1
|
Dolange V, Slamanig S, Abdeljawad A, Lai TY, Lemus N, Singh G, Carreño JM, Abbad A, Srivastava K, Simon V, Sachithanandham J, Pekosz A, Sullivan D, Krammer F, Sun W, Palese P, González-Domínguez I. A Surrogate Enzyme-Linked Immunosorbent Assay to Select High-Titer Human Convalescent Plasma for Treating Immunocompromised Patients Infected With Severe Acute Respiratory Syndrome Coronavirus 2 Variants of Concern. J Infect Dis 2025; 231:e723-e733. [PMID: 39749487 PMCID: PMC11998579 DOI: 10.1093/infdis/jiae645] [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: 09/09/2024] [Revised: 11/07/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND The emergence of new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants challenges the treatment of immunocompromised patients against coronavirus disease 2019 (COVID-19). High-titer COVID-19 convalescent plasma (CCP) remains one of the few available therapeutics for these patients. We have revisited the selection of CCP samples and evaluated their efficacy against the Omicron XBB.1.5 variant, the dominant strain in 2023. METHODS A surrogate enzyme-linked immunoassay was reviewed to select CCP samples that ensure a protective level of neutralizing antibodies as the main correlate of protection. Antibody titers were analyzed in 500 serum samples from a population-based serosurvey at Mount Sinai Hospital in early 2023, and the results were validated with CCP samples (collected in 2020-2023) using an immunosuppressed mouse model. RESULTS Using logistic regression modeling, we have redefined high-titer CCP against the new variant in the postpandemic era, where over 97% of the population has natural or vaccine-induced antibodies against earlier SARS-CoV-2 strains. Treatment of immunocompromised mice with two doses (100 μL/dose) of CCP plasma via intraperitoneal injection reduced lung viral titers by 46-fold 3 days post-XBB.1.5 infection. CONCLUSIONS These findings will guide future efforts in selecting high-titer CCP for emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Victoria Dolange
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Stefan Slamanig
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
| | - Adam Abdeljawad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Tsoi Ying Lai
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Nicholas Lemus
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Anass Abbad
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Komal Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
- Department of Pathology, Molecular and Cell-Based Medicine
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Jaiprasath Sachithanandham
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Center for Vaccine Research and Pandemic Preparedness
- Department of Pathology, Molecular and Cell-Based Medicine
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Austria
| | - Weina Sun
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Peter Palese
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | | |
Collapse
|
2
|
Focosi D, Sullivan DJ, Franchini M. Development of antiviral drugs for COVID-19 in 2025: unmet needs and future challenges. Expert Rev Anti Infect Ther 2025:1-8. [PMID: 40007187 DOI: 10.1080/14787210.2025.2473044] [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: 12/11/2024] [Revised: 02/06/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
INTRODUCTION The success in the coronavirus infectious disease 2019 (COVID-19) pandemic containment largely originated from vaccine- and infection-elicited immunity, with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection only marginally mitigated by the availability of antiviral drugs. The current lack of effective antiviral prophylactic and therapeutic agents in immunocompromised patients highlights the need for a radical change in the design of both drug manufacturing and clinical trials. AREAS COVERED In this review, the authors summarize their suggestions for manufacturers, by reviewing classes of small molecule antivirals and passive immunotherapies and highlighting their limitations and unexploited potential. EXPERT OPINION Molecular and serological testing of patients can improve appropriateness. Efficacy of antivirals can be improved by combining different therapeutic classes while preserving economical sustainability. Respiratory delivery should be better investigated in clinical trials.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - David J Sullivan
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Massimo Franchini
- Division of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| |
Collapse
|
3
|
Shoham S, Focosi D, Franchini M, Atamna A. Novel approaches for preventing COVID-19 infection in immunocompromised patients with hematologic malignancies. Expert Rev Hematol 2025; 18:39-46. [PMID: 39610334 DOI: 10.1080/17474086.2024.2436973] [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: 08/27/2024] [Accepted: 11/28/2024] [Indexed: 11/30/2024]
Abstract
INTRODUCTION COVID-19 is a continuing challenge for immunocompromised patients with hematological malignancies. Such patients are at increased risk for complications, including hospitalization, respiratory failure, delayed anti-cancer therapies, and even death. In addition to non-pharmacologic interventions, the main strategies for prevention in such patients are vaccination and pre-exposure prophylaxis. AREAS COVERED In this narrative review, which relied on a review of the PubMed and bioRxiv databases (starting 1 November 2019), we summarize the epidemiology of COVID-19 and vaccine responses in patients with hematological malignancies and the use of antiviral agents as prophylaxis. A limitation to vaccination is suboptimal immune responses in immunocompromised patients, particularly those with abnormalities in lymphocyte count and function. A limitation to prophylaxis, which has only been proven effective for antiviral monoclonal antibodies (mAbs), is the emergence of resistant strains in the general population. EXPERT OPINION For immunocompromised patients with hematological malignancies, we recommend vaccinations as guided by evolving US Centers for Disease Control and Prevention (CDC) recommendations, consideration of pre-exposure prophylaxis with antiviral mAbs, providing that they are effective against circulating viral strains, and rapid diagnostic testing linked to early therapy for the prevention of severe complications of COVID-19 in those who have broken through the prevention strategies.
Collapse
Affiliation(s)
- Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantua, Italy
| | - Alaa Atamna
- Infectious Diseases, Rabin Medical Center (Beilinson Campus), Petah Tikva, Israel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
McLeod C, Dymock M, Flanagan KL, Plebanski M, Marshall HS, Estcourt MJ, Wadia U, Tjiam MC, Blyth CC, Subbarao K, Mordant FL, Nicholson S, Cain N, Brizuela R, Faust SN, Thornton RB, Ellis Z, Mckenzie A, Marsh JA, Snelling TL, Richmond PC. The Platform Trial In COVID-19 priming and BOOsting (PICOBOO): The immunogenicity, reactogenicity, and safety of licensed COVID-19 vaccinations administered as a second booster in BNT162b2 primed individuals aged 18-<50 and 50-<70 years old. J Infect 2024; 89:106346. [PMID: 39550007 DOI: 10.1016/j.jinf.2024.106346] [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: 08/20/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024]
Abstract
OBJECTIVES PICOBOO is a randomised, adaptive trial evaluating the immunogenicity, reactogenicity, and safety of COVID-19 booster strategies. Here, we present data for second boosters among individuals aged 18-<50 and 50-<70 years old primed with BNT162b2 until Day (D) 84. METHODS Immunocompetent adults who had received two doses of BNT162b2 and any licensed COVID-19 booster at least three months prior were eligible. Participants were randomly allocated to BNT162b2, mRNA-1273 or NVX-CoV2373 1:1:1. The log10 concentration of anti-spike Ig Total was summarised as the geometric mean concentration (GMC). Reactogenicity and safety outcomes were captured. RESULTS Between Mar 2022 and Aug 2023, 743 participants were recruited to the trial and had D28 samples available. Of these, 120 and 103 belonged to the 18-<50 y and 50-<70 y strata, respectively. The mean adjusted GMCs (95% credible intervals) peaked at D28; these were 41 262 (31 611, 51 105), 45 585 (34 194, 57 441) and 25 281 (20 021, 31 234) U/mL in the 18-<50 y stratum and 30 753 (25 071, 36 704), 35 132 (27 523, 42 239) and 17 322 (13 983, 20 641) U/mL in the 50-<70 y stratum following BNT162b2, mRNA-1273 and NVX-CoV2373, respectively. Limited neutralisation against Omicron subvariants was found following boosting with all vaccines. There were 4 possibly or probably-related adverse events in the 18-<50 y stratum and 5 events in the 50-<70 y stratum, and severe reactogenicity events were <10% and <11% in these strata, respectively. CONCLUSIONS Vaccines targeting Ancestral virus elicited boosted antibody responses to Ancestral virus but minimal neutralising antibody against Omicron variants.
Collapse
Affiliation(s)
- C McLeod
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Infectious Diseases Department, Perth Children's Hospital, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia; Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia.
| | - M Dymock
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; School of Population and Global Health, University of Western Australia, Nedlands, Australia
| | - K L Flanagan
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Tasmania, Australia; School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, Australia; School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University (RMIT), Melbourne, Australia
| | - M Plebanski
- School of Health and Biomedical Sciences, Royal Melbourne Institute of Technology University (RMIT), Melbourne, Australia
| | - H S Marshall
- Women's and Children's Health Network, North Adelaide, Australia; Robinson Research Institute and Adelaide Medical School, The University of Adelaide, Australia
| | - M J Estcourt
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia
| | - U Wadia
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Infectious Diseases Department, Perth Children's Hospital, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia
| | - M C Tjiam
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia
| | - C C Blyth
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Infectious Diseases Department, Perth Children's Hospital, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia; Department of Microbiology, Pathwest Laboratory Medicine WA, QEII Medical Centre, Nedlands, Australia
| | - K Subbarao
- WHO Collaborating Centre for Reference and Research on Influenza, University of Melbourne, Parkville, Victoria, Australia; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - F L Mordant
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - S Nicholson
- Victorian Infectious Diseases Reference Laboratory, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Department of Infectious Diseases, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - N Cain
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - R Brizuela
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - S N Faust
- National Institute of Health Research Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom; Faculty of Medicine and Institute for Life Sciences, University of Southampton, United Kingdom
| | - R B Thornton
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia
| | - Z Ellis
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia
| | - A Mckenzie
- The Kids Research Institute of Australia, Nedlands, Australia
| | - J A Marsh
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia
| | - T L Snelling
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Australia
| | - P C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute of Australia, Nedlands, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia; Centre for Child Health Research, University of Western Australia, Crawley, Australia; General Paediatrics and Immunology Departments, Perth Children's Hospital, Nedlands, Australia
| |
Collapse
|
5
|
Kajova M, Khawaja T, Levonen I, Pietilä JP, Virtanen J, Pakkanen SH, Välimaa H, Nousiainen A, Hepojoki J, Sironen T, Vierikko A, Ihalainen J, Vapalahti O, Kantele A. Convalescent plasma therapy for COVID-19 - Donor selection strategies and establishment of a plasma bank. New Microbes New Infect 2024; 62:101525. [PMID: 39584055 PMCID: PMC11584594 DOI: 10.1016/j.nmni.2024.101525] [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: 08/27/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
Background Early in the COVID-19 pandemic, convalescent plasma (CP) emerged as a potentially effective treatment neutralising SARS-CoV-2. Early CP therapy with high neutralising antibody (NAb) titre may benefit COVID-19 outpatients and, in sufficient quantities even some hospitalised patients. This study details the process of setting up a CP bank, containing high- and low-titre CP for a clinical trial. Study design and methods We identified 18-65-year-old convalescents with SARS-CoV-2 NAb titres of ≥1:40 in microneutralisation test (MNT). Following eligibility pre-screening, the Finnish Red Cross Blood Service (FRCBS) determined suitability as CP donors. Results Of the 6466 COVID-19 convalescents contacted, 1481 provided serum, with 851 (57.5 %) exhibiting NAb titres ≥1:40. Participation barriers included reluctance, advanced age and, for women, insufficient body size. Of the volunteers, 125 were evaluated at FRCBS, with major exclusions for HLA antibodies (42 women), interferon antibodies (five men), and NAb titres waning below 1:20 (16 participants). Finally, 70 underwent plasmapheresis, resulting in 50 suitable CP donors (0.8 % of initial contacts and 3.4 % of those tested for NAb). Discussion The process of setting up a CP bank proved challenging. Excessive laboratory workloads during a pandemic hamper their ability to conduct MNT, underscoring the need for rapid screening tests. Only a small proportion of our convalescents exhibited high-titre CP, this fraction declining over time because of waning immunity. Strict plasmapheresis criteria further constrained donor eligibility. Establishing a plasma bank requires meticulous planning to maximize efficiency. Detailed insights from current experiences may prove critical in future pandemics before other remedies and vaccines become available.
Collapse
Affiliation(s)
- Mikael Kajova
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Tamim Khawaja
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Iris Levonen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| | - Jenni Virtanen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - Sari H. Pakkanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Hanna Välimaa
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Arttu Nousiainen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
| | - Jussi Hepojoki
- Department of Virology, University of Helsinki, Helsinki, Finland
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Tarja Sironen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | | | | | - Olli Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Center, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - Anu Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, University of Helsinki, Finland
- FIMAR, Finnish Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Finland
| |
Collapse
|
6
|
Al Khalaf R, Bernasconi A, Pinoli P. Systematic analysis of SARS-CoV-2 Omicron subvariants' impact on B and T cell epitopes. PLoS One 2024; 19:e0307873. [PMID: 39298436 PMCID: PMC11412522 DOI: 10.1371/journal.pone.0307873] [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] [Received: 03/22/2024] [Accepted: 07/14/2024] [Indexed: 09/21/2024] Open
Abstract
INTRODUCTION Epitopes are specific structures in antigens that are recognized by the immune system. They are widely used in the context of immunology-related applications, such as vaccine development, drug design, and diagnosis / treatment / prevention of disease. The SARS-CoV-2 virus has represented the main point of interest within the viral and genomic surveillance community in the last four years. Its ability to mutate and acquire new characteristics while it reorganizes into new variants has been analyzed from many perspectives. Understanding how epitopes are impacted by mutations that accumulate on the protein level cannot be underrated. METHODS With a focus on Omicron-named SARS-CoV-2 lineages, including the last WHO-designated Variants of Interest, we propose a workflow for data retrieval, integration, and analysis pipeline for conducting a database-wide study on the impact of lineages' characterizing mutations on all T cell and B cell linear epitopes collected in the Immune Epitope Database (IEDB) for SARS-CoV-2. RESULTS Our workflow allows us to showcase novel qualitative and quantitative results on 1) coverage of viral proteins by deposited epitopes; 2) distribution of epitopes that are mutated across Omicron variants; 3) distribution of Omicron characterizing mutations across epitopes. Results are discussed based on the type of epitope, the response frequency of the assays, and the sample size. Our proposed workflow can be reproduced at any point in time, given updated variant characterizations and epitopes from IEDB, thereby guaranteeing to observe a quantitative landscape of mutations' impact on demand. CONCLUSION A big data-driven analysis such as the one provided here can inform the next genomic surveillance policies in combatting SARS-CoV-2 and future epidemic viruses.
