1
|
Kunze KL, Johnson PW. The Importance of Geographic Proximity of Convalescent Plasma Donors. Curr Top Microbiol Immunol 2024. [PMID: 39117845 DOI: 10.1007/82_2024_270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Donor-recipient proximity emerged as an important factor influencing the efficacy of COVID-19 convalescent plasma (CCP) treatment during the early stages of the COVID-19 pandemic. This relationship was uncovered while analyzing data collected in the collaborative Expanded Access Program (EAP) for CCP at Mayo Clinic, a project aimed to establish protocols for CCP use amid the uncertainty of the novel disease. Analysis of data from nearly 28,000 patients revealed a significant reduction in risk of 30-day mortality for those receiving near-sourced plasma when compared to those receiving distantly sourced plasma [pooled relative risk, 0.73 (95% CI 0.67-0.80)], prompting adjustments in treatment protocols at selected institutions, and highlighting the importance of proximity in optimizing CCP outcomes. Despite its significance, subsequent studies of CCP effectiveness in COVID-19 have often overlooked donor-recipient proximity. Our findings emphasize the importance of donor-recipient proximity in CCP treatment in the current pandemic, and we discuss potential methods for improving CCP efficacy in future pandemics. Our recommendations include prioritizing virus genotyping for vulnerable patients, establishing a robust testing infrastructure, and collecting additional donor data to enhance plasma selection. This chapter underscores the importance of comprehensive data collection and sharing to navigate the evolving landscape of newly emerging infectious diseases.
Collapse
Affiliation(s)
- Katie L Kunze
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ, USA.
- Digital Innovation Lab, Mayo Clinic, Jacksonville, FL, USA.
| | - Patrick W Johnson
- Digital Innovation Lab, Mayo Clinic, Jacksonville, FL, USA
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
2
|
Yoon H, Pirofski LA. Generating the Evidence Base for Convalescent Plasma Use for a New Infectious Disease. Curr Top Microbiol Immunol 2024. [PMID: 39117847 DOI: 10.1007/82_2024_275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) swept across the world in the waning months of 2019 and emerged as the cause of the coronavirus disease 19 (COVID-19) pandemic in early 2020. The use of convalescent plasma (CP) for prior respiratory pandemics provided a strong biological rationale for the rapid deployment of COVID-19 convalescent plasma (CCP) in early 2020 when no validated treatments or prior immunity existed. CCP is an antiviral agent, with its activity against SARS-CoV-2 stemming from specific antibodies elicited by the virus. Early efforts to investigate the efficacy of CCP in randomized clinical trials (RCTs) that targeted hospitalized patients with COVID-19 did not demonstrate the overall efficacy of CCP despite signals of benefit in certain subgroups, such as those treated earlier in disease. In contrast, studies adhering to the principles of antibody therapy in their study design, choice of patient population, and product qualification, i.e., those that administered high levels of specific antibody during the viral phase of disease in immunocompromised or very early in immunocompetent individuals, demonstrated benefits. In this chapter, we leverage the knowledge gained from clinical studies of CCP for COVID-19 to propose a framework for future studies of CP for a new infectious disease. This framework includes obtaining high-quality CP and designing clinical studies that adhere to the principles of antibody therapy to generate a robust evidence base for using CP.
