1
|
Thomas SM, Harrington RA, Yancy CW, Nugent D, Erzurum S, Bernard GR, Cushman M, Hochman JS, Ridker PM, Ortel TL, Collins SP, Callaway CW, Nolen TL, Womack KN, Brown SM, Gelijns A, Geraci M, Ginde AA, Key NS, Krishnan JA, LaVange L, Wisniewski SR, Berdan L, Punturieri A, Goff DC, Patterson AP. Lessons Learned from National Heart, Lung, and Blood Institute Covid-19 Clinical Trials. NEJM EVIDENCE 2024; 3:EVIDctcs2300291. [PMID: 39437132 DOI: 10.1056/evidctcs2300291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
AbstractIn response to the Covid-19 pandemic, the National Heart, Lung, and Blood Institute launched five multisite clinical trials testing candidate host tissue-directed medical interventions to hasten recovery, improve function, and reduce morbidity and mortality. Speed, flexibility, and collaboration were essential. This article from the Steering and Executive committees describes the Collaborating Network of Networks for Evaluating Covid-19 and Therapeutic Strategies (CONNECTS) research program that enrolled 6690 participants and evaluated 18 intervention strategies using 10 molecular agents across the care continuum (outpatient, inpatient, and post discharge), and reports lessons learned from this initiative. Successes include rapid trial execution through collaboration and adaptive platform designs. Challenges that impeded efficiency included time required to execute subcontracts, constraints on clinical research workforce, and limited research infrastructure in nonacademic settings.
Collapse
Affiliation(s)
| | | | - Clyde W Yancy
- Northwestern University Feinberg School of Medicine, Chicago
| | | | | | | | | | - Judith S Hochman
- New York University Grossman School of Medicine, NYU Langone Health, New York
| | | | | | - Sean P Collins
- Vanderbilt University Medical Center and Veterans Affairs Tennessee Valley Healthcare System, Geriatric Research, Education and Clinical Center (GRECC), Nashville
| | | | | | | | | | | | | | - Adit A Ginde
- University of Colorado School of Medicine, Aurora
| | - Nigel S Key
- University of North Carolina School of Medicine, Chapel Hill
| | | | - Lisa LaVange
- University of North Carolina Gillings School of Global Public Health, Chapel Hill
| | | | - Lisa Berdan
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| | | | - David C Goff
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| | - Amy P Patterson
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, MD
| |
Collapse
|
2
|
Iasella CJ, Hannan SJ, Lyons EJ, Lieber SC, Das A, Dimitrov D, Li W, Saul M, Popescu I, Koshy R, Burke R, Lape B, Brown MJ, Chen X, Sembrat JC, Devonshire K, Kitsios GD, Konstantinidis I, Snyder ME, Chen BB, Merlo CA, Hager DN, Kiss JE, Yazer MH, Wells AH, Morris A, McVerry BJ, McMahon DK, Triulzi DJ, McDyer JF. Impact of variable titer COVID-19 convalescent plasma and recipient SARS-CoV2-specific humoral immunity on survival in hospitalized patients. PLoS One 2024; 19:e0309449. [PMID: 39446792 PMCID: PMC11500870 DOI: 10.1371/journal.pone.0309449] [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: 03/13/2024] [Accepted: 08/12/2024] [Indexed: 10/26/2024] Open
Abstract
COVID-19 convalescent plasma (CCP) was one of the first therapies to receive emergency use authorization for management of COVID-19. We assessed the effectiveness of CCP in a propensity-matched analysis, and whether the presence of antibodies in the recipient at the time of treatment or the titer of antibodies in the administered CCP influenced clinical effectiveness. In an inpatient population within a single large health system, a total of 290 CCP patients were matched to 290 controls. While CCP increased titers of anti-SARS-CoV-2 RBD IgG titers post-CCP (p = <0.0001), no differences in 30-day survival were observed between CCP patients and controls in univariate and multivariate analyses. Survival at 30 days was numerically lower in recipients who were seronegative prior to CCP administration, compared to those with low titer and high titer anti-SARS-CoV-2 RBD IgG, respectively, but did not reach statistical significance (56% vs 82% vs 75%, p = 0.16). Patients who received 2 units of high-titer CCP had numerically better survival versus those who received fewer high-titer units, but this was not statistically significant (p = 0.08). CCP did not improve 30-day survival compared to propensity matched controls. Together these data support that CCP therapy provides limited benefit to hospitalized patients with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carlo J. Iasella
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Stefanie J. Hannan
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Emily J. Lyons
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sophia C. Lieber
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Antu Das
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Dimiter Dimitrov
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Wei Li
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Melissa Saul
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Iulia Popescu
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ritchie Koshy
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robin Burke
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Braidon Lape
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Mark J. Brown
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Xiaoping Chen
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John C. Sembrat