Collapse
Affiliation(s)
- Ruba Al Khalaf
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milano, Italia
| | - Anna Bernasconi
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milano, Italia
| | - Pietro Pinoli
- Dipartimento di Elettronica, Informazione e Bioingegneria (DEIB), Politecnico di Milano, Milano, Italia
| |
Collapse
|
7
|
Focosi D. Monoclonal Antibody Therapies Against SARS-CoV-2: Promises and Realities. Curr Top Microbiol Immunol 2024. [PMID: 39126484 DOI: 10.1007/82_2024_268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Monoclonal antibodies targeting the Spike protein of SARS-CoV-2 have been widely deployed in the ongoing COVID-19 pandemic. I review here the impact of those therapeutics in the early pandemic, ranging from structural classification to outcomes in clinical trials to in vitro and in vivo evidence of basal and treatment-emergent immune escape. Unfortunately, the Omicron variant of concern has completely reset all achievements so far in mAb therapy for COVID-19. Despite the intrinsic limitations of this strategy, future developments such as respiratory delivery of further engineered mAb cocktails could lead to improved outcomes.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy.
| |
Collapse
|
8
|
Sullivan DJ. Convalescent Plasma and Other Antibody Therapies for Infectious Diseases-Lessons Learned from COVID-19 and Future Prospects. Curr Top Microbiol Immunol 2024. [PMID: 39117846 DOI: 10.1007/82_2024_273] [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] [Indexed: 08/10/2024]
Abstract
Antiviral passive antibody therapy includes convalescent plasma, hyperimmune globulin, and monoclonal antibodies. Passive antibodies have proven effective in reducing morbidity and mortality for SARS-CoV-2 and other infectious diseases when given early in the disease course with sufficiently high specific total and neutralizing antibody levels. Convalescent plasma can be delivered to patients before vaccination implementation or novel drug production. Carefully designed and executed randomized controlled trials near the pandemic outset are important for regulatory bodies, healthcare workers, guideline committees, the public, and the government. Unfortunately, many otherwise well-designed antibody-based clinical trials in COVID-19 were futile, either because they intervened too late in the disease or provided plasma with insufficient antibodies. The need for early treatment mandates outpatient clinical trials in parallel with inpatient trials. Early outpatient COVID-19 convalescent plasma transfusion with high antibody content within 9 days of symptom onset has proven effective in blunting disease progression and reducing hospitalization, thus reducing hospital overcrowding in a pandemic. Convalescent plasma offers the opportunity for hope by enabling community participation in outpatient curative therapy while monoclonal therapies, vaccines, and drugs are being developed. Maintaining the appropriate infrastructure for antibody infusion in both outpatient and inpatient facilities is critical for future pandemic readiness.
Collapse
Affiliation(s)
- David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St Rm W4606, Baltimore, MD, 21205, USA.
| |
Collapse
|
9
|
Shoham S. Convalescent Plasma for Immunocompromised Patients. Curr Top Microbiol Immunol 2024. [PMID: 39117848 DOI: 10.1007/82_2024_272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
COVID-19 convalescent plasma (CCP) is an important therapeutic option for immunocompromised patients with COVID-19. Such patients are at increased risk for serious complications of infection and may also develop a unique syndrome of persistent infection. This article reviews the rationale for CCP utilization in immunocompromised patients and the evidence for its value in immunosuppressed patients with both acute and persistent COVID-19. Both historical precedence and understanding of the mechanisms of action of antibody treatment support this use, as do several lines of evidence derived from case series, comparative studies, randomized trials, and systematic reviews of the literature. A summary of recommendations from multiple practice guidelines is also provided.
Collapse
Affiliation(s)
- Shmuel Shoham
- Department of Medicine, Johns Hopkins School of Medicine, 1830 East Monument St., Room 447, Baltimore, MD, 21205, USA.
| |
Collapse
|
10
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
11
|
Karbiener M, Kindle G, Meyts I, Seppänen MRJ, Candotti F, Kamieniak M, Ilk R, Kreil TR, Seidel MG. Clinical efficacy of SARS-CoV-2 Omicron-neutralizing antibodies in immunoglobulin preparations for the treatment of agammaglobulinemia in patients with primary antibody deficiency. J Med Virol 2024; 96:e29738. [PMID: 38884390 DOI: 10.1002/jmv.29738] [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: 03/28/2024] [Revised: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Immunocompromised individuals are at significantly elevated risk for severe courses of coronavirus disease 2019 (COVID-19). In addition to vaccination, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralizing antibodies (nAbs) have been applied throughout the pandemic, with time of treatment onset and potency against the currently prevailing virus variant identified as relevant factors for medical benefit. Using data from the European Society for Immunodeficiencies (ESID) registry, the present study evaluated COVID-19 cases in three groups of patients with inborn errors of immunity (IEI; 981 agammaglobulinemia patients on immunoglobulin replacement therapy (IGRT); 8960 non-agammaglobulinemia patients on IGRT; 14 428 patients without IGRT), and the neutralizing capacity of 1100 immunoglobulin lots against SARS-CoV-2 ("Wuhan" and Omicron strains), throughout 3 years. From the first (2020/2021) to the second (2021/2022) cold season, i.e., during the virus drift to the more contagious Omicron variants, an increase in case numbers was recorded that was comparable (~2- to 3-fold) for all three study groups. During the same period, immunoglobulin lots showed a profound nAb increase against the archetypal SARS-CoV-2 strain, yet only low levels of Omicron nAbs. Notably, shortly before the third (2022/2023) cold season, Omicron-neutralizing capacity of released immunoglobulin lots had plateaued at high levels. From the second to the third cold season, COVID-19 cases dropped markedly. While a ~6-fold case reduction was recorded for the groups of non-agammaglobulinemia patients on IGRT and IEI patients not receiving IGRT, the decline was ~30-fold for the group of agammaglobulinemia patients on IGRT. These findings suggest a substantial COVID-19-protective effect of IGRT, at least for distinct groups of antibody-deficient patients.
Collapse
Affiliation(s)
- Michael Karbiener
- Global Pathogen Safety, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Gerhard Kindle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Biobanking FREEZE, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, KU Leuven, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
- ERN-RITA Core Center, RITAFIN, Helsinki, Finland
- Rare Disease Center and Pediatric Research Center, Children and Adolescents, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Fabio Candotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Marta Kamieniak
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts, USA
| | - Reinhard Ilk
- Global Manufacturing Sciences, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Thomas R Kreil
- Global Pathogen Safety, Takeda Manufacturing Austria AG, Vienna, Austria
| | - Markus G Seidel
- on behalf of the ESID Registry Working Party; Styrian Children's Cancer Research Unit for Cancer and Inborn Errors of the Blood and Immunity in Children, Medical University of Graz, Graz, Austria
- Division of Pediatric Hematology and Oncology, Department of Pediatric and Adolescent Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
12
|
Franchini M, Focosi D. Towards the identification of the correct place for convalescent plasma among COVID-19 therapies. Infect Dis (Lond) 2024; 56:421-422. [PMID: 38549506 DOI: 10.1080/23744235.2024.2333977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 04/24/2024] Open
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
13
|
Khawaja T, Kajova M, Levonen I, Pietilä JP, Välimaa H, Paajanen J, Pakkanen SH, Patjas A, Montonen R, Miettinen S, Virtanen J, Smura T, Sironen T, Fagerlund R, Ugurlu H, Iheozor-Ejiofor R, Saksela K, Vahlberg T, Ranki A, Vierikko A, Ihalainen J, Vapalahti O, Kantele A. Double-blinded, randomised, placebo-controlled trial of convalescent plasma for COVID-19: analyses by neutralising antibodies homologous to recipients' variants. Infect Dis (Lond) 2024; 56:423-433. [PMID: 38513074 DOI: 10.1080/23744235.2024.2329957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024] Open
Abstract
INTRODUCTION Convalescent plasma (CP) emerged as potential treatment for COVID-19 early in the pandemic. While efficacy in hospitalised patients has been lacklustre, CP may be beneficial at the first stages of disease. Despite multiple new variants emerging, no trials have involved analyses on variant-specific antibody titres of CP. METHODS We recruited hospitalised COVID-19 patients within 10 days of symptom onset and, employing a double-blinded approach, randomised them to receive 200 ml convalescent plasma with high (HCP) or low (LCP) neutralising antibody (NAb) titre against the ancestral strain (Wuhan-like variant) or placebo in 1:1:1 ratio. Primary endpoints comprised intubation, corticosteroids for symptom aggravation, and safety assessed as serious adverse events. For a preplanned ad hoc analysis, the patients were regrouped by infused CP's NAb titers to variants infecting the recipients i.e. by titres of homologous HCP (hHCP) or LCP (hLCP). RESULTS Of the 57 patients, 18 received HCP, 19 LCP and 20 placebo, all groups smaller than planned. No significant differences were found for primary endpoints. In ad hoc analysis, hHCPrecipients needed significantly less respiratory support, and appeared to be given corticosteroids less frequently (1/14; 7.1%) than those receiving hLCP (9/23; 39.1%) or placebo (8/20; 40%), (p = 0.077). DISCUSSION Our double-blinded, placebo-controlled CP therapy trial remained underpowered and does not allow any firm conclusions for early-stage hospitalised COVID-19 patients. Interestingly, however, regrouping by homologous - recipients' variant-specific - CP titres suggested benefits for hHCP. We encourage similar re-analysis of ongoing/previous larger CP studies. TRIAL REGISTRATION ClinTrials.gov identifier: NCT0473040.
Collapse
Affiliation(s)
- T Khawaja
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - M Kajova
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - I Levonen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J P Pietilä
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - H Välimaa
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - J Paajanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S H Pakkanen
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - A Patjas
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| | - R Montonen
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - S Miettinen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - J Virtanen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - T Smura
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - T Sironen
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - R Fagerlund
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - H Ugurlu
- Department of Virology, University of Helsinki, Helsinki, Finland
| | - R Iheozor-Ejiofor
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
| | - K Saksela
- Department of Virology, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - T Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - A Ranki
- Department of Dermatology, Allergology and Venereology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - A Vierikko
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - J Ihalainen
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - O Vapalahti
- Department of Virology, University of Helsinki, Helsinki, Finland
- Department of Veterinary Biosciences, Faculty of Veterinary Medicine, University of Helsinki, Helsinki, Finland
- HUS Diagnostic Centre, HUSLAB, Clinical Microbiology, Helsinki University Hospital, Helsinki, Finland
| | - A Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Infectious Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- FIMAR, Multidisciplinary Center of Excellence in Antimicrobial Resistance Research, University of Helsinki, Helsinki, Finland
| |
Collapse
|
14
|
Chen X, Mohapatra A, Nguyen HTV, Schimanski L, Kit Tan T, Rijal P, Chen CP, Cheng SH, Lee WH, Chou YC, Townsend AR, Ma C, Huang KYA. The presence of broadly neutralizing anti-SARS-CoV-2 RBD antibodies elicited by primary series and booster dose of COVID-19 vaccine. PLoS Pathog 2024; 20:e1012246. [PMID: 38857264 PMCID: PMC11192315 DOI: 10.1371/journal.ppat.1012246] [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] [Received: 12/01/2023] [Revised: 06/21/2024] [Accepted: 05/08/2024] [Indexed: 06/12/2024] Open
Abstract
Antibody-mediated immunity plays a key role in protection against SARS-CoV-2. We characterized B-cell-derived anti-SARS-CoV-2 RBD antibody repertoires from vaccinated and infected individuals and elucidate the mechanism of action of broadly neutralizing antibodies and dissect antibodies at the epitope level. The breadth and clonality of anti-RBD B cell response varies among individuals. The majority of neutralizing antibody clones lose or exhibit reduced activities against Beta, Delta, and Omicron variants. Nevertheless, a portion of anti-RBD antibody clones that develops after a primary series or booster dose of COVID-19 vaccination exhibit broad neutralization against emerging Omicron BA.2, BA.4, BA.5, BQ.1.1, XBB.1.5 and XBB.1.16 variants. These broadly neutralizing antibodies share genetic features including a conserved usage of the IGHV3-53 and 3-9 genes and recognize three clustered epitopes of the RBD, including epitopes that partially overlap the classically defined set identified early in the pandemic. The Fab-RBD crystal and Fab-Spike complex structures corroborate the epitope grouping of antibodies and reveal the detailed binding mode of broadly neutralizing antibodies. Structure-guided mutagenesis improves binding and neutralization potency of antibody with Omicron variants via a single amino-substitution. Together, these results provide an immunological basis for partial protection against severe COVID-19 by the ancestral strain-based vaccine and indicate guidance for next generation monoclonal antibody development and vaccine design.
Collapse
Affiliation(s)
- Xiaorui Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | - Hong Thuy Vy Nguyen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Chemical Biology and Molecular Biophysics program, Taiwan International Graduate Program, Academia Sinica, Taipei, Taiwan
- Institute of Biochemical Sciences, National Taiwan University, Taipei, Taiwan
| | - Lisa Schimanski
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Tiong Kit Tan
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Pramila Rijal
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Cheng-Pin Chen
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shu-Hsing Cheng
- Department of Infectious Diseases, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan, and School of Public Health, Taipei Medical University, Taipei, Taiwan
| | - Wen-Hsin Lee
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chi Chou
- Biomedical Translation Research Center, Academia Sinica, Taipei, Taiwan
| | - Alain R. Townsend
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Che Ma
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Kuan-Ying A. Huang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Immunology and Department of Pediatrics, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
15
|
Li J, Chu LT, Hartanto H, Guo G, Liu L, Wu J, Wu M, Cui C, Wang G, Liu W, Kwong HK, Wu S, Chen TH. Microfluidic particle counter visualizing mucosal antibodies against SARS-CoV-2 in the upper respiratory tract for rapid evaluation of immune protection. LAB ON A CHIP 2024; 24:2658-2668. [PMID: 38660972 DOI: 10.1039/d4lc00118d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Mucosal antibodies in the upper respiratory tract are the earliest and most critical responders to prevent respiratory infections, providing an indication for the rapid evaluation of immune protection. Here, we report a microfluidic particle counter that directly visualizes mucosal antibody levels in nasal mucus. The mucosal anti-SARS-CoV-2 spike receptor binding domain (RBD) antibodies in nasal secretions first react with magnetic microparticles (MMPs) and polystyrene microparticles (PMPs) that are surface-modified to form a "MMPs-anti-spike RBD IgG-PMPs" complex when RBD is present. After magnetic separation and loading into the microfluidic particle counter, the free PMPs, which are reduced with increasing anti-spike RBD IgG antibody levels, are trapped by a microfluidic particle dam and accumulate in the trapping channel. A sensitive mode [limit of detection (LOD): 14.0 ng mL-1; sample-to-answer time: 70 min] and an equipment-free rapid mode (LOD: 37.4 ng mL-1; sample-to-answer time: 20 min) were achieved. Eighty-seven nasal secretion (NS) samples from vaccinees were analyzed using our microfluidic particle counter, and the results closely resemble those of the gold-standard enzyme-linked immunosorbent assay (ELISA). The analysis shows that higher antibody levels were found in convalescent volunteers compared to noninfected volunteers. Together, we demonstrate a rapid kit that directly indicates immune status, which can guide vaccine strategy for individuals and the government.
Collapse
Affiliation(s)
- Jiaheng Li
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Lok Ting Chu
- Department of Chemistry and Molecular Biology, School of Basic Medical Sciences, Guang Dong Medical University, Zhanjiang, China
| | - Hogi Hartanto
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Guihuan Guo
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Lu Liu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Jianpeng Wu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Minghui Wu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Chenyu Cui
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong, SAR, China
| | - Gaobo Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Wengang Liu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Hoi Kwan Kwong
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Siying Wu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
| | - Ting-Hsuan Chen
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong, SAR, China.