Collapse
Affiliation(s)
- Hyunah Yoon
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
3
|
Cognasse F, Hamzeh-Cognasse H, Rosa M, Corseaux D, Bonneaudeau B, Pierre C, Huet J, Arthaud CA, Eyraud MA, Prier A, Duchez AC, Ebermeyer T, Heestermans M, Audoux-Caire E, Philippot Q, Le Voyer T, Hequet O, Fillet AM, Chavarin P, Legrand D, Richard P, Pirenne F, Gallian P, Casanova JL, Susen S, Morel P, Lacombe K, Bastard P, Tiberghien P. Inflammatory markers and auto-Abs to type I IFNs in COVID-19 convalescent plasma cohort study. EBioMedicine 2022; 87:104414. [PMID: 36535107 PMCID: PMC9758484 DOI: 10.1016/j.ebiom.2022.104414] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/10/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) contains neutralising anti-SARS-CoV-2 antibodies that may be useful as COVID-19 passive immunotherapy in patients at risk of developing severe disease. Such plasma from convalescent patients may also have additional immune-modulatory properties when transfused to COVID-19 patients. METHODS CCP (n = 766) was compared to non-convalescent control plasma (n = 166) for soluble inflammatory markers, ex-vivo inflammatory bioactivity on endothelial cells, neutralising auto-Abs to type I IFNs and reported adverse events in the recipients. FINDINGS CCP exhibited a statistically significant increase in IL-6 and TNF-alpha levels (0.531 ± 0.04 vs 0.271 ± 0.04; (95% confidence interval [CI], 0.07371-0.4446; p = 0.0061) and 0.900 ± 0.07 vs 0.283 ± 0.07 pg/mL; (95% [CI], 0.3097-0.9202; p = 0.0000829) and lower IL-10 (0.731 ± 0.07 vs 1.22 ± 0.19 pg/mL; (95% [CI], -0.8180 to -0.1633; p = 0.0034) levels than control plasma. Neutralising auto-Abs against type I IFNs were detected in 14/766 (1.8%) CCPs and were not associated with reported adverse events when transfused. Inflammatory markers and bioactivity in CCP with or without auto-Abs, or in CCP whether or not linked to adverse events in transfused patients, did not differ to a statistically significant extent. INTERPRETATION Overall, CCP exhibited moderately increased inflammatory markers compared to the control plasma with no discernible differences in ex-vivo bioactivity. Auto-Abs to type I IFNs detected in a small fraction of CCP were not associated with reported adverse events or differences in inflammatory markers. Additional studies, including careful clinical evaluation of patients treated with CCP, are required in order to further define the clinical relevance of these findings. FUNDING French National Blood Service-EFS, the Association "Les Amis de Rémi" Savigneux, France, the "Fondation pour la Recherche Médicale (Medical Research Foundation)-REACTing 2020".
Collapse
Affiliation(s)
- Fabrice Cognasse
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France,Corresponding author. Etablissement Français du Sang Auvergne-Rhône-Alpes, INSERM U1059, Campus Santé Innovation - 10 rue de la Marandière, 42270, Saint-Priest-en-Jarez, France.
| | - Hind Hamzeh-Cognasse
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Mickael Rosa
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1011- EGID, F-59000 Lille, France,Centre National de la Recherche Scientifique (National Scientific Research Centre), Surgical Critical Care, Department of Anaesthesiology and Critical Care, U1019 - Unité Mixte de Recherche 9017 (Mixed Research Unit 9017) – Lille Centre for Infection and Immunity, France
| | - Delphine Corseaux
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1011- EGID, F-59000 Lille, France,Centre National de la Recherche Scientifique (National Scientific Research Centre), Surgical Critical Care, Department of Anaesthesiology and Critical Care, U1019 - Unité Mixte de Recherche 9017 (Mixed Research Unit 9017) – Lille Centre for Infection and Immunity, France
| | | | - Chloe Pierre
- Etablissement Français du Sang, La Plaine, St Denis, France
| | - Julie Huet
- Etablissement Français du Sang, La Plaine, St Denis, France
| | - Charles Antoine Arthaud
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Marie Ange Eyraud
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Amélie Prier
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Anne Claire Duchez
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Theo Ebermeyer
- Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Marco Heestermans
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Estelle Audoux-Caire