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kaitlyn Devonshire
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Georgios D. Kitsios
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ioannis Konstantinidis
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Mark E. Snyder
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Bill B. Chen
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Christian A. Merlo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - David N. Hager
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Joseph E. Kiss
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Mark H. Yazer
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alan H. Wells
- Department of Pathology, Division of Laboratory Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alison Morris
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Bryan J. McVerry
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Deborah K. McMahon
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Darrell J. Triulzi
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John F. McDyer
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
3
|
Verbrugghe C, Wouters E, Devloo R, Nurmi V, Seghers S, De Bleser D, Harvala H, Compernolle V, Feys HB. Biochemical rationale for transfusion of high titre COVID-19 convalescent plasma. Sci Rep 2024; 14:23579. [PMID: 39384892 PMCID: PMC11464705 DOI: 10.1038/s41598-024-75093-x] [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: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024] Open
Abstract
We aimed to model binding of donor antibodies to virus that infects COVID-19 patients following transfusion of convalescent plasma (CCP). An immunosorbent assay was developed to determine apparent affinity (Kd, app). Antibody binding to virus was modelled using antibody concentration and estimations of viral load. Assay and model were validated using reference antibodies and clinical data of monoclonal antibody therapy. A single Kd, app or two resolvable Kd, app were found for IgG in 11% or 89% of CCP donations, respectively. For IgA this was 50%-50%. Median IgG Kd, app was 0.8nM and 3.6nM for IgA, ranging from 0.1-14.7nM and 0.2-156.0nM respectively. The median concentration of IgG was 44.0nM (range 8.4-269.0nM) and significantly higher than IgA at 2.0nM (range 0.4-11.4nM). The model suggested that a double CCP transfusion (i.e. 500 mL) allows for > 80% binding of antibody to virus provided Kd, app was < 1nM and concentration > 150nM. In our cohort from the pre-vaccination era, 4% of donations fulfilled these criteria. Low and mid-range viral loads are found early post exposure, suggesting that convalescent plasma will be most effective then. This study provides a biochemical rationale for selecting high affinity and high antibody concentration CCP transfused early in the disease course.
Collapse
Affiliation(s)
- Caro Verbrugghe
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Elise Wouters
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Rosalie Devloo
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Visa Nurmi
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Sabrina Seghers
- Transfusion Innovation Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | | | - Heli Harvala
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Microbiology Services, NHS Blood and Transplant, Colindale, UK
- Infection and Immunity, University College of London, London, UK
| | - Veerle Compernolle
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
- Transfusion Innovation Center, Belgian Red Cross-Flanders, Ghent, Belgium
- Blood Services of the Belgian Red Cross-Flanders, Mechelen, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium.
- Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium.
- , Ottergemsesteenweg 413, Ghent, 9000, Belgium.
| |
Collapse
|
4
|
Dragotakes Q, Johnson PW, Buras MR, Carter RE, Joyner MJ, Bloch E, Gebo KA, Hanley DF, Henderson JP, Pirofski LA, Shoham S, Senefeld JW, Tobian AA, Wiggins CC, Wright RS, Paneth NS, Sullivan DJ, Casadevall A. Estimates of actual and potential lives saved in the United States from the use of COVID-19 convalescent plasma. Proc Natl Acad Sci U S A 2024; 121:e2414957121. [PMID: 39352932 PMCID: PMC11474081 DOI: 10.1073/pnas.2414957121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
In the Spring of 2020, the United States of America (USA) deployed COVID-19 convalescent plasma (CCP) to treat hospitalized patients. Over 500,000 patients were treated with CCP during the first year of the pandemic. In this study, we estimated the number of actual inpatient lives saved by CCP treatment in the United States of America based on CCP weekly use, weekly national mortality data, and CCP mortality reduction data from meta-analyses of randomized controlled trials and real-world data. We also estimate the potential number of lives saved if CCP had been deployed for 100% of hospitalized patients or used in 15 to 75% of outpatients. Depending on the assumptions modeled in stratified analyses, we estimated that CCP saved between 16,476 and 66,296 lives. The CCP ideal use might have saved as many as 234,869 lives and prevented 1,136,133 hospitalizations. CCP deployment was a successful strategy for ameliorating the impact of the COVID-19 pandemic in the USA. This experience has important implications for convalescent plasma use in future infectious disease emergencies.
