind, randomized, placebo-controlled trial of adintrevimab, an extended-half-life monoclonal antibody, for postexposure (PEP) and pre-exposure prophylaxis (PrEP) of symptomatic COVID-19. Methods Eligible participants (vaccine-naive, aged ≥12 years) were randomized 1:1 to receive a single 300-mg intramuscular injection of adintrevimab or placebo. Primary efficacy end points were reverse transcription polymerase chain reaction (RT-PCR)-confirmed symptomatic COVID-19 through day 28 in the PEP cohort (RT-PCR-negative at baseline) and through month 3 in the PrEP cohort (RT-PCR-negative and seronegative at baseline) among participants randomized before emergence of the severe acute respiratory syndrome coronavirus 2 Omicron variant (November 30, 2021). Safety was assessed through 6 months. Results Between April 27, 2021, and January 11, 2022, 2582 participants were randomized. In the primary efficacy analysis, RT-PCR-confirmed symptomatic COVID-19 occurred in 3/175 (1.7%) vs 12/176 (6.8%) adintrevimab- and placebo-treated PEP participants, respectively (74.9% relative risk reduction [RRR]; standardized risk difference, -5.0%; 95% CI, -8.87% to -1.08%; P = .0123) and in 12/752 (1.6%) vs 40/728 (5.5%) adintrevimab- and placebo-treated PrEP participants, respectively (71.0% RRR; standardized risk difference, -3.9%; 95% CI, -5.75% to -2.01%; P < .0001). In a prespecified exploratory analysis of 428 PrEP participants randomized after the emergence of Omicron, adintrevimab reduced RT-PCR-confirmed symptomatic COVID-19 by 40.6% (standardized risk difference -8.4%; 95% CI, -15.35% to -1.46%; nominal P = .0177) vs placebo. Adintrevimab was well tolerated, with no serious drug-related adverse events reported. Conclusions A single intramuscular injection of adintrevimab provided prophylactic efficacy against COVID-19 due to susceptible variants without safety concerns. Clinical trial registration. NCT04859517.
Collapse
Affiliation(s)
- Michael G Ison
- Respiratory Diseases Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | | | - Marta Dobryanska
- Department of Emergency Care and ARENSIA Exploratory Medicine, Kyiv City Clinical Hospital No. 12, Kyiv, Ukraine
| | | | | | - Yong Li
- Invivyd, Inc., Waltham, Massachusetts, USA
| | | | | | | | | | | | | | | | | | - Pete Schmidt
- Correspondence: Pete Schmidt, MD, MS, Invivyd, Inc., 1601 Trapelo Road, Suite 178, Waltham, MA 02451 ()
| | | |
Collapse
|
21
|
Follmann D, O'Brien MP, Fintzi J, Fay MP, Montefiori D, Mateja A, Herman GA, Hooper AT, Turner KC, Chan KC, Forleo-Neto E, Isa F, Baden LR, El Sahly HM, Janes H, Doria-Rose N, Miller J, Zhou H, Dang W, Benkeser D, Fong Y, Gilbert PB, Marovich M, Cohen MS. Examining protective effects of SARS-CoV-2 neutralizing antibodies after vaccination or monoclonal antibody administration. Nat Commun 2023; 14:3605. [PMID: 37330602 PMCID: PMC10276829 DOI: 10.1038/s41467-023-39292-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/07/2023] [Indexed: 06/19/2023] Open
Abstract
While new vaccines for SARS-CoV-2 are authorized based on neutralizing antibody (nAb) titer against emerging variants of concern, an analogous pathway does not exist for preventative monoclonal antibodies. In this work, nAb titers were assessed as correlates of protection against COVID-19 in the casirivimab + imdevimab monoclonal antibody (mAb) prevention trial (ClinicalTrials.gov #NCT4452318) and in the mRNA-1273 vaccine trial (ClinicalTrials.gov #NCT04470427). In the mAb trial, protective efficacy of 92% (95% confidence interval (CI): 84%, 98%) is associated with a nAb titer of 1000 IU50/ml, with lower efficacy at lower nAb titers. In the vaccine trial, protective efficacies of 93% [95% CI: 91%, 95%] and 97% (95% CI: 95%, 98%) are associated with nAb titers of 100 and 1000 IU50/ml, respectively. These data quantitate a nAb titer correlate of protection for mAbs benchmarked alongside vaccine induced nAb titers and support nAb titer as a surrogate endpoint for authorizing new mAbs.