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Science Park, Hong Kong, SAR, China
| |
Collapse
|
16
|
Meganck RM, Edwards CE, Mallory ML, Lee RE, Dang H, Bailey AB, Wykoff JA, Gallant SC, Zhu DR, Yount BL, Kato T, Shaffer KM, Nakano S, Cawley AM, Sontake V, Wang JR, Hagan RS, Miller MB, Tata PR, Randell SH, Tse LV, Ehre C, Okuda K, Boucher RC, Baric RS. SARS-CoV-2 variant of concern fitness and adaptation in primary human airway epithelia. Cell Rep 2024; 43:114076. [PMID: 38607917 PMCID: PMC11165423 DOI: 10.1016/j.celrep.2024.114076] [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: 09/15/2023] [Revised: 02/09/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 pandemic is characterized by the emergence of novel variants of concern (VOCs) that replace ancestral strains. Here, we dissect the complex selective pressures by evaluating variant fitness and adaptation in human respiratory tissues. We evaluate viral properties and host responses to reconstruct forces behind D614G through Omicron (BA.1) emergence. We observe differential replication in airway epithelia, differences in cellular tropism, and virus-induced cytotoxicity. D614G accumulates the most mutations after infection, supporting zoonosis and adaptation to the human airway. We perform head-to-head competitions and observe the highest fitness for Gamma and Delta. Under these conditions, RNA recombination favors variants encoding the B.1.617.1 lineage 3' end. Based on viral growth kinetics, Alpha, Gamma, and Delta exhibit increased fitness compared to D614G. In contrast, the global success of Omicron likely derives from increased transmission and antigenic variation. Our data provide molecular evidence to support epidemiological observations of VOC emergence.
Collapse
Affiliation(s)
- Rita M Meganck
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Caitlin E Edwards
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Michael L Mallory
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Rhianna E Lee
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Hong Dang
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Alexis B Bailey
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Jason A Wykoff
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Samuel C Gallant
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Deanna R Zhu
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Boyd L Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Takafumi Kato
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Kendall M Shaffer
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Satoko Nakano
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Anne Marie Cawley
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | | | - Jeremy R Wang
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Robert S Hagan
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Division of Pulmonary Diseases and Critical Care Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Melissa B Miller
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | | | - Scott H Randell
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Longping V Tse
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, MO 63104, USA
| | - Camille Ehre
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Kenichi Okuda
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Richard C Boucher
- Marsico Lung Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA.
| |
Collapse
|
17
|
Billi B, Cholley P, Grobost V, Clément M, Rieu V, Le Guenno G, Lobbes H. Intravenous immunoglobulins for the treatment of prolonged COVID-19 in immunocompromised patients: a brief report. Front Immunol 2024; 15:1399180. [PMID: 38707896 PMCID: PMC11069322 DOI: 10.3389/fimmu.2024.1399180] [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: 03/11/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Primary humoral deficiency and secondary B-cell depletion may lead to prolonged Sars-Cov-2 infection due to a decreased viral clearance. Prolonged infection is mainly driven by the lack of anti-Sars-Cov-2 immunoglobulin (IVIg) especially in patients with no vaccine response. Anti-spike immunoglobulin can be provided by infusion of convalescent patients' plasma: recent studies highlighted that commercial immunoglobulin show high titers of neutralizing IgG. We conducted a single center retrospective cohort. We included 9 patients (6 males, median age 74 years old): one patient with X-linked agammaglobulinemia and 8 patients treated with rituximab (2 granulomatosis with polyangiitis, 1 neuromyelitis optica, 4 low grade B-cell lymphoma and 1 EBV post-transplant lymphoproliferative disorder). Mean serum globulin was 4 ± 1.6 g/L. 7/8 had received at least 3 doses of mRNA anti-Sars-Cov-2 vaccine (median 4) with no response (anti-Spike IgG 0 for 6 patients). In this specific population requiring oxygen therapy but no intensive care support, the administration of IVIg was well tolerated and provided a swift improvement of clinical status, a significant decrease of inflammation associated to the an improvement of radiological patterns. Our results suggest that immunoglobulin could be used as a salvage therapy as an alternative to convalescent plasma but highly stringent patient selection is required due to the worldwide shortage of IVIg.
Collapse
Affiliation(s)
- Bénédicte Billi
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Paul Cholley
- Service de Radiologie, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Grobost
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mélissa Clément
- Service de Médecine Interne, Hôpital Henri Mondor, Aurillac, France
| | - Virginie Rieu
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Guillaume Le Guenno
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Hervé Lobbes
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Institut Pascal, Unité Mixte de Recherche (UMR) 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
18
|
Padilla S, Ledesma C, García-Abellán J, García JA, Fernández-González M, de la Rica A, Galiana A, Gutiérrez F, Masiá M. Long COVID across SARS-CoV-2 variants, lineages, and sublineages. iScience 2024; 27:109536. [PMID: 38585665 PMCID: PMC10995878 DOI: 10.1016/j.isci.2024.109536] [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: 11/09/2023] [Revised: 02/09/2024] [Accepted: 03/17/2024] [Indexed: 04/09/2024] Open
Abstract
This prospective study aimed to determine the prevalence of long COVID in patients hospitalized for SARS-CoV-2 infection from March 2020 to July 2022 and assess the impact of different viral lineages. A total of 2,524 patients were followed up for 12 months, with persistent symptoms reported in 35.2% at one month, decreasing thereafter. Omicron variant patients initially showed higher symptom intensity, but this trend diminished over time. Certain viral lineages, notably Delta lineages AY.126 and AY.43, and Omicron sublineages BA.1.17, BA.2.56, and BA.5.1, consistently correlated with more severe symptoms. Overall, long COVID prevalence and severity were similar across SARS-CoV-2 variants. Specific lineages may influence post-COVID sequelae persistence and severity.
Collapse
Affiliation(s)
- Sergio Padilla
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- Universidad Miguel Hernández de Elche, San Juan de Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Christian Ledesma
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
| | - Javier García-Abellán
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- Universidad Miguel Hernández de Elche, San Juan de Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - José Alberto García
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Fernández-González
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Alba de la Rica
- Microbiology Service, Hospital General Universitario de Elche, Alicante, Spain
| | - Antonio Galiana
- Microbiology Service, Hospital General Universitario de Elche, Alicante, Spain
| | - Félix Gutiérrez
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- Universidad Miguel Hernández de Elche, San Juan de Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Masiá
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
- Universidad Miguel Hernández de Elche, San Juan de Alicante, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
19
|
Antia A, Alvarado DM, Zeng Q, Casorla-Perez LA, Davis DL, Sonnek NM, Ciorba MA, Ding S. SARS-CoV-2 Omicron BA.1 Variant Infection of Human Colon Epithelial Cells. Viruses 2024; 16:634. [PMID: 38675974 PMCID: PMC11055019 DOI: 10.3390/v16040634] [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: 03/05/2024] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
The Omicron variant of SARS-CoV-2, characterized by multiple subvariants including BA.1, XBB.1.5, EG.5, and JN.1, became the predominant strain in early 2022. Studies indicate that Omicron replicates less efficiently in lung tissue compared to the ancestral strain. However, the infectivity of Omicron in the gastrointestinal tract is not fully defined, despite the fact that 70% of COVID-19 patients experience digestive disease symptoms. Here, using primary human colonoids, we found that, regardless of individual variability, Omicron infects colon cells similarly or less effectively than the ancestral strain or the Delta variant. The variant induced limited type III interferon expression and showed no significant impact on epithelial integrity. Further experiments revealed inefficient cell-to-cell spread and spike protein cleavage in the Omicron spike protein, possibly contributing to its lower infectious particle levels. The findings highlight the variant-specific replication differences in human colonoids, providing insights into the enteric tropism of Omicron and its relevance to long COVID symptoms.
Collapse
Affiliation(s)
- Avan Antia
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.A.); (Q.Z.)
| | - David M. Alvarado
- Inflammatory Bowel Diseases Center, Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (D.M.A.); (D.L.D.); (N.M.S.)
| | - Qiru Zeng
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.A.); (Q.Z.)
| | - Luis A. Casorla-Perez
- Inflammatory Bowel Diseases Center, Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (D.M.A.); (D.L.D.); (N.M.S.)
| | - Deanna L. Davis
- Inflammatory Bowel Diseases Center, Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (D.M.A.); (D.L.D.); (N.M.S.)
| | - Naomi M. Sonnek
- Inflammatory Bowel Diseases Center, Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (D.M.A.); (D.L.D.); (N.M.S.)
| | - Matthew A. Ciorba
- Inflammatory Bowel Diseases Center, Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (D.M.A.); (D.L.D.); (N.M.S.)
| | - Siyuan Ding
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA; (A.A.); (Q.Z.)
| |
Collapse
|
20
|
Mercolini F, Abram N, Cesaro S. Managing acute COVID-19 in immunocompromised pediatric patients. Expert Rev Clin Immunol 2024; 20:349-357. [PMID: 38099388 DOI: 10.1080/1744666x.2023.2295982] [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: 05/14/2023] [Accepted: 12/13/2023] [Indexed: 03/16/2024]
Abstract
INTRODUCTION SARS-CoV-2 infection is a potentially life-threatening infection in immunocompromised pediatric patients, and its management has rapidly evolved during the pandemic. To control SARS-CoV-2 infection over time, the scenario changed for the better with the introduction of specific treatments such as antiviral drugs, vaccines, and monoclonal antibodies, together with drugs blocking the inflammatory cytokine cascade and improvements in supportive care. AREAS COVERED This paper discusses the therapeutic strategies to apply for patients affected by COVID-19 in the pediatric population, with a focus on the immunocompromised patients. EXPERT OPINION Treatment in pediatric patients retraces the therapies investigated and approved in adults and must be calibrated on the basis of the severity of the infection (anti-spike monoclonal antibody, antivirals, anti-inflammatory drugs, and immunomodulators). Transmission prevention policies and vaccination reduce the risk of infection, while early intervention in the immunocompromised patients at high-risk of progression to severe-critical COVID-19 may reduce the period of viral shedding and the need for hospitalization, intensive care admission, and death. In hemato-oncological patients, the delayed treatment of SARS-CoV-2 infection or COVID-19 disease represents a frequent complication and its impact on the patient outcome remains a matter of research for the next few years.
Collapse
Affiliation(s)
- Federico Mercolini
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS, Azienda Ospedaliero-Universitaria di Bologna institution, Bologna, Italy
| | - Nicoletta Abram
- Pediatric Oncology and Hematology "Lalla Seràgnoli", IRCCS, Azienda Ospedaliero-Universitaria di Bologna institution, Bologna, Italy
| | - Simone Cesaro
- Pediatric Hematology Oncology, Department of Mother and Child, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| |
Collapse
|
21
|
Ny Mioramalala DJ, Ratovoson R, Tagnouokam-Ngoupo PA, Abessolo Abessolo H, Mindimi Nkodo JM, Bouting Mayaka G, Tsoungui Atangana PC, Randrianarisaona F, Pélembi P, Nzoumbou-Boko R, Coti-Reckoundji CSG, Manirakiza A, Rahantamalala A, Randremanana RV, Tejiokem MC, Schoenhals M. SARS-CoV-2 Neutralizing Antibodies in Three African Countries Following Multiple Distinct Immune Challenges. Vaccines (Basel) 2024; 12:363. [PMID: 38675745 PMCID: PMC11054809 DOI: 10.3390/vaccines12040363] [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: 02/23/2024] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic has affected Madagascar, Cameroon, and the Central African Republic (CAR), with each experiencing multiple waves by mid-2022. This study aimed to evaluate immunity against SARS-CoV-2 strains Wuhan (W) and BA.2 (BA.2) among healthcare workers (HCWs) in these countries, focusing on vaccination and natural infection effects. METHODS HCWs' serum samples were analyzed for neutralizing antibodies (nAbs) against W and BA.2 variants, with statistical analyses comparing responses between countries and vaccination statuses. RESULTS Madagascar showed significantly higher nAb titers against both strains compared to CAR and Cameroon. Vaccination notably increased nAb levels against W by 2.6-fold in CAR and 1.8-fold in Madagascar, and against BA.2 by 1.6-fold in Madagascar and 1.5-fold in CAR. However, in Cameroon, there was no significant difference in nAb levels between vaccinated and unvaccinated groups. CONCLUSION This study highlights the complex relationship between natural and vaccine-induced immunity, emphasizing the importance of assessing immunity in regions with varied epidemic experiences and low vaccination rates.
Collapse
Affiliation(s)
- Diary Juliannie Ny Mioramalala
- Institut Pasteur of Madagascar, Immunology of Infectious Diseases, Antananarivo 101, Madagascar; (D.J.N.M.); (F.R.); (A.R.)
| | - Rila Ratovoson
- Institut Pasteur of Madagascar, Epidemiology and Clinical Research, Antananarivo 101, Madagascar; (R.R.); (R.V.R.)
| | - Paul Alain Tagnouokam-Ngoupo
- Centre Pasteur du Cameroon, Epidemiology and Public Health, Yaoundé P.O. Box 1274, Cameroon; (P.A.T.-N.); (M.C.T.)
| | | | | | | | | | - Fanirisoa Randrianarisaona
- Institut Pasteur of Madagascar, Immunology of Infectious Diseases, Antananarivo 101, Madagascar; (D.J.N.M.); (F.R.); (A.R.)
| | - Pulchérie Pélembi
- Institut Pasteur of Bangui, Epidemiology, Bangui P.O. Box 923, Central African Republic; (P.P.); (R.N.-B.); (C.S.G.C.-R.); (A.M.)
| | - Romaric Nzoumbou-Boko
- Institut Pasteur of Bangui, Epidemiology, Bangui P.O. Box 923, Central African Republic; (P.P.); (R.N.-B.); (C.S.G.C.-R.); (A.M.)
| | | | - Alexandre Manirakiza
- Institut Pasteur of Bangui, Epidemiology, Bangui P.O. Box 923, Central African Republic; (P.P.); (R.N.-B.); (C.S.G.C.-R.); (A.M.)
| | - Anjanirina Rahantamalala
- Institut Pasteur of Madagascar, Immunology of Infectious Diseases, Antananarivo 101, Madagascar; (D.J.N.M.); (F.R.); (A.R.)
| | - Rindra Vatosoa Randremanana
- Institut Pasteur of Madagascar, Epidemiology and Clinical Research, Antananarivo 101, Madagascar; (R.R.); (R.V.R.)
| | - Mathurin Cyrille Tejiokem
- Centre Pasteur du Cameroon, Epidemiology and Public Health, Yaoundé P.O. Box 1274, Cameroon; (P.A.T.-N.); (M.C.T.)
| | - Matthieu Schoenhals
- Institut Pasteur of Madagascar, Immunology of Infectious Diseases, Antananarivo 101, Madagascar; (D.J.N.M.); (F.R.); (A.R.)