Collapse
Affiliation(s)
- Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Patrick W. Johnson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL32224
| | - Matthew R. Buras
- Division of Biostatistics and Clinical Trials, Department of Quantitative Health Sciences, Scottsdale, AZ85259
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL32224
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN55905
| | - Evan Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Kelly A. Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniel F. Hanley
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jeffrey P. Henderson
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Louis, St. Louis, MO63110
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine, New York, NY10461
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jonathon W. Senefeld
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Chad C. Wiggins
- Department of Kinesiology, Michigan State University, East Lansing, MI48823
| | - R. Scott Wright
- Departments of Cardiovascular Medicine and Human Research Protection Program, Mayo Clinic, Rochester, MN55905
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI48823
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI48823
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| |
Collapse
|
5
|
Franchini M, Casadevall A, Dragotakes Q, Focosi D. Avoided and Avoidable Deaths with the Use of COVID-19 Convalescent Plasma in Italy during the First Two Years of Pandemic. Life (Basel) 2024; 14:1207. [PMID: 39337989 PMCID: PMC11433499 DOI: 10.3390/life14091207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Italy was the first western country to be hit by the COVID-19 pandemic and has suffered nearly 200,000 deaths so far during the four years of the pandemic. In March 2020, Italy first deployed COVID-19 convalescent plasma (CCP) to treat hospitalized patients. Despite this initial effort, the proportion of COVID-19 patients treated with CCP during the first two years of the pandemic (2020-2021) was very low (approximately 2% of individuals hospitalized for COVID-19). In this study, we estimated the number of actual inpatient lives saved by CCP treatment in Italy using national mortality data, and CCP mortality reduction data from meta-analyses of randomized controlled trials and real-world data. We also estimated the potential number of lives saved if CCP had been deployed to 100% of hospitalized patients or used in 15% to 75% of outpatients. According to these models, CCP usage in 2020-2021 saved between 385-1304 lives, but this number would have increased to 17,751-60,079 if 100% of inpatients had been transfused with CCP. Similarly, broader (15-75%) usage in outpatients could have prevented 21,187-190,689 hospitalizations (desaturating hospitals) and 6144-81,926 deaths. These data have important implications for convalescent plasma use in future infectious disease emergencies.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy;
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (A.C.); (Q.D.)
| | - Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (A.C.); (Q.D.)
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| |
Collapse
|
6
|
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
|
7
|
Park H, Yu C, Pirofski LA, Yoon H, Wu D, Li Y, Tarpey T, Petkova E, Antman EM, Troxel AB. Association between COVID-19 convalescent plasma antibody levels and COVID-19 outcomes stratified by clinical status at presentation. BMC Infect Dis 2024; 24:639. [PMID: 38926676 PMCID: PMC11201301 DOI: 10.1186/s12879-024-09529-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND There is a need to understand the relationship between COVID-19 Convalescent Plasma (CCP) anti-SARS-CoV-2 IgG levels and clinical outcomes to optimize CCP use. This study aims to evaluate the relationship between recipient baseline clinical status, clinical outcomes, and CCP antibody levels. METHODS The study analyzed data from the COMPILE study, a meta-analysis of pooled individual patient data from 8 randomized clinical trials (RCTs) assessing the efficacy of CCP vs. control, in adults hospitalized for COVID-19 who were not receiving mechanical ventilation at randomization. SARS-CoV-2 IgG levels, referred to as 'dose' of CCP treatment, were retrospectively measured in donor sera or the administered CCP, semi-quantitatively using the VITROS Anti-SARS-CoV-2 IgG chemiluminescent immunoassay (Ortho-Clinical Diagnostics) with a signal-to-cutoff ratio (S/Co). The association between CCP dose and outcomes was investigated, treating dose as either continuous or categorized (higher vs. lower vs. control), stratified by recipient oxygen supplementation status at presentation. RESULTS A total of 1714 participants were included in the study, 1138 control- and 576 CCP-treated patients for whom donor CCP anti-SARS-CoV2 antibody levels were available from the COMPILE study. For participants not receiving oxygen supplementation at baseline, higher-dose CCP (/control) was associated with a reduced risk of ventilation or death at day 14 (OR = 0.19, 95% CrI: [0.02, 1.70], posterior probability Pr(OR < 1) = 0.93) and day 28 mortality (OR = 0.27 [0.02, 2.53], Pr(OR < 1) = 0.87), compared to lower-dose CCP (/control) (ventilation or death at day 14 OR = 0.79 [0.07, 6.87], Pr(OR < 1) = 0.58; and day 28 mortality OR = 1.11 [0.10, 10.49], Pr(OR < 1) = 0.46), exhibiting a consistently positive CCP dose effect on clinical outcomes. For participants receiving oxygen at baseline, the dose-outcome relationship was less clear, although a potential benefit for day 28 mortality was observed with higher-dose CCP (/control) (OR = 0.66 [0.36, 1.13], Pr(OR < 1) = 0.93) compared to lower-dose CCP (/control) (OR = 1.14 [0.73, 1.78], Pr(OR < 1) = 0.28). CONCLUSION Higher-dose CCP is associated with its effectiveness in patients not initially receiving oxygen supplementation, however, further research is needed to understand the interplay between CCP anti-SARS-CoV-2 IgG levels and clinical outcome in COVID-19 patients initially receiving oxygen supplementation.