Collapse
Affiliation(s)
- Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | | | - Jonathan Fintzi
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Michael P Fay
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - David Montefiori
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | | | - Flonza Isa
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY, USA
| | | | - Hana M El Sahly
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicole Doria-Rose
- Vaccine Research Center, National Institutes of Health, Bethesda, MD, USA
| | | | | | | | - David Benkeser
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Youyi Fong
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Mary Marovich
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- Division of AIDS, National Institute of Allergy and Infectious Diseases, Bethesda, USA
| | - Myron S Cohen
- Institute for Global Health and Infectious Diseases, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Saleh NA. In-silico study: docking simulation and molecular dynamics of peptidomimetic fullerene-based derivatives against SARS-CoV-2 M pro. 3 Biotech 2023; 13:185. [PMID: 37193325 PMCID: PMC10182551 DOI: 10.1007/s13205-023-03608-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 05/03/2023] [Indexed: 05/18/2023] Open
Abstract
COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2, has become a global pandemic resulting in significant morbidity and mortality. This study presents 12 new peptidomimetic fullerene-based derivatives in three groups that are investigated theoretically as SARS-CoV-2 Mpro inhibitors to increase the chance of treating COVID-19. Studied compounds are designed and optimized at B88-LYP/DZVP method. Molecular descriptors results show the stability and reactivity of the compounds with Mpro, especially in the 3rd group (Ser compounds). However, Lipinski's Rule of Five values indicates that the compounds are not suitable as oral drugs. Furthermore, molecular docking simulations are carried out to investigate the binding affinity and interaction modes of the top five compounds (compounds 1, 9, 11, 2, and 10) with the Mpro protein, which have the lowest binding energy. Molecular dynamics simulations are also performed to evaluate the stability of the protein-ligand complexes with compounds 1 and 9 and compare them with natural substrate interaction. The analysis of RMSD, H-bonds, Rg, and SASA indicates that both compounds 1 (Gly-α acid) and 9 (Ser-α acid) have good stability and strong binding affinity with the Mpro protein. However, compound 9 shows slightly better stability and binding affinity compared to compound 1.
Collapse
Affiliation(s)
- Noha A. Saleh
- Department of Physics, College of Science, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia
- Basic and Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, 31441 Dammam, Saudi Arabia
| |
Collapse
|
23
|
Hausinger RI, Bachmann Q, Crone-Rawe T, Hannane N, Monsef I, Haller B, Heemann U, Skoetz N, Kreuzberger N, Schmaderer C. Effectiveness, Immunogenicity and Harms of Additional SARS-CoV-2 Vaccine Doses in Kidney Transplant Recipients: A Systematic Review. Vaccines (Basel) 2023; 11:vaccines11040863. [PMID: 37112775 PMCID: PMC10141039 DOI: 10.3390/vaccines11040863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/05/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Kidney transplant recipients (KTRs) who have a highly impaired immune response are in need of intensified and safe vaccination strategies to achieve seroconversion and prevent severe disease. METHODS We searched the Web of Science Core Collection, the Cochrane COVID-19 Study Register and the WHO COVID-19 global literature on coronavirus disease from January 2020 to 22 July 2022 for prospective studies that assessed immunogenicity and efficacy after three or more SARS-CoV-2 vaccine doses. RESULTS In 37 studies on 3429 patients, de novo seroconversion after three and four vaccine doses ranged from 32 to 60% and 25 to 37%. Variant-specific neutralization was 59 to 70% for Delta and 12 to 52% for Omicron. Severe disease after infection was rarely reported but all concerned KTRs lacked immune responses after vaccination. Studies investigating the clinical course of COVID-19 found remarkably higher rates of severe disease than in the general population. Serious adverse events and acute graft rejections were very rare. Substantial heterogeneity between the studies limited their comparability and summary. CONCLUSION Additional SARS-CoV-2 vaccine doses are potent and safe in general terms as well as regarding transplant-specific outcomes whilst the Omicron wave remains a significant threat to KTRs without adequate immune responses.
Collapse
Affiliation(s)
- Renate Ilona Hausinger
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Quirin Bachmann
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Timotius Crone-Rawe
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Nora Hannane
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Ina Monsef
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Bernhard Haller
- Institute for AI and Informatics in Medicine, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Uwe Heemann
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | - Nicole Skoetz
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Nina Kreuzberger
- Evidence-Based Medicine, Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum Rechts der Isar, TUM School of Medicine, Technical University of Munich, 81675 Munich, Germany
| |
Collapse
|
24
|
Meziyerh S, Bouwmans P, van Gelder T, van der Helm D, Messchendorp L, van der Boog PJM, de Fijter JW, Moes DJAR, de Vries APJ. Mycophenolic Acid Exposure Determines Antibody Formation Following SARS-CoV-2 Vaccination in Kidney Transplant Recipients: A Nested Cohort Study. Clin Pharmacol Ther 2023. [PMID: 36789469 DOI: 10.1002/cpt.2872] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
Despite (repeated) boosting, kidney transplant recipients (KTRs) may remain at increased risk of severe COVID-19 since a substantial number of individuals remain seronegative or with low antibody titers. In particular, mycophenolic acid use has been shown to affect antibody formation negatively and may be an important modifiable risk factor. We investigated the exposure-response relationship between mycophenolic acid 12-hour area under the curve (AUC0-12h ) exposure and seroconversion including antibody titers after vaccination using mRNA-1273 SARS-CoV-2 vaccine (Moderna) in 316 KTRs from our center that participated in the national Dutch renal patients COVID-19 vaccination - long term efficacy and safety of SARS-CoV-2 vaccination in kidney disease patients vaccination study. After two vaccination doses, 162 (51%) KTRs seroconverted. KTRs treated with mycophenolic acid showed less seroconversion and lower antibody titers compared with KTRs without mycophenolic acid (44% vs. 77%, and 36 binding antibody units (BAU)/mL vs. 340 BAU/mL; P < 0.001). The mean mycophenolic acid AUC0-12h exposure was significantly lower in KTRs who seroconverted compared with KTRs who did not (39 vs. 29 mg⋅h/L; P < 0.001). High mycophenolic acid exposure (±90 mg⋅h/L) and no exposure to mycophenolic acid resulted in a seroconversion rate ranging from 10% to 80%. Every 10 mg⋅h/L increase in mycophenolic acid AUC0-12h gave an adjusted odds ratio for seroconversion of 0.87 (95% confidence interval (CI), 0.79-0.97; P = 0.010) and 0.89 (95% CI, 0.85-0.93; P < 0.001) for KTRs on dual and triple maintenance immunosuppressive therapy, respectively. Higher mycophenolic acid AUC0-12h correlated with lower antibody titers (R = 0.44, P < 0.001). This study demonstrates the exposure-response relationship between gold standard mycophenolic acid exposure and antibody formation to support interventional studies investigating mycophenolic acid adjustment to improve antibody formation after further boosting.