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France,Univ Jean Monnet, Mines Saint-Étienne, INSERM, U 1059 Sainbiose, 42023, Saint-Étienne, France
| | - Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163 (National Institute for Health and Medical Research), Necker Hospital for Sick Children, Paris, France,University of Paris, Imagine Institute, Paris, France
| | - Tom Le Voyer
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163 (National Institute for Health and Medical Research), Necker Hospital for Sick Children, Paris, France,University of Paris, Imagine Institute, Paris, France
| | - Olivier Hequet
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| | | | - Patricia Chavarin
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| | - Dominique Legrand
- Établissement Français du Sang Auvergne-Rhône-Alpes, Saint-Étienne, France
| | | | - France Pirenne
- Univ Paris Est Creteil, INSERM U955, Institut Mondor de Recherche Biomédicale (Mondor Biomedical Research Institute) (IMRB), Creteil, France & Laboratory of Excellence GR-Ex, Paris, France
| | - Pierre Gallian
- Etablissement Français du Sang, La Plaine, St Denis, France,UMR “Unité des Virus Emergents” (Emerging Virus Unit), Aix-Marseille University - IRD 190 - INSERM 1207 - IRBA - EFS - IHU Méditerranée Infection, Marseille, France
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163 (National Institute for Health and Medical Research), Necker Hospital for Sick Children, Paris, France,University of Paris, Imagine Institute, Paris, France,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA,Howard Hughes Medical Institute, New York, NY, USA
| | - Sophie Susen
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1011- EGID, F-59000 Lille, France,Centre National de la Recherche Scientifique (National Scientific Research Centre), Surgical Critical Care, Department of Anaesthesiology and Critical Care, U1019 - Unité Mixte de Recherche 9017 (Mixed Research Unit 9017) – Lille Centre for Infection and Immunity, France
| | - Pascal Morel
- Etablissement Français du Sang, La Plaine, St Denis, France
| | - Karine Lacombe
- Sorbonne University, Inserm IPLESP, Infectious Diseases Department, Saint-Antoine Hospital, APHP (University Hospital Trust), Paris, France
| | - Paul Bastard
- Etablissement Français du Sang, La Plaine, St Denis, France,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163 (National Institute for Health and Medical Research), Necker Hospital for Sick Children, Paris, France,St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Pierre Tiberghien
- Etablissement Français du Sang, La Plaine, St Denis, France,UMR RIGHT U1098, INSERM, Etablissement Français du Sang, University of Franche-Comté, Besançon, France
| |
Collapse
|
4
|
Herman JD, Wang C, Burke JS, Zur Y, Compere H, Kang J, Macvicar R, Taylor S, Shin S, Frank I, Siegel D, Tebas P, Choi GH, Shaw PA, Yoon H, Pirofski LA, Julg BD, Bar KJ, Lauffenburger D, Alter G. Nucleocapsid-specific antibody function is associated with therapeutic benefits from COVID-19 convalescent plasma therapy. Cell Rep Med 2022; 3:100811. [PMID: 36351430 PMCID: PMC9595358 DOI: 10.1016/j.xcrm.2022.100811] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 06/22/2022] [Accepted: 10/16/2022] [Indexed: 11/05/2022]
Abstract
Coronavirus disease 2019 (COVID-19) convalescent plasma (CCP), a passive polyclonal antibody therapeutic agent, has had mixed clinical results. Although antibody neutralization is the predominant approach to benchmarking CCP efficacy, CCP may also influence the evolution of the endogenous antibody response. Using systems serology to comprehensively profile severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) functional antibodies of hospitalized people with COVID-19 enrolled in a randomized controlled trial of CCP (ClinicalTrials.gov: NCT04397757), we find that the clinical benefits of CCP are associated with a shift toward reduced inflammatory Spike (S) responses and enhanced nucleocapsid (N) humoral responses. We find that CCP has the greatest clinical benefit in participants with low pre-existing anti-SARS-CoV-2 antibody function and that CCP-induced immunomodulatory Fc glycan profiles and N immunodominant profiles persist for at least 2 months. We highlight a potential mechanism of action of CCP associated with durable immunomodulation, outline optimal patient characteristics for CCP treatment, and provide guidance for development of a different class of COVID-19 hyperinflammation-targeting antibody therapeutic agents.