Collapse
Affiliation(s)
- Hyung Park
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Chang Yu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Liise-Anne Pirofski
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Hyunah Yoon
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Danni Wu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Yi Li
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Thaddeus Tarpey
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Eva Petkova
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Elliott M Antman
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea B Troxel
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
8
|
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] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [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
|
9
|
Singh S, Boyd S, Schilling WHK, Watson JA, Mukaka M, White NJ. The relationship between viral clearance rates and disease progression in early symptomatic COVID-19: a systematic review and meta-regression analysis. J Antimicrob Chemother 2024; 79:935-945. [PMID: 38385479 PMCID: PMC11062948 DOI: 10.1093/jac/dkae045] [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: 10/09/2023] [Accepted: 01/31/2024] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Effective antiviral drugs accelerate viral clearance in acute COVID-19 infections; the relationship between accelerating viral clearance and reducing severe clinical outcomes is unclear. METHODS A systematic review was conducted of randomized controlled trials (RCTs) of antiviral therapies in early symptomatic COVID-19, where viral clearance data were available. Treatment benefit was defined clinically as the relative risk of hospitalization/death during follow-up (≥14 days), and virologically as the SARS-CoV-2 viral clearance rate ratio (VCRR). The VCRR is the ratio of viral clearance rates between the intervention and control arms. The relationship between the clinical and virological treatment effects was assessed by mixed-effects meta-regression. RESULTS From 57 potentially eligible RCTs, VCRRs were derived for 44 (52 384 participants); 32 had ≥1 clinical endpoint in each arm. Overall, 9.7% (R2) of the variation in clinical benefit was explained by variation in VCRRs with an estimated linear coefficient of -0.92 (95% CI: -1.99 to 0.13; P = 0.08). However, this estimate was highly sensitive to the inclusion of the recent very large PANORAMIC trial. Omitting this outlier, half the variation in clinical benefit (R2 = 50.4%) was explained by variation in VCRRs [slope -1.47 (95% CI -2.43 to -0.51); P = 0.003], i.e. higher VCRRs were associated with an increased clinical benefit. CONCLUSION Methods of determining viral clearance in COVID-19 studies and the relationship to clinical outcomes vary greatly. As prohibitively large sample sizes are now required to show clinical treatment benefit in antiviral therapeutic assessments, viral clearance is a reasonable surrogate endpoint.
Collapse
Affiliation(s)
- Shivani Singh
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
| | - Simon Boyd
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - William H K Schilling
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - James A Watson
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
- Biostatistics Department, Oxford University Clinical Research Unit, 764 Vo Van Kiet, Quan 5, Ho Chi Minh City, Vietnam
| | - Mavuto Mukaka
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| | - Nicholas J White
- Faculty of Tropical Medicine, Mahidol Oxford Tropical Medicine Research Unit, Mahidol University, Bangkok, Thailand
- Nuffield Department of Medicine, Centre for Tropical Medicine and Global Health, Oxford University, Oxford, UK
| |
Collapse
|
10
|
Huygens S, Preijers T, Swaneveld FH, Kleine Budde I, GeurtsvanKessel CH, Koch BCP, Rijnders BJA. Dosing of Convalescent Plasma and Hyperimmune Anti-SARS-CoV-2 Immunoglobulins: A Phase I/II Dose-Finding Study. Clin Pharmacokinet 2024; 63:497-509. [PMID: 38427270 PMCID: PMC11052786 DOI: 10.1007/s40262-024-01351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND AND OBJECTIVE During the COVID-19 pandemic, trials on convalescent plasma (ConvP) were performed without preceding dose-finding studies. This study aimed to assess potential protective dosing regimens by constructing a population pharmacokinetic (popPK) model describing anti-SARS-CoV-2 antibody titers following the administration of ConvP or hyperimmune globulins (COVIg). METHODS Immunocompromised patients, testing negative for anti-SARS-CoV-2 spike antibodies despite vaccination, received a range of anti-SARS-CoV-2 antibodies in the form of COVIg or ConvP infusion. The popPK analysis was performed using NONMEM v7.4. Monte Carlo simulations were performed to assess potential COVIg and ConvP dosing regimens for prevention of COVID-19. RESULTS Forty-four patients were enrolled, and data from 42 were used for constructing the popPK model. A two-compartment elimination model with mixed residual error best described the Nab-titers after administration. Inter-individual variation was associated to CL (44.3%), V1 (27.3%), and V2 (29.2%). Lean body weight and type of treatment (ConvP/COVIg) were associated with V1 and V2, respectively. Median elimination half-life was 20 days (interquartile range: 17-25 days). Simulations demonstrated that even monthly infusions of 600 mL of the ConvP or COVIg used in this trial would not achieve potentially protective serum antibody titers for > 90% of the time. However, as a result of hybrid immunity and/or repeated vaccination, plasma donors with extremely high antibody titers are now readily available, and a > 90% target attainment should be possible. CONCLUSION The results of this study may inform future intervention studies on the prophylactic and therapeutic use of antiviral antibodies in the form of ConvP or COVIg. CLINICAL TRIAL REGISTRATION NUMBER NL9379 (The Netherlands Trial Register).