| |
Collapse
|
22
|
Park HS, Yin A, Barranta C, Lee JS, Caputo CA, Sachithanandham J, Li M, Yoon S, Sitaras I, Jedlicka A, Eby Y, Ram M, Fernandez RE, Baker OR, Shenoy AG, Mosnaim GS, Fukuta Y, Patel B, Heath SL, Levine AC, Meisenberg BR, Spivak ES, Anjan S, Huaman MA, Blair JE, Currier JS, Paxton JH, Gerber JM, Petrini JR, Broderick PB, Rausch W, Cordisco ME, Hammel J, Greenblatt B, Cluzet VC, Cruser D, Oei K, Abinante M, Hammitt LL, Sutcliffe CG, Forthal DN, Zand MS, Cachay ER, Raval JS, Kassaye SG, Marshall CE, Yarava A, Lane K, McBee NA, Gawad AL, Karlen N, Singh A, Ford DE, Jabs DA, Appel LJ, Shade DM, Lau B, Ehrhardt S, Baksh SN, Shapiro JR, Ou J, Na YB, Knoll MD, Ornelas-Gatdula E, Arroyo-Curras N, Gniadek TJ, Caturegli P, Wu J, Ndahiro N, Betenbaugh MJ, Ziman A, Hanley DF, Casadevall A, Shoham S, Bloch EM, Gebo KA, Tobian AA, Laeyendecker O, Pekosz A, Klein SL, Sullivan DJ. Outpatient COVID-19 convalescent plasma recipient antibody thresholds correlated to reduced hospitalizations within a randomized trial. JCI Insight 2024; 9:e178460. [PMID: 38483534 PMCID: PMC11141865 DOI: 10.1172/jci.insight.178460] [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: 12/14/2023] [Accepted: 03/06/2024] [Indexed: 03/26/2024] Open
Abstract
BACKGROUNDCOVID-19 convalescent plasma (CCP) virus-specific antibody levels that translate into recipient posttransfusion antibody levels sufficient to prevent disease progression are not defined.METHODSThis secondary analysis correlated donor and recipient antibody levels to hospitalization risk among unvaccinated, seronegative CCP recipients within the outpatient, double-blind, randomized clinical trial that compared CCP to control plasma. The majority of COVID-19 CCP arm hospitalizations (15/17, 88%) occurred in this unvaccinated, seronegative subgroup. A functional cutoff to delineate recipient high versus low posttransfusion antibody levels was established by 2 methods: (i) analyzing virus neutralization-equivalent anti-Spike receptor-binding domain immunoglobulin G (anti-S-RBD IgG) responses in donors or (ii) receiver operating characteristic (ROC) curve analysis.RESULTSSARS-CoV-2 anti-S-RBD IgG antibody was volume diluted 21.3-fold into posttransfusion seronegative recipients from matched donor units. Virus-specific antibody delivered was approximately 1.2 mg. The high-antibody recipients transfused early (symptom onset within 5 days) had no hospitalizations. A CCP-recipient analysis for antibody thresholds correlated to reduced hospitalizations found a statistical significant association between early transfusion and high antibodies versus all other CCP recipients (or control plasma), with antibody cutoffs established by both methods-donor-based virus neutralization cutoffs in posttransfusion recipients (0/85 [0%] versus 15/276 [5.6%]; P = 0.03) or ROC-based cutoff (0/94 [0%] versus 15/267 [5.4%]; P = 0.01).CONCLUSIONIn unvaccinated, seronegative CCP recipients, early transfusion of plasma units in the upper 30% of study donors' antibody levels reduced outpatient hospitalizations. High antibody level plasma units, given early, should be reserved for therapeutic use.TRIAL REGISTRATIONClinicalTrials.gov NCT04373460.FUNDINGDepartment of Defense (W911QY2090012); Defense Health Agency; Bloomberg Philanthropies; the State of Maryland; NIH (3R01AI152078-01S1, U24TR001609-S3, 1K23HL151826NIH); the Mental Wellness Foundation; the Moriah Fund; Octapharma; the Healthnetwork Foundation; the Shear Family Foundation; the NorthShore Research Institute; and the Rice Foundation.
Collapse
Affiliation(s)
- Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anna Yin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Caelan Barranta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - John S. Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christopher A. Caputo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jaiprasath Sachithanandham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maggie Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Steve Yoon
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Anne Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Yolanda Eby
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Malathi Ram
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Reinaldo E. Fernandez
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Owen R. Baker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aarthi G. Shenoy
- Department of Medicine, Division of Hematology and Oncology, MedStar Washington Hospital Center, Washington DC, USA
| | - Giselle S. Mosnaim
- Division of Allergy and Immunology, Department of Medicine, NorthShore University Health System, Evanston, Illinois, USA
| | - Yuriko Fukuta
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Bela Patel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Sonya L. Heath
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Adam C. Levine
- Department of Emergency Medicine, Rhode Island Hospital, Brown University, Providence, Rhode Island, USA
| | | | - Emily S. Spivak
- Department of Medicine, Division of Infectious Diseases, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Shweta Anjan
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Moises A. Huaman
- Department of Medicine, Division of Infectious Diseases, University of Cincinnati, Cincinnati, Ohio, USA
| | - Janis E. Blair
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic Hospital, Phoenix, Arizona, USA
| | - Judith S. Currier
- Department of Medicine, Division of Infectious Diseases, UCLA, Los Angeles, California, USA
| | - James H. Paxton
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jonathan M. Gerber
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | | | | | | | | | - Jean Hammel
- Nuvance Health Norwalk Hospital, Norwalk, Connecticut, USA
| | | | - Valerie C. Cluzet
- Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, New York, USA
| | - Daniel Cruser
- Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, New York, USA
| | - Kevin Oei
- Ascada Research, Fullerton, California, USA
| | | | - Laura L. Hammitt
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Catherine G. Sutcliffe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Donald N. Forthal
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine, California, USA
| | - Martin S. Zand
- Department of Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Edward R. Cachay
- Department of Medicine, Division of Infectious Diseases, UCSD, San Diego, California, USA
| | - Jay S. Raval
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - Seble G. Kassaye
- Department of Medicine, Division of Infectious Diseases, Georgetown University Medical Center, Washington DC, USA
| | - Christi E. Marshall
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Karen Lane
- Department of Neurology, Brain Injury Outcomes
| | | | | | | | - Atika Singh
- Department of Neurology, Brain Injury Outcomes
| | | | - Douglas A. Jabs
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Lawrence J. Appel
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David M. Shade
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Bryan Lau
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Stephan Ehrhardt
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sheriza N. Baksh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Janna R. Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Jiangda Ou
- Department of Neurology, Brain Injury Outcomes
| | - Yu Bin Na
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Maria D. Knoll
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Elysse Ornelas-Gatdula
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, Maryland, USA
| | - Netzahualcoyotl Arroyo-Curras
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas J. Gniadek
- Department of Pathology and Laboratory Medicine, Northshore University Health System, Evanston, Illinois, USA
| | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jinke Wu
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nelson Ndahiro
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael J. Betenbaugh
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Alyssa Ziman
- Department of Pathology and Laboratory Medicine, Wing-Kwai and Alice Lee-Tsing Chung Transfusion Service, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | | | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kelly A. Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases (NIAID), Baltimore, Maryland, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Sabra L. Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - David J. Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Bedding MJ, Franck C, Johansen-Leete J, Aggarwal A, Maxwell JWC, Patel K, Hawkins PME, Low JKK, Siddiquee R, Sani HM, Ford DJ, Turville S, Mackay JP, Passioura T, Christie M, Payne RJ. Discovery of High Affinity Cyclic Peptide Ligands for Human ACE2 with SARS-CoV-2 Entry Inhibitory Activity. ACS Chem Biol 2024; 19:141-152. [PMID: 38085789 DOI: 10.1021/acschembio.3c00568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
The development of effective antiviral compounds is essential for mitigating the effects of the COVID-19 pandemic. Entry of SARS-CoV-2 virions into host cells is mediated by the interaction between the viral spike (S) protein and membrane-bound angiotensin-converting enzyme 2 (ACE2) on the surface of epithelial cells. Inhibition of this viral protein-host protein interaction is an attractive avenue for the development of antiviral molecules with numerous spike-binding molecules generated to date. Herein, we describe an alternative approach to inhibit the spike-ACE2 interaction by targeting the spike-binding interface of human ACE2 via mRNA display. Two consecutive display selections were performed to direct cyclic peptide ligand binding toward the spike binding interface of ACE2. Through this process, potent cyclic peptide binders of human ACE2 (with affinities in the picomolar to nanomolar range) were identified, two of which neutralized SARS-CoV-2 entry. This work demonstrates the potential of targeting ACE2 for the generation of anti-SARS-CoV-2 therapeutics as well as broad spectrum antivirals for the treatment of SARS-like betacoronavirus infection.
Collapse
Affiliation(s)
- Max J Bedding
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Charlotte Franck
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jason Johansen-Leete
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | | | - Joshua W C Maxwell
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Karishma Patel
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Paige M E Hawkins
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jason K K Low
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Rezwan Siddiquee
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Hakimeh Moghaddas Sani
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Daniel J Ford
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| | | | - Joel P Mackay
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Toby Passioura
- Sydney Analytical Core Research Facility, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Mary Christie
- School of Life and Environmental Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
- School of Medical Sciences, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Richard J Payne
- School of Chemistry, The University of Sydney, Sydney, New South Wales 2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
24
|
Park HS, Yin A, Barranta C, Lee JS, Caputo CA, Sachithanandham J, Li M, Yoon S, Sitaras I, Jedlicka A, Eby Y, Ram M, Fernandez RE, Baker OR, Shenoy AG, Mosnaim GS, Fukuta Y, Patel B, Heath SL, Levine AC, Meisenberg BR, Spivak ES, Anjan S, Huaman MA, Blair JE, Currier JS, Paxton JH, Gerber JM, Petrini JR, Broderick PB, Rausch W, Cordisco ME, Hammel J, Greenblatt B, Cluzet VC, Cruser D, Oei K, Abinante M, Hammitt LL, Sutcliffe CG, Forthal DN, Zand MS, Cachay ER, Raval JS, Kassaye SG, Marshall CE, Yarava A, Lane K, McBee NA, Gawad AL, Karlen N, Singh A, Ford DE, Jabs DA, Appel LJ, Shade DM, Lau B, Ehrhardt S, Baksh SN, Shapiro JR, Ou J, Na YB, Knoll MD, Ornelas-Gatdula E, Arroyo-Curras N, Gniadek TJ, Caturegli P, Wu J, Ndahiro N, Betenbaugh MJ, Ziman A, Hanley DF, Casadevall A, Shoham S, Bloch EM, Gebo KA, Tobian AAR, Laeyendecker O, Pekosz A, Klein SL, Sullivan DJ. Outpatient COVID-19 convalescent plasma recipient antibody thresholds correlated to reduced hospitalizations within a randomized trial. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.13.23288353. [PMID: 37131659 PMCID: PMC10153328 DOI: 10.1101/2023.04.13.23288353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
BACKGROUND The COVID-19 convalescent plasma (CCP) viral specific antibody levels that translate into recipient post-transfusion antibody levels sufficient to prevent disease progression is not defined. METHODS This secondary analysis correlated donor and recipient antibody levels to hospitalization risk among unvaccinated, seronegative CCP recipients within the outpatient, double blind, randomized clinical trial that compared CCP to control plasma. The majority of COVID-19 CCP arm hospitalizations (15/17, 88%) occurred in this unvaccinated, seronegative subgroup. A functional cutoff to delineate recipient high versus low post-transfusion antibody levels was established by two methods: 1) analyzing virus neutralization-equivalent anti-S-RBD IgG responses in donors or 2) receiver operating characteristic (ROC) analysis. RESULTS SARS-CoV-2 anti-S-RBD IgG antibody was diluted by a factor of 21.3 into post-transfusion seronegative recipients from matched donor units. Viral specific antibody delivered approximated 1.2 mg. The high antibody recipients transfused early (symptom onset within 5 days) had no hospitalizations. A CCP recipient analysis for antibody thresholds correlated to reduced hospitalizations found a significant association with Fisher's exact test between early and high antibodies versus all other CCP recipients (or control plasma) with antibody cutoffs established by both methods-donor virus neutralization-based cutoff: (0/85; 0% versus 15/276; 5.6%) p=0.03 or ROC based cutoff: (0/94; 0% versus 15/267; 5.4%) p=0.01. CONCLUSION In unvaccinated, seronegative CCP recipients, early transfusion of plasma units corresponding to the upper 30% of all study donors reduced outpatient hospitalizations. These high antibody level plasma units, given early, should be reserved for therapeutic use.Trial registration: NCT04373460. FUNDING Defense Health Agency and others.
Collapse
Affiliation(s)
- Han-Sol Park
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anna Yin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Caelan Barranta
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - John S Lee
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christopher A Caputo
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jaiprasath Sachithanandham
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maggie Li
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Steve Yoon
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Ioannis Sitaras
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Anne Jedlicka
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yolanda Eby
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Malathi Ram
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Reinaldo E Fernandez
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Owen R Baker
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aarthi G Shenoy
- Department of Medicine, Division of Hematology and Oncology, MedStar Washington Hospital Center, Washington DC, USA
| | - Giselle S Mosnaim
- Division of Allergy and Immunology, Department of Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Yuriko Fukuta
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Bela Patel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Texas Health Science Center, Houston, TX, USA
| | - Sonya L Heath
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Adam C Levine
- Department of Emergency Medicine, Rhode Island Hospital, Brown University, Providence, RI, USA
| | | | - Emily S Spivak
- Department of Medicine, Division of Infectious Diseases, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Shweta Anjan
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Moises A Huaman
- Department of Medicine, Division of Infectious Diseases, University of Cincinnati, Cincinnati, OH, USA
| | - Janis E Blair
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic Hospital, Phoenix, AZ, USA
| | - Judith S Currier
- Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, CA, USA
| | - James H Paxton
- Department of Emergency Medicine, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jonathan M Gerber
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | | | | | | | | | - Jean Hammel
- Nuvance Health Norwalk Hospital, Norwalk, CT, USA
| | | | - Valerie C Cluzet
- Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | - Daniel Cruser
- Nuvance Health Vassar Brothers Medical Center, Poughkeepsie, NY, USA
| | | | | | - Laura L Hammitt
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Catherine G Sutcliffe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Donald N Forthal
- Department of Medicine, Division of Infectious Diseases, University of California, Irvine, CA, USA
| | - Martin S Zand
- Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Edward R Cachay
- Department of Medicine, Division of Infectious Diseases, University of California, San Diego, CA, USA
| | - Jay S Raval
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Seble G Kassaye
- Department of Medicine, Division of Infectious Diseases, Georgetown University Medical Center Washington DC, USA
| | - Christi E Marshall
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anusha Yarava
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Karen Lane
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nichol A McBee
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy L Gawad
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nicky Karlen
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Atika Singh
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Daniel E Ford
- Institute for Clinical and Translational Research Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Douglas A Jabs
- Department of Ophthalmology Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Lawrence J Appel
- Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David M Shade
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Bryan Lau
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stephan Ehrhardt
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sheriza N Baksh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Janna R Shapiro
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jiangda Ou
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yu Bin Na
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Maria D Knoll
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Elysse Ornelas-Gatdula
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore MD, USA
| | - Netzahualcoyotl Arroyo-Curras
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thomas J Gniadek
- Department of Pathology and Laboratory Medicine, Northshore University Health System, Evanston, IL
| | - Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jinke Wu
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Nelson Ndahiro
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J Betenbaugh
- Advanced Mammalian Biomanufacturing Innovation Center, Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Alyssa Ziman
- Department of Pathology and Laboratory Medicine, Wing-Kwai and Alice Lee-Tsing Chung Transfusion Service, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Daniel F Hanley
- Department of Neurology, Brain Injury Outcomes, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kelly A Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Baltimore, MD, USA
| | - Andrew Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Sabra L Klein
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - David J Sullivan
- W. Harry Feinstone Department of Molecular Microbiology and Immunology; Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| |
Collapse
|
25
|
Caturegli P, Laeyendecker O, Tobian AAR, Sullivan DJ. Hundred-fold increase in SARS-CoV-2 spike antibody levels over three years in a hospital clinical laboratory. Microbiol Spectr 2023; 11:e0218323. [PMID: 37811983 PMCID: PMC10715067 DOI: 10.1128/spectrum.02183-23] [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: 05/24/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
IMPORTANCE Despite the evolution of SARS-CoV-2 variants of concern and ongoing transmission, COVID-19 hospitalization and mortality rates continue to decline. Both the percent seropositive and antibody levels have risen over the past 3 years. Here, we observe more than 90% seropositivity as well as more than a hundred-fold increase in spike IgG levels in a tertiary hospital clinical immunology laboratory setting. Antibody effector functions (such as neutralization, opsonization, and complement activation) and cell-mediated immunity all contribute to protection from COVID-19 progression to hospitalization, and all correlate to the total SARS-CoV-2 antibody levels. We recommend therapeutic COVID-19 convalescent plasma be restricted to the top 20% of potential donors to maintain activity against ongoing SARS-CoV-2 variant evolution.