Collapse
Affiliation(s)
- Jonathan D Herman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA; Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Yonatan Zur
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Jaewon Kang
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Ryan Macvicar
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Sabian Taylor
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Sally Shin
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Ian Frank
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Don Siegel
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pablo Tebas
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Grace H Choi
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania, Philadelphia, PA, USA
| | - Pamela A Shaw
- Biostatistics Unit, Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - Hyunah Yoon
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA; Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Boris D Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Katharine J Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
5
|
Weisser M, Khanna N, Hedstueck A, Tschudin Sutter S, Roesch S, Stehle G, Sava M, Deigendesch N, Battegay M, Infanti L, Holbro A, Bassetti S, Pargger H, Hirsch HH, Leuzinger K, Kaiser L, Vu D, Baur K, Massaro N, Busch MP, Simmons G, Stone M, Felgner PL, de Assis RR, Khan S, Tsai C, Robinson PV, Seftel D, Irsch J, Bagri A, Buser AS, Corash L. Characterization of Pathogen Inactivated
COVID
‐19 Convalescent Plasma and Responses in Transfused Patients. Transfusion 2022; 62:1997-2011. [PMID: 36054476 PMCID: PMC9538076 DOI: 10.1111/trf.17083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/31/2022] [Accepted: 07/31/2022] [Indexed: 12/15/2022]
Abstract
Background Efficacy of donated COVID‐19 convalescent plasma (dCCP) is uncertain and may depend on antibody titers, neutralizing capacity, timing of administration, and patient characteristics. Study Design and Methods In a single‐center hypothesis‐generating prospective case–control study with 1:2 matched dCCP recipients to controls according to disease severity at day 1, hospitalized adults with COVID‐19 pneumonia received 2 × 200 ml pathogen‐reduced treated dCCP from 2 different donors. We evaluated severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) antibodies in COVID‐19 convalescent plasma donors and recipients using multiple antibody assays including a Coronavirus antigen microarray (COVAM), and binding and neutralizing antibody assays. Outcomes were dCCP characteristics, antibody responses, 28‐day mortality, and dCCP ‐related adverse events in recipients. Results Eleven of 13 dCCPs (85%) contained neutralizing antibodies (nAb). PRT did not affect dCCP antibody activity. Fifteen CCP recipients and 30 controls (median age 64 and 65 years, respectively) were enrolled. dCCP recipients received 2 dCCPs from 2 different donors after a median of one hospital day and 11 days after symptom onset. One dCCP recipient (6.7%) and 6 controls (20%) died (p = 0.233). We observed no dCCP‐related adverse events. Transfusion of unselected dCCP led to heterogeneous SARS CoV‐2 antibody responses. COVAM clustered dCCPs in 4 distinct groups and showed endogenous immune responses to SARS‐CoV‐2 antigens over 14–21 days post dCCP in all except 4 immunosuppressed recipients. Discussion PRT did not impact dCCP anti‐virus neutralizing activity. Transfusion of unselected dCCP did not impact survival and had no adverse effects. Variable dCCP antibodies and post‐transfusion antibody responses indicate the need for controlled trials using well‐characterized dCCP with informative assays.
Collapse
Affiliation(s)
- Maja Weisser
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
- Department of Clinical Research University Hospital Basel Basel Switzerland
| | - Nina Khanna
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
- Department of Clinical Research University Hospital Basel Basel Switzerland
| | - Anemone Hedstueck
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
| | - Sarah Tschudin Sutter
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
- Department of Clinical Research University Hospital Basel Basel Switzerland
| | - Sandra Roesch
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
| | - Gregor Stehle
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | - Mihaela Sava
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
| | | | - Manuel Battegay