Collapse
Affiliation(s)
- Sammy Huygens
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Tim Preijers
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Francis H Swaneveld
- Unit of Transfusion Medicine, Sanquin Blood Supply Foundation, 1066 CX, Amsterdam, The Netherlands
| | - Ilona Kleine Budde
- Clinical Operations, Prothya Biosolutions, 1066 CX, Amsterdam, The Netherlands
| | - Corine H GeurtsvanKessel
- Department of Viroscience, Erasmus University Medical Center Rotterdam, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
11
|
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
| | | | - Amy L Gawad
- Department of Neurology, Brain Injury Outcomes
| | | | - Atika Singh
- Department of Neurology, Brain Injury Outcomes
| | - Daniel E Ford
- Institute for Clinical and Translational Research, and
| | - 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 Ar 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
|
12
|
Casadevall A, Sullivan DJ. Late Administration and Corticosteroid Usage Explain Inefficacy in COVID-19 Convalescent Plasma Trial. J Infect Dis 2024; 229:617-618. [PMID: 37967364 DOI: 10.1093/infdis/jiad509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 11/17/2023] Open
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Room 5132, Baltimore, MD 21205, USA
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, 615 North Wolfe Street, Room 5132, Baltimore, MD 21205, USA
| |
Collapse
|
13
|
Franchini M, Focosi D. Hyperimmune Plasma and Immunoglobulins against COVID-19: A Narrative Review. Life (Basel) 2024; 14:214. [PMID: 38398723 PMCID: PMC10890293 DOI: 10.3390/life14020214] [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: 11/20/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Since late 2019, the new SARS-CoV-2 virus belonging to the Coronaviridae family has been responsible for COVID-19 pandemic, a severe acute respiratory syndrome. Several antiviral therapies, mostly derived from previous epidemics, were initially repurposed to fight this not rarely life-threatening respiratory illness. Among them, however, the only specific antibody-based therapy available against SARS-CoV-2 infection during the first year of the pandemic was represented by COVID-19 convalescent plasma (CCP). CCP, collected from recovered individuals, contains high levels of polyclonal antibodies of different subclasses able to neutralize SARS-CoV-2 infection. Tens of randomized controlled trials have been conducted during the last three years of the pandemic to evaluate the safety and the clinical efficacy of CCP in both hospitalized and ambulatory COVID-19 patients, whose main results will be summarized in this narrative review. In addition, we will present the current knowledge on the development of anti-SARS-CoV-2 hyperimmune polyclonal immunoglobulins.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy;
| |
Collapse
|
14
|
Abstract
The treatment for COVID-19 has evolved rapidly since the start of the pandemic and now consists mainly of antiviral and immunomodulatory agents. Antivirals, such as remdesivir and nirmatrelvir-ritonavir, have proved to be most useful earlier in illness (e.g., as outpatient therapy) and for less severe disease. Immunomodulatory therapies, such as dexamethasone and interleukin-6 or Janus kinase inhibitors, are most useful in severe disease or critical illness. The role of anti-SARS-CoV-2 monoclonal antibodies has diminished because of the emergence of viral variants that are not anticipated to be susceptible to these treatments, and there still is not a consensus on the use of convalescent plasma. COVID-19 has been associated with increased rates of venous thromboembolism, but the role of antithrombotic therapy is limited. Multiple investigational agents continue to be studied, which will alter current treatment paradigms as new data are released.
Collapse
Affiliation(s)
- Hayden S Andrews
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; ,
| | - Jonathan D Herman
- Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA;
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Rajesh T Gandhi
- Division of Infectious Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA; ,
| |
Collapse
|
15
|
Senefeld JW, Marks P, Casadevall A, Joyner MJ. The value of observational registry studies for the next infectious disease emergency. mBio 2023; 14:e0256523. [PMID: 37937981 PMCID: PMC10746279 DOI: 10.1128/mbio.02565-23] [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: 11/09/2023] Open
Abstract
During infectious disease emergencies, it may be necessary to deploy new therapies without conclusive evidence for their effectiveness. During the SARS-CoV-2 pandemic, several countries used registries to track the use of COVID-19 convalescent plasma (CCP). Those registries provided evidence that CCP was effective when used early and with high titer.