Collapse
Affiliation(s)
- Patrizio Caturegli
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Aaron A. R. Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
26
|
Siripongboonsitti T, Nontawong N, Tawinprai K, Suptawiwat O, Soonklang K, Poovorawan Y, Mahanonda N. Efficacy of combined COVID-19 convalescent plasma with oral RNA-dependent RNA polymerase inhibitor treatment versus neutralizing monoclonal antibody therapy in COVID-19 outpatients: a multi-center, non-inferiority, open-label randomized controlled trial (PlasMab). Microbiol Spectr 2023; 11:e0325723. [PMID: 37975699 PMCID: PMC10714803 DOI: 10.1128/spectrum.03257-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023] Open
Abstract
This pivotal study reveals that high neutralizing titer COVID-19 convalescent plasma therapy (CPT) combined with favipiravir (FPV) is non-inferior to sotrovimab in preventing hospitalization and severe outcomes in outpatients with mild-to-moderate COVID-19 and high-risk comorbidities. It underscores the potential of CPT-FPV as a viable alternative to neutralizing monoclonal antibodies like sotrovimab, especially amid emerging variants with spike protein mutations. The study's unique approach, comparing a monoclonal antibody with CPT, demonstrates the efficacy of early intervention using high neutralizing antibody titer CPT, even in populations with a significant proportion of elderly patients. These findings are crucial, considering the alternative treatment challenges, especially in resource-limited countries, posed by the rapidly mutating SARS-CoV-2 virus and the need for adaptable therapeutic strategies.
Collapse
Affiliation(s)
- Taweegrit Siripongboonsitti
- Division of Infectious Diseases, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | | | - Kriangkrai Tawinprai
- Division of Infectious Diseases, Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Ornpreya Suptawiwat
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Kamonwan Soonklang
- Center of Learning and Research in Celebration of HRH Princess Chulabhorn 60th Birthday Anniversary, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Yong Poovorawan
- Department of Pediatrics, Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nithi Mahanonda
- Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
27
|
Herrera S, Aguado JM, Candel FJ, Cordero E, Domínguez-Gil B, Fernández-Ruiz M, Los Arcos I, Len Ò, Marcos MÁ, Muñez E, Muñoz P, Rodríguez-Goncer I, Sánchez-Céspedes J, Valerio M, Bodro M. Executive summary of the consensus statement of the group for the study of infection in transplantation and other immunocompromised host (GESITRA-IC) of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC) on the treatment of SARS-CoV-2 infection in solid organ transplant recipients. Transplant Rev (Orlando) 2023; 37:100788. [PMID: 37591117 DOI: 10.1016/j.trre.2023.100788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/19/2023]
Affiliation(s)
- Sabina Herrera
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain
| | - Jose M Aguado
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Francisco Javier Candel
- Clinical Microbiology & Infectious Diseases, Transplant Coordination, Hospital Clínico Universitario San Carlos, Madrid 28040, Spain; Department of Clinical Microbiology and Infectious Diseases, Hospital Clínico San Carlos, Madrid, Spain
| | - Elisa Cordero
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | | | - Mario Fernández-Ruiz
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Ibai Los Arcos
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | - Òscar Len
- Infectious Diseases Department, Hospital Universitari Vall D'Hebron, Barcelona, Spain
| | | | - Elena Muñez
- Infectious Diseases Unit, Internal Medicine Department, University Hospital Puerta de Hierro, Majadahonda, Madrid, Spain
| | - Patricia Muñoz
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Isabel Rodríguez-Goncer
- Infectious Diseases Unit, Hospital Universitario 12 de Octubre (Madrid), Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Javier Sánchez-Céspedes
- Infectious Diseases Unit, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina Sevilla, Sevilla, Spain
| | - Maricela Valerio
- Department of Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, 9 Madrid, Spain
| | - Marta Bodro
- Department of Infectious Diseases, Hospital Clínic, IDIBAPS (Institut D'Investigacions Biomèdiques Agust Pi I Sunyer), Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
28
|
Focosi D, Franchini M, Nicastri E, Sullivan DJ, Casadevall A. Convalescent Plasma Versus Hyperimmune Immunoglobulins. Clin Infect Dis 2023; 77:1356-1357. [PMID: 37399022 DOI: 10.1093/cid/ciad406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 06/28/2023] [Indexed: 07/04/2023] Open
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Emanuele Nicastri
- Division of Infectious Diseases, National Institute for Infectious Diseases "Lazzaro Spallanzani," Rome, Italy
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
29
|
|
30
|
Buck AM, Deitchman AN, Takahashi S, Lu S, Goldberg SA, Bodansky A, Kung A, Hoh R, Williams MC, Kerbleski M, Maison DP, Deveau TM, Munter SE, Lombardo J, Wrin T, Petropoulos CJ, Durstenfeld MS, Hsue PY, Daniel Kelly J, Greenhouse B, Martin JN, Deeks SG, Peluso MJ, Henrich TJ. The breadth of the neutralizing antibody response to original SARS-CoV-2 infection is linked to the presence of Long COVID symptoms. J Med Virol 2023; 95:e29216. [PMID: 37988251 PMCID: PMC10754238 DOI: 10.1002/jmv.29216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/20/2023] [Accepted: 10/21/2023] [Indexed: 11/23/2023]
Abstract
The associations between longitudinal dynamics and the breadth of SARS-CoV-2 neutralizing antibody (nAb) response with various Long COVID phenotypes before vaccination are not known. The capacity of antibodies to cross-neutralize a variety of viral variants may be associated with ongoing pathology and persistent symptoms. We measured longitudinal neutralizing and cross-neutralizing antibody responses to pre- and post-SARS-CoV-2 Omicron variants in participants infected early in the COVID-19 pandemic, before widespread rollout of SARS-CoV-2 vaccines. Cross-sectional regression models adjusted for clinical covariates and longitudinal mixed-effects models were used to determine the impact of the breadth and rate of decay of neutralizing responses on the development of Long COVID symptoms, as well as Long COVID phenotypes. We identified several novel relationships between SARS-CoV-2 antibody neutralization and the presence of Long COVID symptoms. Specifically, we show that, although nAb responses to the original, infecting strain of SARS-CoV-2 were not associated with Long COVID in cross-sectional analyses, cross-neutralization ID50 levels to the Omicron BA.5 variant approximately 4 months following acute infection was independently and significantly associated with greater odds of Long COVID and with persistent gastrointestinal and neurological symptoms. Longitudinal modeling demonstrated significant associations in the overall levels and rates of decay of neutralization capacity with Long COVID phenotypes. A higher proportion of participants had antibodies capable of neutralizing Omicron BA.5 compared with BA.1 or XBB.1.5 variants. Our findings suggest that relationships between various immune responses and Long COVID are likely complex but may involve the breadth of antibody neutralization responses.
Collapse
Affiliation(s)
- Amanda M. Buck
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Amelia N. Deitchman
- Department of Clinical Pharmacy, University of California San Francisco, San Francisco, CA, United States
| | - Saki Takahashi
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Scott Lu
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Sarah A. Goldberg
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Aaron Bodansky
- Division of Pediatric Critical Care Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Andrew Kung
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, United States
| | - Rebecca Hoh
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Meghann C. Williams
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Marian Kerbleski
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - David P. Maison
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Tyler-Marie Deveau
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Sadie E. Munter
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| | - James Lombardo
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Terri Wrin
- Monogram Biosciences, South San Francisco, CA, United States
| | | | - Matthew S. Durstenfeld
- Division of Cardiology, University of California San Francisco, San Francisco, CA, United States
| | - Priscilla Y. Hsue
- Division of Cardiology, University of California San Francisco, San Francisco, CA, United States
| | - J. Daniel Kelly
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Bryan Greenhouse
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Jeffrey N. Martin
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Steven G. Deeks
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, ID and Global Medicine, University of California San Francisco, San Francisco, CA, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, United States
| |
Collapse
|
31
|
Focosi D, Spezia PG, Maggi F. SARS-CoV-2 BA.2.86 ("Pirola"): Is it Pi or Just Another Omicron Sublineage? Vaccines (Basel) 2023; 11:1634. [PMID: 38005966 PMCID: PMC10674951 DOI: 10.3390/vaccines11111634] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
The SARS-CoV-2 sublineage BA [...].
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| | - Pietro Giorgio Spezia
- National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (P.G.S.); (F.M.)
| | - Fabrizio Maggi
- National Institute for Infectious Diseases “Lazzaro Spallanzani” IRCCS, 00149 Rome, Italy; (P.G.S.); (F.M.)
| |
Collapse
|
32
|
Guo W, Zheng Y, Feng S. Omicron related COVID-19 prevention and treatment measures for patients with hematological malignancy and strategies for modifying hematologic treatment regimes. Front Cell Infect Microbiol 2023; 13:1207225. [PMID: 37928188 PMCID: PMC10622671 DOI: 10.3389/fcimb.2023.1207225] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
The Omicron variant of SARS-CoV-2 has rapidly become the dominant strain worldwide due to its high transmissibility, although it appears to be less pathogenic than previous strains. However, individuals with hematological malignancy (HM) and COVID-19 remain susceptible to severe infection and mortality, especially those with chronic lymphocytic leukemia (CLL) and those undergoing chimeric antigen receptor T-cell (CAR-T) treatment. Hematologists should thoroughly assess the severity of the patient's hematological disease and the potential risk of SARS-CoV-2 infection before initiating chemotherapy or immunosuppressive treatment. Vaccination and booster doses are strongly recommended and patients with a poor vaccine response may benefit from long-acting COVID-19 neutralizing monoclonal antibodies (such as Evusheld). Early use of small molecule antiviral drugs is recommended for managing mild COVID-19 in HM patients and those with severe immunodeficiency may benefit from SARS-CoV-2 neutralizing monoclonal antibody therapy and high-titer COVID-19 convalescent plasma (CCP). For moderate to severe cases, low-dose glucocorticoids in combination with early antiviral treatment can be administered, with cytokine receptor antagonists or JAK inhibitors added if the condition persists or worsens. In the treatment of hematological malignancies, delaying chemotherapy is preferable for CLL, acute leukemia (AL), and low-risk myelodysplastic syndrome (MDS), but if the disease progresses, appropriate adjustments in dosage and frequency of treatment are required, with the avoidance of anti-CD20 monoclonal antibody, CAR-T and hematopoietic stem cell transplantation (HSCT). Patients with chronic myelocytic leukemia (CML) and myeloproliferative neoplasms (MPNs) can continue current treatment. What's more, non-drug protective measures, the development of new vaccines and antiviral drugs, and monitoring of mutations in immunocompromised populations are particularly important.
Collapse
Affiliation(s)
- Wenjing Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yizhou Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
33
|
Sarkar A, Santra D, Sundar Panja A, Maiti S. Immunoinformatics and MD-simulation data suggest that Omicron spike epitopes are more interacting to IgG via better MHC recognition than Delta variant. Int Immunopharmacol 2023; 123:110636. [PMID: 37499394 DOI: 10.1016/j.intimp.2023.110636] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/28/2023] [Accepted: 07/09/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Recently, in Nov 2021, in South Africa, the SARS CoV-2 variant Omicron was found to be highly infectious and transmissible but with the least fatality. It occupies the nasopharynx-oropharynx and easily spreads. The epidemiological data/reports suggest that several vaccines failed to neutralize Omicron. It has a large number of spike mutations and the RNA/protein vaccines were developed from its predecessors that may justify its escape in most neutralization reactions. Its lower immuno-suppression/cytokine-storming/inflammatory-response effects need exploration. OBJECTIVES In the current study, we attempted to delineate the comparative interaction of different variants' spikes with multiple recognition sites on IgG and HLA-typing of MHC class and I and II. METHODS All SARS-CoV-2 spike-proteins/human-IgG/MHC-I & II were obtained from the NCBI/ PDB/GISAID database. Initial 3D-structures of the unavailable proteins were constructed by Homology-Modeling (Swissmodel-Expasy) and optimized (PROCHECK). Molecular-docking of spike-IgG/spike- I & MHC-II was performed (HADDOCK2.4/HawkDock) with active-residue screening (CPORT). Antigenicity of epitopes was determined (Vaxigen v2.0-server) and the epitope-model prepared (PEP-FOLD3-server). The binding-affinity/biological-interfaces/visualize were performed (PRODIGY-PyMOL2). We also examined the genesis of feasible transition pathways of functional docked complexes (iMODs) of MHC with different epitopes and antibodies of IgG with different variants. Further, Molecular-Dynamic-Simulation was performed by GROMACS 2023.1 software package. The MD-simulation was run with 100 ns (300 k-heating/1-atm pressure). RESULTS Surface-area with interactomes, H-bonding and polar/non-polar bonding were the highest in Omicron spike-IgG interaction. Unlike other variants, both the L and H chains of at least three different recognition sites of IgG interact with the N-terminal and C-terminal RBD of the S1-portion and partially bind to S2. In other cases, binding was observed in either NTD or CTD with a lesser number of bonding-interactomes, especially in Delta spike-Ab interaction. In the case of MHC class-I & II, the highest binding affinity/surface was noticed by Omicron and least by the Delta variant. The MD simulation data of lower RMSD values of the Delta and Omicron variants indicate improved structural stability and less departure from the initial conformation. Better binding to the IgG and MHC molecules explains Omicron's little ability in immune invasion.