Collapse
Affiliation(s)
- Soufian Meziyerh
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Pim Bouwmans
- Department of Internal Medicine, Division of Nephrology, Maastricht University Medical Center, Maastricht, The Netherlands.,Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, University of Maastricht, Maastricht, The Netherlands
| | - Teun van Gelder
- Department of Clinical Pharmacy and Toxicology, Leiden University Medical Center, Leiden, The Netherlands
| | - Danny van der Helm
- Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Lianne Messchendorp
- Department of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul J M van der Boog
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Johan W de Fijter
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Dirk Jan A R Moes
- Cardiovascular Research Institute Maastricht School for Cardiovascular Disease, University of Maastricht, Maastricht, The Netherlands
| | - Aiko P J de Vries
- Department of Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, The Netherlands.,Leiden University Medical Center Transplant Center, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
25
|
Zhao J, Zhu J, Huang C, Zhu X, Zhu Z, Wu Q, Yuan R. Uncovering the information immunology journals transmitted for COVID-19: A bibliometric and visualization analysis. Front Immunol 2022; 13:1035151. [PMID: 36405695 PMCID: PMC9670819 DOI: 10.3389/fimmu.2022.1035151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 10/17/2022] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Since the global epidemic of the coronavirus disease 2019 (COVID-19), a large number of immunological studies related to COVID-19 have been published in various immunology journals. However, the results from these studies were discrete, and no study summarized the important immunological information about COVID-19 released by these immunology journals. This study aimed to comprehensively summarize the knowledge structure and research hotspots of COVID-19 published in major immunology journals through bibliometrics. METHODS Publications on COVID-19 in major immunology journals were obtained from the Web of Science Core Collection. CiteSpace, VOSviewer, and R-bibliometrix were comprehensively used for bibliometric and visual analysis. RESULTS 1,331 and 5,000 publications of 10 journals with high impact factors and 10 journals with the most papers were included, respectively. The USA, China, England, and Italy made the most significant contributions to these papers. University College London, National Institute of Allergy and Infectious Diseases, Harvard Medical School, University California San Diego, and University of Pennsylvania played a central role in international cooperation in the immunology research field of COVID-19. Yuen Kwok Yung was the most important author in terms of the number of publications and citations, and the H-index. CLINICAL INFECTIOUS DISEASES and FRONTIERS IN IMMUNOLOGY were the most essential immunology journals. These immunology journals mostly focused on the following topics: "Delta/Omicron variants", "cytokine storm", "neutralization/neutralizing antibody", "T cell", "BNT162b2", "mRNA vaccine", "vaccine effectiveness/safety", and "long COVID". CONCLUSION This study systematically uncovered a holistic picture of the current research on COVID-19 published in major immunology journals from the perspective of bibliometrics, which will provide a reference for future research in this field.
Collapse
Affiliation(s)
- Jiefeng Zhao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jinfeng Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Chao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xiaojian Zhu
- Center for Digestive Disease, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Zhengming Zhu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Qinrong Wu
- Department of General Surgery, Yingtan City People’s Hospital, Yingtan, Jiangxi, China
| | - Rongfa Yuan
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
26
|
Gottlieb RL, Razonable RR. Stewardship of COVID-19 volunteers by nested trial design. THE LANCET INFECTIOUS DISEASES 2022; 22:1400-1401. [PMID: 35803288 PMCID: PMC9255946 DOI: 10.1016/s1473-3099(22)00429-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Robert L Gottlieb
- Center for Advanced Heart and Lung Disease, Baylor University Medical Center, Dallas, TX 75246, USA; Baylor Scott & White Research Institute, Dallas, TX, USA; Texas A&M Health Science Center, Dallas, TX, USA; School of Medicine, Texas Christian University, Fort Worth, TX, USA.
| | | |
Collapse
|