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
- Department of Clinical Research University Hospital Basel Basel Switzerland
| | - Laura Infanti
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | - Andreas Holbro
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | - Stefano Bassetti
- Department of Clinical Research University Hospital Basel Basel Switzerland
- Department of Internal Medicine University Hospital Basel Basel Switzerland
| | - Hans Pargger
- Department of Clinical Research University Hospital Basel Basel Switzerland
- Department of Intensive Care University Hospital Basel Basel Switzerland
| | - Hans H. Hirsch
- Division of Infectious Diseases & Hospital Epidemiology University and University Hospital of Basel Basel Switzerland
- Department of Clinical Research University Hospital Basel Basel Switzerland
- Transplantation & Clinical Virology, Department of Biomedicine University of Basel Basel Switzerland
| | - Karoline Leuzinger
- Transplantation & Clinical Virology, Department of Biomedicine University of Basel Basel Switzerland
| | - Laurent Kaiser
- Geneva Centre for Emerging Viral Diseases, Geneva University Hospitals, 1205 Geneva, Switzerland; Laboratory of Virology, Division of Laboratory Medicine, Geneva University Hospitals & Faculty of Medicine University of Geneva Geneva Switzerland
| | - Diem‐Lan Vu
- Division of Infectious Diseases Geneva University Hospitals Geneva Switzerland
| | - Katharina Baur
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | - Nadine Massaro
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | - Michael Paul Busch
- Department of Laboratory Medicine University of California, San Francisco San Francisco CA USA
- Vitalant Research Institute San Francisco CA
| | - Graham Simmons
- Department of Laboratory Medicine University of California, San Francisco San Francisco CA USA
- Vitalant Research Institute San Francisco CA
| | - Mars Stone
- Department of Laboratory Medicine University of California, San Francisco San Francisco CA USA
- Vitalant Research Institute San Francisco CA
| | - Philip L. Felgner
- Department of Physiology and Biophysics, Vaccine Research and Development Laboratory University of California, Irvine Irvine CA USA
| | - Rafael R. de Assis
- Department of Physiology and Biophysics, Vaccine Research and Development Laboratory University of California, Irvine Irvine CA USA
| | - Saahir Khan
- Division of Infectious Diseases, Department of Medicine, Keck School of Medicine University of Southern California Los Angeles CA USA
| | | | | | | | | | | | - Andreas S. Buser
- Department of Clinical Research University Hospital Basel Basel Switzerland
- Regional Blood Transfusion Service, Swiss Red Cross, Basel Basel Switzerland
| | | |
Collapse
|
6
|
Fodor E, Müller V, Iványi Z, Berki T, Kuten Pella O, Hornyák I, Ambrus M, Sárkány Á, Skázel Á, Madár Á, Kardos D, Kemenesi G, Földes F, Nagy S, Matusovits A, János N, Tordai A, Jakab F, Lacza Z. Early Transfusion of Convalescent Plasma Improves the Clinical Outcome in Severe SARS-CoV2 Infection. Infect Dis Ther 2022; 11:293-304. [PMID: 34817840 PMCID: PMC8611245 DOI: 10.1007/s40121-021-00514-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/26/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Plasma harvested from convalescent COVID-19 patients (CCP) has been applied as first-line therapy in the early phase of the SARS-CoV2 pandemic through clinical studies using various protocols. METHODS We present data from a cohort of 267 hospitalized severe COVID-19 patients who received CCP. No transfusion-related complications were reported, indicating the overall safety of CCP therapy. RESULTS Patients who eventually died from COVID-19 received CCP significantly later (3.95 versus 5.22 days after hospital admission) and had higher interleukin 6 (IL-6) levels (28.9 pg/ml versus 102.5 pg/ml) than those who survived. In addition, CCP transfusion caused a significant reduction in the overall inflammatory status of the patients regardless of the severity of disease or outcome, as evidenced by decreasing C-reactive protein, IL6 and ferritin levels. CONCLUSION We conclude that CCP transfusion is a safe and effective supplementary treatment modality for hospitalized COVID-19 patients characterized by better expected outcome if applied as early as possible. We also observed that IL-6 may be a suitable laboratory parameter for patient selection and monitoring of CCP therapy effectiveness.