Collapse
Affiliation(s)
- Aniket Sarkar
- Post-Graduate Department of Biotechnology, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Dipannita Santra
- Post-Graduate Department of Biotechnology, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore 721102, West Bengal, India
| | - Anindya Sundar Panja
- Department of Biotechnology, Molecular Informatics Laboratory, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore, West Bengal 721102, India
| | - Smarajit Maiti
- Post-Graduate Department of Biotechnology, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore 721102, West Bengal, India; Agricure Biotech Research Society, Midnapore, 721101, India.
| |
Collapse
|
34
|
Meejun T, Srisurapanont K, Manothummetha K, Thongkam A, Mejun N, Chuleerarux N, Sanguankeo A, Phongkhun K, Leksuwankun S, Thanakitcharu J, Lerttiendamrong B, Langsiri N, Torvorapanit P, Worasilchai N, Plongla R, Hirankarn N, Nematollahi S, Permpalung N, Moonla C, Kates OS. Attenuated immunogenicity of SARS-CoV-2 vaccines and risk factors in stem cell transplant recipients: a meta-analysis. Blood Adv 2023; 7:5624-5636. [PMID: 37389818 PMCID: PMC10514108 DOI: 10.1182/bloodadvances.2023010349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/05/2023] [Accepted: 06/25/2023] [Indexed: 07/01/2023] Open
Abstract
Immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is diminished in hematopoietic stem cell transplant (HSCT) recipients. To summarize current evidence and identify risk factors for attenuated responses, 5 electronic databases were searched since database inceptions through 12 January 2023 for studies reporting humoral and/or cellular immunogenicity of SARS-CoV-2 vaccination in the HSCT population. Using descriptive statistics and random-effects models, extracted numbers of responders and pooled odds ratios (pORs) with 95% confidence intervals (CIs) for risk factors of negative immune responses were analyzed (PROSPERO: CRD42021277109). From 61 studies with 5906 HSCT recipients, after 1, 2, and 3 doses of messenger RNA (mRNA) SARS-CoV-2 vaccines, the mean antispike antibody seropositivity rates (95% CI) were 38% (19-62), 81% (77-84), and 80% (75-84); neutralizing antibody seropositivity rates were 52% (40-64), 71% (54-83), and 78% (61-89); and cellular immune response rates were 52% (39-64), 66% (51-79), and 72% (52-86). After 2 vaccine doses, risk factors (pOR; 95% CI) associated with antispike seronegativity were male recipients (0.63; 0.49-0.83), recent rituximab exposure (0.09; 0.03-0.21), haploidentical allografts (0.46; 0.22-0.95), <24 months from HSCT (0.25; 0.07-0.89), lymphopenia (0.18; 0.13-0.24), hypogammaglobulinemia (0.23; 0.10-0.55), concomitant chemotherapy (0.48; 0.29-0.78) and immunosuppression (0.18; 0.13-0.25). Complete remission of underlying hematologic malignancy (2.55; 1.05-6.17) and myeloablative conditioning (1.72; 1.30-2.28) compared with reduced-intensity conditioning were associated with antispike seropositivity. Ongoing immunosuppression (0.31; 0.10-0.99) was associated with poor cellular immunogenicity. In conclusion, attenuated humoral and cellular immune responses to mRNA SARS-CoV-2 vaccination are associated with several risk factors among HSCT recipients. Optimizing individualized vaccination and developing alternative COVID-19 prevention strategies are warranted.
Collapse
Affiliation(s)
- Tanaporn Meejun
- Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | | | - Kasama Manothummetha
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Achitpol Thongkam
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nuthchaya Mejun
- Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nipat Chuleerarux
- Department of Medicine, University of Miami/Jackson Memorial Hospital, Miami, FL
| | - Anawin Sanguankeo
- Department of Preventive and Social Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kasidis Phongkhun
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Surachai Leksuwankun
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | | | - Nattapong Langsiri
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pattama Torvorapanit
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Thai Red Cross Emerging Infectious Diseases Clinical Center, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | | | - Rongpong Plongla
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Nattiya Hirankarn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Saman Nematollahi
- Department of Medicine, University of Arizona College of Medicine, Tucson, AZ
| | - Nitipong Permpalung
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chatphatai Moonla
- Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Center of Excellence in Translational Hematology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Olivia S. Kates
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
35
|
Focosi D, Meschi S, Coen S, Iorio MC, Franchini M, Lanza M, Maggi F. Serum anti-Spike immunoglobulin G levels in random blood donors in Italy: High-titre convalescent plasma is easier than ever to procure. Vox Sang 2023; 118:794-797. [PMID: 37489640 DOI: 10.1111/vox.13498] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND AND OBJECTIVES COVID-19 convalescent plasma (CCP) has retained potency and clinical efficacy against SARS-CoV-2 and is currently of utmost value for seronegative immunocompromised patients. Since most of the effect is due to the vaccine boost of infection-elicited antibodies, there is a theoretical concern that the frequency of suitable donors is declining. MATERIALS AND METHODS In this single-institution serosurvey, we screened 599 consecutive donors attending our area in two different seasons (300 in November 2022 and 299 in February 2023) using the Abbott Alinity® anti-Spike immunoglobulin G assay. RESULTS More than 80% of random donors qualify according to the FDA criteria for high-titre CCP (>4350 AU/mL), with a stable trend. CONCLUSION Despite reduced anti-Spike vaccine boost deployment in the general population, we have shown here that high-titre CCP units are easier than ever to procure. This finding also has implications for the derivation of standard immunoglobulins, which are finally approaching the potency of hyperimmune serum and could soon represent an alternative to CCP.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Sabrina Coen
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| | - Maria Carla Iorio
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | | | - Maria Lanza
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases, Lazzaro Spallanzani IRCCS, Rome, Italy
| |
Collapse
|
36
|
Lin YCJ, Evans DH, Robbins NF, Orjuela G, Abe KT, Rathod B, Colwill K, Gingras AC, Tuite A, Yi QL, O’Brien SF, Drews SJ. Diminished Neutralization Capacity of SARS-CoV-2 Omicron BA.1 in Donor Plasma Collected from January to March 2021. Microbiol Spectr 2023; 11:e0525622. [PMID: 37289096 PMCID: PMC10434250 DOI: 10.1128/spectrum.05256-22] [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: 12/21/2022] [Accepted: 05/25/2023] [Indexed: 06/09/2023] Open
Abstract
The 50% plaque reduction neutralization assay (PRNT50) has been previously used to assess the neutralization capacity of donor plasma against wild-type and variant of concern (VOC) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Emerging data suggest that plasma with an anti-SARS-CoV-2 level of ≥2 × 104 binding antibody units/mL (BAU/mL) protects against SARS-CoV-2 Omicron BA.1 infection. Specimens were collected using a cross-sectional random sampling approach. For PRNT50 studies, 63 previously analyzed specimens by PRNT50 versus SARS-CoV-2 wild-type, Alpha, Beta, Gamma, and Delta were analyzed by PRNT50 versus Omicron BA.1. The 63 specimens plus 4,390 specimens (randomly sampled regardless of serological evidence of infection) were also tested using the Abbott SARS-CoV-2 IgG II Quant assay (anti-spike [S]; Abbott, Chicago, IL, USA; Abbott Quant assay). In the vaccinated group, the percentages of specimens with any measurable PRNT50 versus wild-type or VOC were wild type (21/25 [84%]), Alpha (19/25 [76%]), Beta (18/25 [72%]), Gamma (13/25 [52%]), Delta (19/25 [76%]), and Omicron BA.1 (9/25 [36%]). In the unvaccinated group, the percentages of specimens with any measurable PRNT50 versus wild type or VOC were wild-type SARS-CoV-2 (16/39 [41%]), Alpha (16/39 [41%]), Beta (10/39 [26%]), Gamma (9/39 [23%]), Delta (16/39 [41%]), and Omicron BA.1 (0/39) (Fisher's exact tests, vaccinated versus unvaccinated for each variant, P < 0.05). None of the 4,453 specimens tested by the Abbott Quant assay had a binding capacity of ≥2 × 104 BAU/mL. Vaccinated donors were more likely than unvaccinated donors to neutralize Omicron when assessed by a PRNT50 assay. IMPORTANCE SARS-CoV-2 Omicron emergence occurred in Canada during the period from November 2021 to January 2022. This study assessed the ability of donor plasma collected earlier (January to March 2021) to generate any neutralizing capacity against Omicron BA.1 SARS-CoV-2. Vaccinated individuals, regardless of infection status, were more likely to neutralize Omicron BA.1 than unvaccinated individuals. This study then used a semiquantitative binding antibody assay to screen a larger number of specimens (4,453) for individual specimens that might have high-titer neutralizing capacity against Omicron BA.1. None of the 4,453 specimens tested by the semiquantitative SARS-CoV-2 assay had a binding capacity suggestive of a high-titer neutralizing capacity against Omicron BA.1. These data do not imply that Canadians lacked immunity to Omicron BA.1 during the study period. Immunity to SARS-CoV-2 is complex, and there is still no wide consensus on correlation of protection to SARS-CoV-2.
Collapse
Affiliation(s)
- Yi-Chan J. Lin
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada
| | - David H. Evans
- Department of Medical Microbiology & Immunology, University of Alberta, Edmonton, Canada
| | | | | | - Kento T. Abe
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Bhavisha Rathod
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mt. Sinai Hospital, Sinai Health, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ashleigh Tuite
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Qi-Long Yi
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Sheila F. O’Brien
- Epidemiology and Surveillance, Canadian Blood Services, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Steven J. Drews
- Canadian Blood Services, Microbiology, Edmonton, Alberta, Canada
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
37
|
Metcalf RA, Cohn CS, Bakhtary S, Gniadek T, Gupta G, Harm S, Haspel RL, Hess AS, Jacobson J, Lokhandwala PM, Murphy C, Poston JN, Prochaska MT, Raval JS, Saifee NH, Salazar E, Shan H, Zantek ND, Pagano MB. Current advances in 2022: A critical review of selected topics by the Association for the Advancement of Blood and Biotherapies (AABB) Clinical Transfusion Medicine Committee. Transfusion 2023; 63:1590-1600. [PMID: 37403547 DOI: 10.1111/trf.17475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/16/2023] [Indexed: 07/06/2023]
Abstract
BACKGROUND The Association for the Advancement of Blood and Biotherapies Clinical Transfusion Medicine Committee (CTMC) composes a summary of new and important advances in transfusion medicine (TM) on an annual basis. Since 2018, this has been assembled into a manuscript and published in Transfusion. STUDY DESIGN AND METHODS CTMC members selected original manuscripts relevant to TM that were published electronically and/or in print during calendar year 2022. Papers were selected based on perceived importance and/or originality. References for selected papers were made available to CTMC members to provide feedback. Members were also encouraged to identify papers that may have been omitted initially. They then worked in groups of two to three to write a summary for each new publication within their broader topic. Each topic summary was then reviewed and edited by two separate committee members. The final manuscript was assembled by the first and senior authors. While this review is extensive, it is not a systematic review and some publications considered important by readers may have been excluded. RESULTS For calendar year 2022, summaries of key publications were assembled for the following broader topics within TM: blood component therapy; infectious diseases, blood donor testing, and collections; patient blood management; immunohematology and genomics; hemostasis; hemoglobinopathies; apheresis and cell therapy; pediatrics; and health care disparities, diversity, equity, and inclusion. DISCUSSION This Committee Report reviews and summarizes important publications and advances in TM published during calendar year 2022, and maybe a useful educational tool.
Collapse
Affiliation(s)
- Ryan A Metcalf
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Claudia S Cohn
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sara Bakhtary
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | | | - Gaurav Gupta
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sarah Harm
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Richard L Haspel
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Aaron S Hess
- Departments of Anesthesiology and Pathology & Laboratory Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Jessica Jacobson
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, USA
| | - Parvez M Lokhandwala
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Colin Murphy
- TriCore Reference Laboratories, Albuquerque, New Mexico, USA
| | - Jacqueline N Poston
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
- Department of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Micah T Prochaska
- Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Jay S Raval
- Department of Pathology, University of New Mexico, Albuquerque, New Mexico, USA
| | | | - Eric Salazar
- Department of Pathology, UT Health San Antonio, San Antonio, Texas, USA
| | - Hua Shan
- Department of Pathology, Stanford University, Palo Alto, California, USA
| | - Nicole D Zantek
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Monica B Pagano
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
38
|
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: 73] [Impact Index Per Article: 36.5] [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
|
39
|
Werbel WA, Weld ED, Advani SD, Patel PK, Sundaram ME, Phadke VK. Your Outpatient has Coronavirus Disease 2019: What Are the Treatment Options in the Current Severe Acute Respiratory Syndrome Coronavirus 2 Variant Climate? Clin Infect Dis 2023; 77:32-37. [PMID: 36999905 PMCID: PMC10320072 DOI: 10.1093/cid/ciad178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/01/2023] Open
Abstract
Mutations accumulated by novel Severe Acute Respiratory Syndrome Coronavirus 2 Omicron sublineages contribute to evasion of previously effective monoclonal antibodies for treatment or prevention of Coronavirus Disease 2019 (COVID-19). Other authorized or approved antiviral drugs such as nirmatrelvir/ritonavir, remdesivir, and molnupiravir are, however, predicted to maintain activity against these sublineages and are key tools to reduce severe COVID-19 outcomes in vulnerable populations. A stepwise approach may be taken to target the appropriate antiviral drug to the appropriate patient, beginning with identifying whether a patient is at high risk for hospitalization or other complications of COVID-19. Among higher risk individuals, patient profile (including factors such as age, organ function, and comedications) and antiviral drug access inform suitable antiviral drug selection. When applied in targeted fashion, these therapies serve as a complement to vital ongoing nonpharmaceutical interventions and vaccination strategies that reduce morbidity and maximize protection against COVID-19.
Collapse
Affiliation(s)
- William A Werbel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ethel D Weld
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sonali D Advani
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Payal K Patel
- Division of Infectious Diseases and Clinical Epidemiology, Intermountain Healthcare, Murray, Utah, USA
| | - Maria E Sundaram
- Center for Clinical Epidemiology and Population Health, Marshfield Clinical Research Institute, Marshfield, Wisconsin, USA
| | - Varun K Phadke
- Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
40
|
Chenchula S, Amerneni KC, Ghanta MK, Padmavathi R, Chandra MB, Adusumilli MB, Chavan M, Mudda S, Gupta R, Lakhawat B. Clinical virology and effect of Covid-19 vaccination and monoclonal antibodies against highly infectious SARS- CoV-2 omicron sub variant BF.7 (BA.5.2.1.7): A systematic review. Virology 2023; 584:38-43. [PMID: 37229914 PMCID: PMC10197433 DOI: 10.1016/j.virol.2023.04.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 05/27/2023]
Abstract
Over time, the SARS-CoV-2 virus has acquired several genetic mutations, particularly on the receptor-binding domain (RBD) spike glycoprotein. The Omicron variant is highly infectious, with enhanced immune escape activity, and has given rise to various sub-lineages due to mutations. However, there has been a sudden increase in COVID-19 reports of the Omicron subvariant BF.7 (BA.2.75.2), which has the highest number of reported cases, accounting for 76.2% of all cases worldwide. Hence, the present systematic review aimed to understand the viral mutations and factors associated with the increase in the reports of COVID-19 cases and to assess the effectiveness of vaccines and mAbs against the novel Omicron variant BF.7. The R346T mutation on the spike glycoprotein RBD might be associated with increased infection rates, severity, and resistance to vaccines and mAbs. Booster doses of COVID-19 vaccination with bivalent mRNA booster vaccine shots are effective in curtailing infections and decreasing the severity and mortality by enhancing the neutralizing antibodies (Abs) against the emerging Omicron subvariants of SARS-CoV-2, including BF.7 and future VOCs.