Collapse
Affiliation(s)
- Jonathon W. Senefeld
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Peter Marks
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
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: 4.0] [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
IMPORTANCE 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
|
19
|
Kasten MJ, Lahr BD, Parisapogu A, Yetmar ZA, O'Horo JC, Orenstein R, Moreno Franco P, Razonable RR, Vergidis P, Shah AS, Enzler MJ, Inwards DJ, Bauer PR. COVID-19 outcome is not affected by anti-CD20 or high-titer convalescent plasma in immunosuppressed patients. Sci Rep 2023; 13:21249. [PMID: 38040756 PMCID: PMC10692159 DOI: 10.1038/s41598-023-48145-x] [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: 09/22/2023] [Accepted: 11/22/2023] [Indexed: 12/03/2023] Open
Abstract
The role of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) convalescent plasma in the treatment of Coronavirus Disease 2019 (COVID-19) in immunosuppressed individuals remains controversial. We describe the course of COVID-19 in patients who had received anti-CD20 therapy within the 3 years prior to infection. We compared outcomes between those treated with and those not treated with high titer SARS-CoV2 convalescent plasma. We identified 144 adults treated at Mayo clinic sites who had received anti-CD20 therapies within a median of 5.9 months prior to the COVID-19 index date. About one-third (34.7%) were hospitalized within 14 days and nearly half (47.9%) within 90 days. COVID-19 directed therapy included anti-spike monoclonal antibodies (n = 30, 20.8%), and, among those hospitalized within 14 days (n = 50), remdesivir (n = 45, 90.0%), glucocorticoids (n = 36, 72.0%) and convalescent plasma (n = 24, 48.0%). The duration from receipt of last dose of anti-CD20 therapy did not correlate with outcomes. The overall 90-day mortality rate was 14.7%. Administration of convalescent plasma within 14 days of the COVID-19 diagnosis was not significantly associated with any study outcome. Further study of COVID-19 in CD20-depleted individuals is needed focusing on the early administration of new and potentially combination antiviral agents, associated or not with vaccine-boosted convalescent plasma.
Collapse
Affiliation(s)
- Mary J Kasten
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Brian D Lahr
- Division of Clinical Trials and Biostatistics, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | - Zachary A Yetmar
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Infectious Disease, Integrated Hospital-Care Institute, Cleveland Clinic, Cleveland, OH, USA
| | - John C O'Horo
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | | | | | - Raymund R Razonable
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Paschalis Vergidis
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Aditya S Shah
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - Mark J Enzler
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA
| | - David J Inwards
- Division of Hematology, Emeritus Staff Center, Mayo Clinic, Rochester, MN, USA
| | - Philippe R Bauer
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
20
|
|
21
|
Şimşek-Yavuz S. COVID-19: An Update on Epidemiology, Prevention and Treatment, September-2023. INFECTIOUS DISEASES & CLINICAL MICROBIOLOGY 2023; 5:165-187. [PMID: 38633552 PMCID: PMC10986731 DOI: 10.36519/idcm.2023.251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/19/2023] [Indexed: 04/19/2024]
Abstract
After a downward trend for more than 12 months, the incidence of COVID-19 has increased in the last months. Although COVID-19 is not as frequent as in the first years of the pandemic, case numbers are still very high, and it causes a significant number of deaths. COVID-19 is not seen with a predictable frequency, at least two times more deadly than the flu, continues as an epidemic, and has not reached the endemic level yet. Currently, the Omicron strains EG.5 and XBB.1.16 are dominant worldwide. Although BA.2.86 and FLip variants, including FL.1.5.1 are not widespread at the moment, both were shown to be highly immune-evasive and require close monitoring. Prevention of COVID-19 relies on vaccinations, surveillance, proper ventilation of enclosed spaces, isolation of patients, and mask usage. Currently, monovalent COVID-19 vaccines, including XBB.1.5 Omicron SARS-CoV-2, are recommended for both primary and booster vaccinations against COVID-19. Monovalent vaccines, including only original SARS-CoV-2 strain, and bivalent vaccines, including original virus plus BA4/5 variant, are no longer recommended against COVID-19. Booster vaccination with XBB.1.5 containing vaccine should be prioritized for patients at high risk for severe COVID-19. Bacillus Calmette-Guérin (BCG) vaccination does not seem to be effective in preventing COVID-19. At the current phase of the pandemic, nirmatrelvir/ritonavir, remdesivir, molnupiravir, sotrovimab (for patients from XBB.1.5 variant dominant settings), and convalescent plasma can be considered for the treatment of high-risk early-stage outpatients with COVID-19, while hospitalized patients with more severe disease can be treated with dexamethasone, anti cytokines including tocilizumab, sarilumab, baricitinib, and tofacitinib and antithrombotic agents including enoxaparin. Remdesivir oral analogues and ensitrelvir fumarate are promising agents for treating acute COVID-19, which are in phase trials now; however, ivermectin, fluvoxamine, and metformin were shown to be ineffective.