Collapse
Affiliation(s)
- Eszter Fodor
- Orthosera Kft, Budapest, 1149 Hungary
- Univesity of Physical Education, Budapest, 1223 Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, Budapest, 1083 Hungary
| | - Zsolt Iványi
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, 1082 Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, University of Pécs, Budapest, 7643 Hungary
| | | | - István Hornyák
- Instute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Mira Ambrus
- Univesity of Physical Education, Budapest, 1223 Hungary
| | - Ágnes Sárkány
- Szent György University Teaching Hospital, Székesfehérvár, 8000 Hungary
| | - Árpád Skázel
- Szent György University Teaching Hospital, Székesfehérvár, 8000 Hungary
| | - Ágnes Madár
- Univesity of Physical Education, Budapest, 1223 Hungary
| | | | - Gábor Kemenesi
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Fanni Földes
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Sándor Nagy
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Andrea Matusovits
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Nacsa János
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Attila Tordai
- Department of Transfusiology, Semmelweis University, Budapest, 1089 Hungary
| | - Ferenc Jakab
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Zsombor Lacza
- Orthosera Kft, Budapest, 1149 Hungary
- Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Univesity of Physical Education, Budapest, 1223 Hungary
| |
Collapse
|
7
|
Ortigoza MB, Yoon H, Goldfeld KS, Troxel AB, Daily JP, Wu Y, Li Y, Wu D, Cobb GF, Baptiste G, O'Keeffe M, Corpuz MO, Ostrosky-Zeichner L, Amin A, Zacharioudakis IM, Jayaweera DT, Wu Y, Philley JV, Devine MS, Desruisseaux MS, Santin AD, Anjan S, Mathew R, Patel B, Nigo M, Upadhyay R, Kupferman T, Dentino AN, Nanchal R, Merlo CA, Hager DN, Chandran K, Lai JR, Rivera J, Bikash CR, Lasso G, Hilbert TP, Paroder M, Asencio AA, Liu M, Petkova E, Bragat A, Shaker R, McPherson DD, Sacco RL, Keller MJ, Grudzen CR, Hochman JS, Pirofski LA, Parameswaran L, Corcoran AT, Rohatgi A, Wronska MW, Wu X, Srinivasan R, Deng FM, Filardo TD, Pendse J, Blaser SB, Whyte O, Gallagher JM, Thomas OE, Ramos D, Sturm-Reganato CL, Fong CC, Daus IM, Payoen AG, Chiofolo JT, Friedman MT, Wu DW, Jacobson JL, Schneider JG, Sarwar UN, Wang HE, Huebinger RM, Dronavalli G, Bai Y, Grimes CZ, Eldin KW, Umana VE, Martin JG, Heath TR, Bello FO, Ransford DL, Laurent-Rolle M, Shenoi SV, Akide-Ndunge OB, Thapa B, Peterson JL, Knauf K, Patel SU, Cheney LL, Tormey CA, Hendrickson JE. Efficacy and Safety of COVID-19 Convalescent Plasma in Hospitalized Patients: A Randomized Clinical Trial. JAMA Intern Med 2022; 182:115-126. [PMID: 34901997 PMCID: PMC8669605 DOI: 10.1001/jamainternmed.2021.6850] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
IMPORTANCE There is clinical equipoise for COVID-19 convalescent plasma (CCP) use in patients hospitalized with COVID-19. OBJECTIVE To determine the safety and efficacy of CCP compared with placebo in hospitalized patients with COVID-19 receiving noninvasive supplemental oxygen. DESIGN, SETTING, AND PARTICIPANTS CONTAIN COVID-19, a randomized, double-blind, placebo-controlled trial of CCP in hospitalized adults with COVID-19, was conducted at 21 US hospitals from April 17, 2020, to March 15, 2021. The trial enrolled 941 participants who were hospitalized for 3 or less days or presented 7 or less days after symptom onset and required noninvasive oxygen supplementation. INTERVENTIONS A unit of approximately 250 mL of CCP or equivalent volume of placebo (normal saline). MAIN OUTCOMES AND MEASURES The primary outcome was participant scores on the 11-point World Health Organization (WHO) Ordinal Scale for Clinical Improvement on day 14 after randomization; the secondary outcome was WHO scores determined on day 28. Subgroups were analyzed with respect to age, baseline WHO score, concomitant medications, symptom duration, CCP SARS-CoV-2 titer, baseline SARS-CoV-2 serostatus, and enrollment quarter. Outcomes were analyzed using a bayesian proportional cumulative odds model. Efficacy of CCP was defined as a cumulative adjusted odds ratio (cOR) less than 1 and a clinically meaningful effect as cOR less than 0.8. RESULTS Of 941 participants randomized (473 to placebo and 468 to CCP), 556 were men (59.1%); median age was 63 years (IQR, 52-73); 373 (39.6%) were Hispanic and 132 (14.0%) were non-Hispanic Black. The cOR for the primary outcome adjusted for site, baseline risk, WHO score, age, sex, and symptom duration was 0.94 (95% credible interval [CrI], 0.75-1.18) with posterior probability (P[cOR<1] = 72%); the cOR for