Collapse
Affiliation(s)
- Santenna Chenchula
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Bhopal, Madhya Pradesh, India.
| | | | - Mohan Krishna Ghanta
- Department of Pharmacology, MVJ Medical College and Research Hospital, Bangalore, Karnataka, India.
| | - R Padmavathi
- SVS Medical College and Hospital, Telangana, India.
| | | | | | - Madhavrao Chavan
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Mangalagiri, Andhra Pradesh, India.
| | - Sofia Mudda
- All India Institute of Medical Sciences, Bhopal, India.
| | - Rupesh Gupta
- Department of Internal Medicine, Government Medical College, Shahdol, Madhya Pradesh, India.
| | - Bhawna Lakhawat
- Department of Pharmacology, All India Institute of Medical Sciences, Bhopal, India.
| |
Collapse
|
41
|
Bäuerl C, Zulaica J, Rusu L, Moreno AR, Pérez-Cano FJ, Lerin C, Mena-Tudela D, Aguilar-Camprubí L, Parra-Llorca A, Martínez-Costa C, Geller R, Collado MC. Assessment of SARS-CoV-2 neutralizing antibody titers in breastmilk from convalescent and vaccinated mothers. iScience 2023; 26:106802. [PMID: 37197591 PMCID: PMC10158041 DOI: 10.1016/j.isci.2023.106802] [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: 02/16/2023] [Revised: 03/29/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Breastmilk contains antibodies that could protect breastfed infants from infections. In this work, we examined if antibodies in breastmilk could neutralize SARS-CoV-2 in 84 breastmilk samples from women that were either vaccinated (Comirnaty, mRNA-1273, or ChAdOx1), infected with SARS-CoV-2, or both infected and vaccinated. The neutralization capacity of these sera was tested using pseudotyped vesicular stomatitis virus carrying either the Wuhan-Hu-1, Delta, or BA.1 Omicron spike proteins. We found that natural infection resulted in higher neutralizing titers and that neutralization correlated positively with levels of immunoglobulin A in breastmilk. In addition, significant differences in the capacity to produce neutralizing antibodies were observed between both mRNA-based vaccines and the adenovirus-vectored ChAdOx1 COVID-19 vaccine. Overall, our results indicate that breastmilk from naturally infected women or those vaccinated with mRNA-based vaccines contains SARS-CoV-2 neutralizing antibodies that could potentially provide protection to breastfed infants from infection.
Collapse
Affiliation(s)
- Christine Bäuerl
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
| | - Joao Zulaica
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Luciana Rusu
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Alicia Rodríguez Moreno
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science and Institute of Research in Nutrition and Food Safety (INSA), University of Barcelona (UB), 08028 Barcelona, Spain
| | - Carles Lerin
- Endocrinology department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Desirée Mena-Tudela
- Department of Nursing, Nursing Research Group, Universitat Jaume I, Castellón, Spain
| | | | - Anna Parra-Llorca
- Health Research Institute La Fe, Neonatal Research Group, Spain and University and Polytechnic Hospital La Fe, Division of Neonatology, 46026 Valencia, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain. Nutrition Research Group of INCLIVA, 46010 Valencia, Spain
| | - Ron Geller
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
| | - MilkCORONA study team
- Institute of Agrochemistry and Food Technology- National Research Council (IATA-CSIC), 46980 Paterna, Valencia, Spain
- Institute for Integrative Systems Biology (ISysBio), University of Valencia-CSIC, 46980 Paterna, Valencia, Spain
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science and Institute of Research in Nutrition and Food Safety (INSA), University of Barcelona (UB), 08028 Barcelona, Spain
- Endocrinology department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Nursing, Nursing Research Group, Universitat Jaume I, Castellón, Spain
- LactApp Women Health, Barcelona, Spain
- Health Research Institute La Fe, Neonatal Research Group, Spain and University and Polytechnic Hospital La Fe, Division of Neonatology, 46026 Valencia, Spain
- Department of Pediatrics, Hospital Clínico Universitario, University of Valencia, Spain. Nutrition Research Group of INCLIVA, 46010 Valencia, Spain
| |
Collapse
|
42
|
Tang J, Liu H, Wang Q, Gu X, Wang J, Li W, Luo Y, Li Y, Deng L, Luo Y, Du X, Tan D, Fu X, Chen X. Predictors of high SARS-CoV-2 immunoglobulin G titers in COVID-19 convalescent whole-blood donors: a cross-sectional study in China. Front Immunol 2023; 14:1191479. [PMID: 37388736 PMCID: PMC10303911 DOI: 10.3389/fimmu.2023.1191479] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/02/2023] [Indexed: 07/01/2023] Open
Abstract
Background Demographic information has been shown to help predict high antibody titers of COVID-19 convalescent plasma (CCP) in CCP donors. However, there is no research on the Chinese population and little evidence on whole-blood donors. Therefore, we aimed to investigate these associations among Chinese blood donors after SARS-CoV-2 infection. Methods In this cross-sectional study, 5,064 qualified blood donors with confirmed or suspected SARS-CoV-2 infection completed a self-reported questionnaire and underwent tests of SARS-CoV-2 Immunoglobulin G (IgG) antibody and ABO blood type. Logistic regression models were used to calculate odds ratios (ORs) for high SARS-CoV-2 IgG titers according to each factor. Results Totally, 1,799 participants (with SARS-CoV-2 IgG titers≥1:160) had high-titer CCPs. Multivariable analysis showed that a 10-year increment in age and earlier donation were associated with higher odds of high-titer CCP, while medical personnel was associated with lower odds. The ORs (95% CIs) of high-titer CCP were 1.17 (1.10-1.23, p< 0.001) and 1.41 (1.25-1.58, p< 0.001) for each 10-year increment in age and earlier donation, respectively. The OR of high-titer CCP was 0.75 (0.60-0.95, p = 0.02) for medical personnel. Female early donors were associated with increased odds of high-titer CCP, but this association was insignificant for later donors. Donating after 8 weeks from the onset was associated with decreased odds of having high-titer CCP compared to donating within 8 weeks from the onset, and the HR was 0.38 (95% CI: 0.22-0.64, p <0.001). There was no significant association between ABO blood type or race and the odds of high-titer CCP. Discussion Older age, earlier donation, female early donors, and non-medical-related occupations are promising predictors of high-titer CCP in Chinese blood donors. Our findings highlight the importance of CCP screening at the early stage of the pandemic.
Collapse
Affiliation(s)
- Jingyun Tang
- Blood Research Laboratory, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Humin Liu
- Department of Blood Testing, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Qing Wang
- Department of Blood Collection, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Xiaobo Gu
- Department of Blood Collection, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Jia Wang
- Department of Blood Collection, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Wenjun Li
- Department of Blood Testing, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Yinglan Luo
- Department of Blood Testing, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Yan Li
- Department of Blood Collection, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Lan Deng
- Department of Blood Collection, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Yue Luo
- Blood Research Laboratory, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Xinman Du
- Blood Research Laboratory, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Donglin Tan
- Department of Blood Processing, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Xuemei Fu
- Blood Research Laboratory, Chengdu Blood Center, Chengdu, Sichuan, China
| | - Xue Chen
- Blood Research Laboratory, Chengdu Blood Center, Chengdu, Sichuan, China
| |
Collapse
|
43
|
Bloch EM, Focosi D, Shoham S, Senefeld J, Tobian AAR, Baden LR, Tiberghien P, Sullivan DJ, Cohn C, Dioverti V, Henderson JP, So-Osman C, Juskewitch JE, Razonable RR, Franchini M, Goel R, Grossman BJ, Casadevall A, Joyner MJ, Avery RK, Pirofski LA, Gebo KA. Guidance on the Use of Convalescent Plasma to Treat Immunocompromised Patients With Coronavirus Disease 2019. Clin Infect Dis 2023; 76:2018-2024. [PMID: 36740590 PMCID: PMC10249987 DOI: 10.1093/cid/ciad066] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/23/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) convalescent plasma (CCP) is a safe and effective treatment for COVID-19 in immunocompromised (IC) patients. IC patients have a higher risk of persistent infection, severe disease, and death from COVID-19. Despite the continued clinical use of CCP to treat IC patients, the optimal dose, frequency/schedule, and duration of CCP treatment has yet to be determined, and related best practices guidelines are lacking. A group of individuals with expertise spanning infectious diseases, virology and transfusion medicine was assembled to render an expert opinion statement pertaining to the use of CCP for IC patients. For optimal effect, CCP should be recently and locally collected to match circulating variant. CCP should be considered for the treatment of IC patients with acute and protracted COVID-19; dosage depends on clinical setting (acute vs protracted COVID-19). CCP containing high-titer severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies, retains activity against circulating SARS-CoV-2 variants, which have otherwise rendered monoclonal antibodies ineffective.
Collapse
Affiliation(s)
- Evan M Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jonathon Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lindsey R Baden
- Department of Infectious Diseases, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Pierre Tiberghien
- Etablissement Français du Sang, La Plaine-St-Denis and Université de Franche-Comté, Besançon, France
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Claudia Cohn
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Veronica Dioverti
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jeffrey P Henderson
- Departments of Internal Medicine (Division of Infectious Diseases) and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Cynthia So-Osman
- Department Transfusion Medicine, Division Blood Bank, Sanquin Blood Supply Foundation, Amsterdam, The Netherlands
- Department Haematology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Justin E Juskewitch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester campus, Minnesota, USA
| | - Raymund R Razonable
- Department of Internal Medicine, Division of Infectious Diseases, Mayo Clinic, Rochester, Minnesota, USA
| | - Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Ruchika Goel
- Division of Hematology/Oncology, Simmons Cancer Institute at SIU School of Medicine and Mississippi Valley Regional Blood Center, Springfield, Illinois, USA
| | - Brenda J Grossman
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin K Avery
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Liise-anne Pirofski
- Department of Medicine, Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kelly A Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
44
|
Blickstein D, Izak M, Filipovich-Rimon T, Garach-Jehoshua O, Rahimi-Levene N, Shinar E, Hamad RA, Bar-Chaim A, Koren-Michowitz M. Antiphospholipid antibodies in convalescent plasma of donors recovered from mild COVID-19 infection. Vox Sang 2023. [PMID: 37191363 DOI: 10.1111/vox.13439] [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: 02/08/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND AND OBJECTIVES Passive immunization by the infusion of convalescent plasma (CP) obtained from patients who have recently recovered from COVID-19, thus having antibodies to severe acute respiratory syndrome coronavirus 2, is a potential strategy to reduce the severity of illness. A high prevalence of antiphospholipid antibodies (APLA) in patients with COVID-19 has been reported during the pandemic, raising a concern whether the use of CP could increase the risk of thrombosis in transfused patients. We aimed to evaluate the prevalence of APLA in COVID-19 CP (CCP) in order to assess the potential prothrombotic influence of transfused CCP to COVID-19 patients. MATERIALS AND METHODS We studied the prevalence of APLA in 122 CCP samples collected from healthy donors who recovered from mild-COVID-19 at two time periods: September 2020-January 2021 (defined as 'early period' samples) and April-May 2021 (defined as 'late period' samples). Thirty-four healthy subjects unexposed to COVID-19 were used as controls. RESULTS APLA were present in 7 of 122 (6%) CCP samples. One donor had anti-β2-glycoprotein 1(anti-β2GP1) IgG, one had anti-β2GP1 IgM and five had lupus anticoagulant (LAC) using silica clotting time (SCT), all in 'late period' donors. In the control group, one subject had anti-β2GP1 IgG, two had LAC using dilute Russell viper venom time (dRVVT) and four had LAC SCT (both LAC SCT and LAC dRVVT in one subject). CONCLUSION The low prevalence of APLA in CCP donors reassures the safety of CCP administration to patients with severe COVID-19.
Collapse
Affiliation(s)
- Dorit Blickstein
- Department of Hematology, Shamir Medical Center, Be'er Ya'akov, Israel
| | - Marina Izak
- Magen David Adom National Blood Services, Ramat Gan, Israel
| | | | | | | | - Eilat Shinar
- Magen David Adom National Blood Services, Ramat Gan, Israel
| | - Ramzia Abu Hamad
- Clinical Chemistry Laboratory, Shamir Medical Center, Be'er Ya'akov, Israel
| | - Adina Bar-Chaim
- Clinical Laboratories, Shamir Medical Center, Be'er Ya'akov, Israel
| | - Maya Koren-Michowitz
- Department of Hematology, Shamir Medical Center, Be'er Ya'akov, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
45
|
Casadevall A, Joyner MJ, Pirofski LA, Senefeld JW, Shoham S, Sullivan D, Paneth N, Focosi D. Convalescent plasma therapy in COVID-19: Unravelling the data using the principles of antibody therapy. Expert Rev Respir Med 2023:1-15. [PMID: 37129285 DOI: 10.1080/17476348.2023.2208349] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
INTRODUCTION When the COVID-19 pandemic struck no specific therapies were available and many turned to COVID-19 convalescent plasma (CCP), a form of antibody therapy. The literature provides mixed evidence for CCP efficacy. AREAS COVERED PubMed was searched using the words COVID-19 and convalescent plasma and individual study designs were evaluated for adherence to the three principles of antibody therapy, i.e. that plasma 1) contain specific antibody; 2) have enough specific antibody to mediate a biological effect; and 3) be administered early in the course of disease. Using this approach, a diverse and seemingly contradictory collection of clinical findings was distilled into a consistent picture whereby CCP was effective when used according to the above principles of antibody therapy. In addition, CCP therapy in immunocompromised patients is useful at any time in the course of disease. EXPERT OPINION CCP is safe and effective when used appropriately. Today, most of humanity has some immunity to SARS-CoV-2 from vaccines and infection, which has lessened the need for CCP in the general population. However, COVID-19 in immunocompromised patients is a major therapeutic challenge, and with the deauthorization of all SARS-CoV-2-spike protein-directed monoclonal antibodies, CCP is the only antibody therapy available for this population.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nigel Paneth
- Departments of Epidemiology & Biostatistics and Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
46
|
Sullivan DJ, Franchini M, Senefeld JW, Joyner MJ, Casadevall A, Focosi D. Plasma after both SARS-CoV-2 boosted vaccination and COVID-19 potently neutralizes BQ.1.1 and XBB.1. J Gen Virol 2023; 104:001854. [PMID: 37167085 PMCID: PMC10336427 DOI: 10.1099/jgv.0.001854] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Recent 2022 SARS-CoV-2 Omicron variants, have acquired resistance to most neutralizing anti-Spike monoclonal antibodies authorized, and the BQ.1.* sublineages are notably resistant to all authorized monoclonal antibodies. Polyclonal antibodies from individuals both vaccinated and recently recovered from Omicron COVID-19 (VaxCCP) could retain new Omicron neutralizing activity. Here we reviewed BQ.1.* virus neutralization data from 920 individual patient samples from 43 separate cohorts defined by boosted vaccinations (Vax) with or without recent Omicron COVID-19, as well as infection without vaccination (CCP) to determine level of BQ.1.* neutralizing antibodies and percent of plasma samples with neutralizing activity. More than 90 % of the plasma samples from individuals in the recently (within 6 months) boosted VaxCCP study cohorts neutralized BQ.1.1, and BF.7 with 100 % neutralization of WA-1, BA.4/5, BA.4.6 and BA.2.75. The geometric mean of the geometric mean 50 % neutralizing titres (GM (GMT50) were 314, 78 and 204 for BQ.1.1, XBB.1 and BF.7, respectively. Compared to VaxCCP, plasma sampled from COVID-19 naïve subjects who also recently (within 6 months) received at least a third vaccine dose had about half of the GM (GMT50) for all viral variants. Boosted VaxCCP characterized by either recent vaccine dose or infection event within 6 months represents a robust, variant-resilient, neutralizing antibody source against the new Omicron BQ.1.1, XBB.1 and BF.7 variants.