Collapse
Affiliation(s)
- Serap Şimşek-Yavuz
- Department of Infectious Diseases and Clinical Microbiology, İstanbul University School of Medicine, İstanbul, Türkiye
| |
Collapse
|
22
|
Huaman MA, Raval JS, Paxton JH, Mosnaim GS, Patel B, Anjan S, Meisenberg BR, Levine AC, Marshall CE, Yarava A, Shenoy AG, Heath SL, Currier JS, Fukuta Y, Blair JE, Spivak ES, Petrini JR, Broderick PB, Rausch W, Cordisco M, Hammel J, Greenblatt B, Cluzet VC, Cruser D, Oei K, Abinante M, Hammitt LL, Sutcliffe CG, Forthal DN, Zand MS, Cachay ER, Kassaye SG, Ram M, Wang Y, Das P, Lane K, McBee NA, Gawad AL, Karlen N, Ford DE, Laeyendecker O, Pekosz A, Klein SL, Ehrhardt S, Lau B, Baksh SN, Shade DM, Casadevall A, Hanley DF, Ou J, Gniadek TJ, Ziman A, Shoham S, Gebo KA, Bloch EM, Tobian AAR, Sullivan DJ, Gerber JM. Transfusion reactions associated with COVID-19 convalescent plasma in outpatient clinical trials. Transfusion 2023; 63:1639-1648. [PMID: 37534607 PMCID: PMC10720768 DOI: 10.1111/trf.17485] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/20/2023] [Accepted: 06/20/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) is an important therapeutic option for outpatients at high risk of hospitalization from SARS-CoV-2 infection. We assessed the safety of outpatient CCP transfusions administered during clinical trials. STUDY DESIGN AND METHODS We analyzed data pertaining to transfusion-related reactions from two randomized controlled trials in the U.S. that evaluated the efficacy of CCP versus control plasma in various ambulatory settings. Multivariable logistic regression was used to assess whether CCP was associated with transfusion reactions, after adjusting for potential confounders. RESULTS The combined study reported 79/1351 (5.9%) adverse events during the transfusion visit, with the majority 62/1351 (4.6%) characterized by mild, allergic-type findings of urticaria, and/or pruritus consistent with minor allergic transfusion reactions; the other reported events were attributed to the patients' underlying disease, COVID-19, or vasovagal in nature. We found no difference in the likelihood of allergic transfusion reactions between those receiving CCP versus control plasma (adjusted odds ratio [AOR], 0.75; 95% CI, 0.43-1.31). Risk of urticaria and/or pruritus increased with a pre-existing diagnosis of asthma (AOR, 2.33; 95% CI, 1.16-4.67). We did not observe any CCP-attributed antibody disease enhancement in participants with COVID-19 or increased risk of infection. There were no life-threatening severe transfusion reactions and no patients required hospitalization related to transfusion-associated complications. DISCUSSION Outpatient plasma administration was safely performed for nearly 1400 participants. CCP is a safe therapeutic option for outpatients at risk of hospitalization from COVID-19.
Collapse
Affiliation(s)
- Moises A Huaman
- Department of Internal Medicine, Division of Infectious Diseases, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jay S Raval
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, New Mexico, USA
| | - James H Paxton
- Department of Emergency Medicine, Wayne State University, Detroit, Michigan, USA
| | - Giselle S Mosnaim
- Department of Medicine, Division of Allergy and Immunology, NorthShore University Health System, Evanston, Illinois, USA
| | - Bela Patel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Texas Health Science Center, Houston, Texas, USA
| | - Shweta Anjan
- Department of Medicine, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, Florida, USA
| | | | - Adam C Levine
- Department of Emergency Medicine, Rhode Island Hospital & Brown University, Providence, Rhode Island, USA
| | - Christi E Marshall
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Anusha Yarava
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Aarthi G Shenoy
- Department of Medicine, Division of Hematology and Oncology, MedStar Washington Hospital Center, DC, USA
| | - Sonya L Heath
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Judith S Currier
- Department of Medicine, Division of Infectious Diseases, University of California, Los Angeles, USA
| | - Yuriko Fukuta
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, Texas, USA
| | - Janis E Blair
- Department of Medicine, Division of Infectious Diseases, Mayo Clinic Hospital, Phoenix, Arizona, USA
| | - Emily S Spivak
- Department of Medicine, Division of Infectious Diseases, University of Utah School of Medicine, Salt Lake City, Utah, 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, Johns Hopkins University, Baltimore, Maryland, USA
| | - Catherine G Sutcliffe
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, 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, University of California, San Diego, California, USA
| | - Seble G Kassaye
- Department of Medicine, Division of Infectious Diseases, Georgetown University Medical Center, DC, USA
| | - Malathi Ram
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ying Wang
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Piyali Das
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Karen Lane
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nichol A McBee
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Amy L Gawad
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nicky Karlen
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel E Ford
- Institute for Clinical and Translational Research, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Oliver Laeyendecker
- The Division of Intramural Research, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, USA
| | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sabra L Klein
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Stephan Ehrhardt
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Bryan Lau
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sheriza N Baksh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - David M Shade
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Daniel F Hanley
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jiangda Ou
- Department of Neurology, Brain Injury Outcomes Division, 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
| | - 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, California, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, 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
| | - Evan M Bloch
- Department of Pathology, 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
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jonathan M Gerber
- Department of Medicine, Division of Hematology and Oncology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
23
|
Richier Q, Hueso T, Tiberghien P, Lacombe K. [COVID-19: Still a place for the convalescent plasma? Focus on the immunocompromised patients]. Rev Med Interne 2023; 44:467-471. [PMID: 37689526 DOI: 10.1016/j.revmed.2023.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/29/2023] [Indexed: 09/11/2023]
Affiliation(s)
- Q Richier
- Sorbonne université, Paris, France; Service de maladies infectieuses, hôpital Saint-Antoine, AP-HP, 75012 Paris, France.