Collapse
Affiliation(s)
- David J. Sullivan
- Johns Hopkins Bloomberg School of Public Health and School of Medicine, Baltimore, MD 21218, USA
| | - Massimo Franchini
- Division of Transfusion Medicine, Carlo Poma Hospital, 46100 Mantua, Italy
| | - Jonathon W. Senefeld
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Michael J. Joyner
- Department of Anesthesiology & Perioperative Medicine, Mayo Clinic, Rochester, MN 55902, USA
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health and School of Medicine, Baltimore, MD 21218, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| |
Collapse
|
47
|
Shoham S, Batista C, Ben Amor Y, Ergonul O, Hassanain M, Hotez P, Kang G, Kim JH, Lall B, Larson HJ, Naniche D, Sheahan T, Strub-Wourgaft N, Sow SO, Wilder-Smith A, Yadav P, Bottazzi ME. Vaccines and therapeutics for immunocompromised patients with COVID-19. EClinicalMedicine 2023; 59:101965. [PMID: 37070102 PMCID: PMC10091856 DOI: 10.1016/j.eclinm.2023.101965] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/19/2023] Open
Abstract
The COVID-19 pandemic has disproportionately impacted immunocompromised patients. This diverse group is at increased risk for impaired vaccine responses, progression to severe disease, prolonged hospitalizations and deaths. At particular risk are people with deficiencies in lymphocyte number or function such as transplant recipients and those with hematologic malignancies. Such patients' immune responses to vaccination and infection are frequently impaired leaving them more vulnerable to prolonged high viral loads and severe complications of COVID-19. Those in turn, have implications for disease progression and persistence, development of immune escape variants and transmission of infection. Data to guide vaccination and treatment approaches in immunocompromised people are generally lacking and extrapolated from other populations. The large clinical trials leading to authorisation and approval of SARS-CoV-2 vaccines and therapeutics included very few immunocompromised participants. While experience is accumulating, studies focused on the special circumstances of immunocompromised patients are needed to inform prevention and treatment approaches.
Collapse
Affiliation(s)
- Shmuel Shoham
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carolina Batista
- Médecins Sans Frontières, Rio de Janeiro, Brazil
- Baraka Impact Finance, Geneva, Switzerland
| | - Yanis Ben Amor
- Center for Sustainable Development, Columbia University, New York, NY, USA
| | - Onder Ergonul
- Koc University Research Center for Infectious Diseases, Istanbul, Turkey
| | - Mazen Hassanain
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Peter Hotez
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Bhavna Lall
- University of Houston Tilman J. Fertitta Family College of Medicine, Houston, TX, USA
| | | | - Denise Naniche
- ISGlobal, Barcelona Institute for Global Health, Hospital Clinic, University of Barcelona, Spain
| | - Timothy Sheahan
- University of North Carolina, Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Nathalie Strub-Wourgaft
- ISGlobal, Barcelona Institute for Global Health, Hospital Clinic, University of Barcelona, Spain
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
| | - Samba O. Sow
- Center for Vaccine Development, Bamako, Mali
- University of Maryland, MD, USA
| | - Annelies Wilder-Smith
- London School of Hygiene & Tropical Medicine, London, UK
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| | - Prashant Yadav
- Center for Global Development, Washington, DC, USA
- Harvard Medical School, Boston, MA, USA
- Technology and Operations Management, INSEAD, Fontainebleau, France
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
| | - Lancet Commission on COVID-19 Vaccines and Therapeutics Task Force
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Médecins Sans Frontières, Rio de Janeiro, Brazil
- Baraka Impact Finance, Geneva, Switzerland
- Center for Sustainable Development, Columbia University, New York, NY, USA
- Koc University Research Center for Infectious Diseases, Istanbul, Turkey
- College of Medicine, King Saud University, Riyadh, Saudi Arabia
- Texas Children's Hospital Center for Vaccine Development, Baylor College of Medicine, Houston, TX, USA
- Christian Medical College, Vellore, India
- International Vaccine Institute, Seoul, South Korea
- University of Houston Tilman J. Fertitta Family College of Medicine, Houston, TX, USA
- London School of Hygiene & Tropical Medicine, London, UK
- ISGlobal, Barcelona Institute for Global Health, Hospital Clinic, University of Barcelona, Spain
- University of North Carolina, Gillings School of Global Public Health, Chapel Hill, NC, USA
- Drugs for Neglected Diseases Initiative, Geneva, Switzerland
- Center for Vaccine Development, Bamako, Mali
- University of Maryland, MD, USA
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
- Center for Global Development, Washington, DC, USA
- Harvard Medical School, Boston, MA, USA
- Technology and Operations Management, INSEAD, Fontainebleau, France
| |
Collapse
|
48
|
Desmarets M, Hoffmann S, Vauchy C, Rijnders BJA, Toussirot E, Durrbach A, Körper S, Schrezenmeier E, van der Schoot CE, Harvala H, Brunotte G, Appl T, Seifried E, Tiberghien P, Bradshaw D, Roberts DJ, Estcourt LJ, Schrezenmeier H. Early, very high-titre convalescent plasma therapy in clinically vulnerable individuals with mild COVID-19 (COVIC-19): protocol for a randomised, open-label trial. BMJ Open 2023; 13:e071277. [PMID: 37105693 PMCID: PMC10151238 DOI: 10.1136/bmjopen-2022-071277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/29/2023] Open
Abstract
INTRODUCTION COVID-19 convalescent plasma (CCP) is a possible treatment option for COVID-19. A comprehensive number of clinical trials on CCP efficacy have already been conducted. However, many aspects of CCP treatment still require investigations: in particular (1) Optimisation of the CCP product, (2) Identification of the patient population in need and most likely to benefit from this treatment approach, (3) Timing of administration and (4) CCP efficacy across viral variants in vivo. We aimed to test whether high-titre CCP, administered early, is efficacious in preventing hospitalisation or death in high-risk patients. METHODS AND ANALYSIS COVIC-19 is a multicentre, randomised, open-label, adaptive superiority phase III trial comparing CCP with very high neutralising antibody titre administered within 7 days of symptom onset plus standard of care versus standard of care alone. We will enrol patients in two cohorts of vulnerable patients [(1) elderly 70+ years, or younger with comorbidities; (2) immunocompromised patients]. Up to 1020 participants will be enrolled in each cohort (at least 340 with a sample size re-estimation after reaching 102 patients). The primary endpoint is the proportion of participants with (1) Hospitalisation due to progressive COVID-19, or (2) Who died by day 28 after randomisation. Principal analysis will follow the intention-to-treat principle. ETHICS AND DISSEMINATION Ethical approval has been granted by the University of Ulm ethics committee (#41/22) (lead ethics committee for Germany), Comité de protection des personnes Sud-Est I (CPP Sud-Est I) (#2022-A01307-36) (ethics committee for France), and ErasmusMC ethics committee (#MEC-2022-0365) (ethics committee for the Netherlands). Signed informed consent will be obtained from all included patients. The findings will be published in peer-reviewed journals and presented at relevant stakeholder conferences and meetings. TRIAL REGISTRATION Clinical Trials.gov (NCT05271929), EudraCT (2021-006621-22).
Collapse
Affiliation(s)
- Maxime Desmarets
- Centre d'Investigation Clinique Inserm CIC1431, CHU Besançon, Besançon, Bourgogne Franche-Comté, France
- UMR 1098 Right, Inserm, Établissement Français du Sang, Université de Franche-Comté, Besançon, Bourgogne Franche-Comté, France
| | - Simone Hoffmann
- Blood Transfusion Service Baden-Württemberg-Hessen, German Red Cross, Ulm, Baden-Württemberg, Germany
| | - Charline Vauchy
- Centre d'Investigation Clinique Inserm CIC1431, CHU Besançon, Besançon, Bourgogne Franche-Comté, France
- UMR 1098 Right, Inserm, Établissement Français du Sang, Université de Franche-Comté, Besançon, Bourgogne Franche-Comté, France
| | - Bart J A Rijnders
- University Medical Center, Erasmus MC, Rotterdam, Zuid-Holland, Netherlands
| | - Eric Toussirot
- Centre d'Investigation Clinique Inserm CIC1431, CHU Besançon, Besançon, Bourgogne Franche-Comté, France
- UMR 1098 Right, Inserm, Établissement Français du Sang, Université de Franche-Comté, Besançon, Bourgogne Franche-Comté, France
| | - Antoine Durrbach
- Department of Nephrology, AP-HP Hôpital Henri Mondor, Créteil, Île-de-France, France
| | - Sixten Körper
- Blood Transfusion Service Baden-Württemberg-Hessen, German Red Cross, Ulm, Baden-Württemberg, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Baden-Württemberg, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research, Amsterdam, Noord-Holland, Netherlands
| | - Heli Harvala
- Microbiology Services, NHS Blood and Transplant, Colindale, London, UK
| | - Gaëlle Brunotte
- Centre d'investigation clinique Inserm CIC1431, CHU Besançon, Besançon, France
| | - Thomas Appl
- Blood Transfusion Service Baden-Württemberg-Hessen, German Red Cross, Ulm, Baden-Württemberg, Germany
| | - Erhard Seifried
- Blood Transfusion Service Baden-Württemberg-Hessen, German Red Cross, Ulm, Baden-Württemberg, Germany
| | - Pierre Tiberghien
- UMR 1098 Right, Inserm, Établissement Français du Sang, Université de Franche-Comté, Besançon, Bourgogne Franche-Comté, France
- Etablissement Francais du Sang, La Plaine Saint-Denis, Île-de-France, France
| | - Daniel Bradshaw
- Virus Reference Department, UK Health Security Agency, London, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, Oxfordshire, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Lise J Estcourt
- NHS Blood and Transplant, Oxford, Oxfordshire, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Hubert Schrezenmeier
- Blood Transfusion Service Baden-Württemberg-Hessen, German Red Cross, Ulm, Baden-Württemberg, Germany
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, Ulm, Baden-Württemberg, Germany
| |
Collapse
|
49
|
Song Q, Zhao L, Mai W, Xia D, Ding W, Zhou X, Deng M, Lei Y, Chen L, Li Y, Mai X, Zhang L, Chen Z, Qin Y, Ren R, Wei W, Ji T. Handheld NIR-to-NIR Platform for on-site evaluating protective neutralizing antibody against SARS-CoV-2 ancestral strain and Omicron variant after vaccination or infection. Biosens Bioelectron 2023; 234:115353. [PMID: 37120945 PMCID: PMC10127741 DOI: 10.1016/j.bios.2023.115353] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/14/2023] [Accepted: 04/24/2023] [Indexed: 05/02/2023]
Abstract
Lateral flow assays (LFAs) are promising points-of-care tests, playing a vital role in diseases screening, diagnosis and surveillance. However, development of portable, cheap, and smart LFAs platform for sensitive and accurate quantification of disease biomarkers in complex media is challenging. Here, a cheap handheld device was developed to realize on-site detection of disease biomarkers by Nd3+/Yb3+ co-doped near-infrared (NIR)-to-NIR downconversion nanoparticles (DCNPs) based LFA. Its sensitivity is at least 8-fold higher for detecting NIR light signal from Nd3+/Yb3+ co-doped nanoparticles than conventional expensive InGaAs camera based detection platform. Additionally, we enhance NIR quantum yield of Nd3+/Yb3+ co-doped nanoparticles up to 35.5% via simultaneous high dopant of sensitizer ions Nd3+ and emitter ions Yb3+. Combination of NIR-to-NIR handheld detection device and ultra-bright NIR emitting NaNbF4:Yb60%@NaLuF4 nanoparticle probe allows the detection sensitivity of SARS-CoV-2 ancestral strain and Omicron variants specific neutralizing antibodies LFA up to the level of commercial enzyme linked immunosorbent assay kit. Furthermore, by this robust method, enhanced neutralizing antibodies against SARS-CoV-2 ancestral strain and Omicron variants are observed in healthy participants with Ad5-nCoV booster on top of two doses of inactivated vaccine. This NIR-to-NIR handheld platform provides a promising strategy for on-site evaluating protective humoral immunity after SARS-CoV-2 vaccination or infection.
Collapse
Affiliation(s)
- Qingwei Song
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China; MOE & Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, PR China
| | - Lei Zhao
- MOE & Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, PR China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Daoyu Xia
- MOE & Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, PR China
| | - Weidong Ding
- Shenzhen Highcreation Technology Co., Ltd, Shenzhen, 518122, PR China
| | - Xiaomian Zhou
- Guangzhou Biotron Technology Co, Ltd, Guangzhou, 510530, PR China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Yantao Li
- MOE & Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, PR China
| | - Xiaorui Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Lei Zhang
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, 510260, PR China
| | - Zheng Chen
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, 510260, PR China
| | - Yiru Qin
- Guangdong Provincial Key Laboratory of Occupational Disease Prevention and Treatment, Guangdong Province Hospital for Occupational Disease Prevention and Treatment, Guangzhou, 510300, PR China
| | - Ruiwen Ren
- Guangdong Arbovirus Diseases Emergency Technology Research Center, Guangzhou, 510507, PR China
| | - Wei Wei
- MOE & Guangdong Provincial Key Laboratory of Laser Life Science, Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou, 510631, PR China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| |
Collapse
|
50
|
McDyer JF, Azimpouran M, Durkalski-Mauldin VL, Clevenger RG, Yeatts SD, Deng X, Barsan W, Silbergleit R, El Kassar N, Popescu I, Dimitrov D, Li W, Lyons EJ, Lieber SC, Stone M, Korley FK, Callaway CW, Dumont LJ, Norris PJ. COVID-19 convalescent plasma boosts early antibody titer and does not influence the adaptive immune response. JCI Insight 2023; 8:e167890. [PMID: 36862515 PMCID: PMC10174456 DOI: 10.1172/jci.insight.167890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Multiple randomized, controlled clinical trials have yielded discordant results regarding the efficacy of convalescent plasma in outpatients, with some showing an approximately 2-fold reduction in risk and others showing no effect. We quantified binding and neutralizing antibody levels in 492 of the 511 participants from the Clinical Trial of COVID-19 Convalescent Plasma in Outpatients (C3PO) of a single unit of COVID-19 convalescent plasma (CCP) versus saline infusion. In a subset of 70 participants, peripheral blood mononuclear cells were obtained to define the evolution of B and T cell responses through day 30. Binding and neutralizing antibody responses were approximately 2-fold higher 1 hour after infusion in recipients of CCP compared with saline plus multivitamin, but levels achieved by the native immune system by day 15 were almost 10-fold higher than those seen immediately after CCP administration. Infusion of CCP did not block generation of the host antibody response or skew B or T cell phenotype or maturation. Activated CD4+ and CD8+ T cells were associated with more severe disease outcome. These data show that CCP leads to a measurable boost in anti-SARS-CoV-2 antibodies but that the boost is modest and may not be sufficient to alter disease course.
Collapse
Affiliation(s)
| | | | | | | | - Sharon D. Yeatts
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - William Barsan
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Robert Silbergleit
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Nahed El Kassar
- National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Iulia Popescu
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Wei Li
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | - Frederick K. Korley
- Department of Emergency Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | | | - Larry J. Dumont
- Vitalant Research Institute, San Francisco, California, USA
- University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Philip J. Norris
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, UCSF, San Francisco, California, USA
| | | |
Collapse
|