| | - T Hueso
- Service d'hématologie clinique, hôpital Avicenne, Sorbonne université Paris-Nord, AP-HP, Bobigny, France
| | - P Tiberghien
- Établissement français du sang, La Plaine Saint-Denis, France; Établissement français du sang, UMR 1098 Right Inserm, université de Franche-Comté, Besançon, France
| | - K Lacombe
- Sorbonne université, Paris, France; Service de maladies infectieuses, hôpital Saint-Antoine, AP-HP, 75012 Paris, France; Institut Pierre-Louis épidémiologie et santé publique, Inserm UMR-S1136, Paris, France
| |
Collapse
|
24
|
Casadevall A, Pirofski LA. Misinterpretation of Clinical Research Findings and COVID-19 Mortality. Ann Intern Med 2023; 176:993-994. [PMID: 37186922 PMCID: PMC10186249 DOI: 10.7326/m23-0737] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
The first 3 years of the COVID-19 pandemic witnessed an unprecedented pace of research that dramatically lessened morbidity and mortality due to COVID-19. This commentary discusses research findings that led to clinical practice recommendations that were later associated with excess mortality.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland (A.C.)
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York (L.P.)
| |
Collapse
|
25
|
Wouters E, Verbrugghe C, Abdelnabi R, Devloo R, De Clippel D, Jochmans D, De Bleser D, Weynand B, Compernolle V, Neyts J, Feys HB. Intranasal administration of convalescent plasma protects against SARS-CoV-2 infection in hamsters. EBioMedicine 2023; 92:104597. [PMID: 37148586 PMCID: PMC10171892 DOI: 10.1016/j.ebiom.2023.104597] [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: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Convalescent plasma (CP) transfusion is an early option for treating infections with pandemic potential, often preceding vaccine or antiviral drug rollout. Heterogenous findings from randomized clinical trials on transfusion of COVID-19 CP (CCP) have been reported. However, meta-analysis suggests that transfusion of high titer CCP is associated with a mortality benefit for COVID-19 outpatients or inpatients treated within 5 days after symptom onset, indicating the importance of early administration. METHODS We tested if CCP is an effective prophylactic against SARS-CoV-2 infection by the intranasal administration of 25 μL CCP/nostril (i.e. 0.01-0.06 mg anti-RBD antibodies/kg) in hamsters exposed to infected littermates. FINDINGS In this model, 40% of CCP treated hamsters were fully protected and 40% had significantly reduced viral loads, the remaining 20% was not protected. The effect seems dose-dependent because high-titer CCP from a vaccinated donor was more effective than low-titer CCP from a donation prior to vaccine rollout. Intranasal administration of human CCP resulted in a reactive (immune) response in hamster lungs, however this was not observed upon administration of hamster CCP. INTERPRETATION We conclude that CCP is an effective prophylactic when used directly at the site of primary infection. This option should be considered in future prepandemic preparedness plans. FUNDING Flanders Innovation & Entrepreneurship (VLAIO) and the Foundation for Scientific Research of the Belgian Red Cross Flanders.
Collapse
Affiliation(s)
- Elise Wouters
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Caro Verbrugghe
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Rosalie Devloo
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | | | - Dirk Jochmans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | | | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Division of Translational Cell and Tissue Research, B-3000, Leuven, Belgium
| | - Veerle Compernolle
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Blood Services of the Belgian Red Cross-Flanders, Mechelen, Belgium; Transfusion Innovation Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Blood Services of the Belgian Red Cross-Flanders, Mechelen, Belgium.
| |
Collapse
|
26
|
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: 17] [Impact Index Per Article: 17.0] [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
|
27
|
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: 14] [Impact Index Per Article: 14.0] [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
|
28
|
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: 3.0] [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
|
29
|
Sullivan DJ, Franchini M, Senefeld JW, Joyner MJ, Casadevall A, Focosi D. Outpatient Treatment of Confirmed COVID-19. Ann Intern Med 2023; 176:eL230099. [PMID: 37068278 DOI: 10.7326/l23-0099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/19/2023] Open
|