the secondary adjusted outcome was 0.92 (95% CrI, 0.74-1.16; P[cOR<1] = 76%). Exploratory subgroup analyses suggested heterogeneity of treatment effect: at day 28, cORs were 0.72 (95% CrI, 0.46-1.13; P[cOR<1] = 93%) for participants enrolled in April-June 2020 and 0.65 (95% CrI, 0.41 to 1.02; P[cOR<1] = 97%) for those not receiving remdesivir and not receiving corticosteroids at randomization. Median CCP SARS-CoV-2 neutralizing titer used in April to June 2020 was 1:175 (IQR, 76-379). Any adverse events (excluding transfusion reactions) were reported for 39 (8.2%) placebo recipients and 44 (9.4%) CCP recipients (P = .57). Transfusion reactions occurred in 2 (0.4) placebo recipients and 8 (1.7) CCP recipients (P = .06). CONCLUSIONS AND RELEVANCE In this trial, CCP did not meet the prespecified primary and secondary outcomes for CCP efficacy. However, high-titer CCP may have benefited participants early in the pandemic when remdesivir and corticosteroids were not in use. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04364737.
Collapse
Affiliation(s)
- Mila B Ortigoza
- Division of Infectious Disease, Department of Medicine, NYU Grossman School of Medicine, New York, New York.,Department of Microbiology, NYU Grossman School of Medicine, New York, New York
| | - Hyunah Yoon
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Keith S Goldfeld
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Andrea B Troxel
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Johanna P Daily
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Yinxiang Wu
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Yi Li
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Danni Wu
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Gia F Cobb
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Gillian Baptiste
- Department of Surgery, NYU Grossman School of Medicine, New York, New York
| | - Mary O'Keeffe
- Department of Medicine, NYU Long Island School of Medicine, Mineola, New York
| | - Marilou O Corpuz
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Luis Ostrosky-Zeichner
- Division of Infectious Disease, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Amee Amin
- Department of Emergency Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Ioannis M Zacharioudakis
- Division of Infectious Disease, Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Dushyantha T Jayaweera
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida.,Miami Clinical and Translational Science Institute, University of Miami Miller School of Medicine Miami, Florida
| | - Yanyun Wu
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida
| | - Julie V Philley
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Tyler, UTHealth East Texas, Tyler
| | - Megan S Devine
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Tyler, UTHealth East Texas, Tyler
| | - Mahalia S Desruisseaux
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Alessandro D Santin
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut
| | - Shweta Anjan
- Division of Infectious Disease, Department of Medicine, University of Miami Miller School of Medicine, Miami, Florida
| | - Reeba Mathew
- Division of Critical Care, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Bela Patel
- Division of Critical Care, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Masayuki Nigo
- Division of Infectious Disease, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Rabi Upadhyay
- Department of Medicine, NYU Grossman School of Medicine, New York, New York.,Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| | - Tania Kupferman
- Division of Infectious Disease, Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Andrew N Dentino
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg
| | - Rahul Nanchal
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee
| | - Christian A Merlo
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - David N Hager
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Jonathan R Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Johanna Rivera
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Chowdhury R Bikash
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Timothy P Hilbert
- Department of Pathology, NYU Grossman School of Medicine, New York, New York
| | - Monika Paroder
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York
| | - Andrea A Asencio
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Mengling Liu
- Department of Population Health, NYU Grossman School of Medicine, New York, New York.,Department of Environmental Health, NYU Grossman School of Medicine, New York, New York
| | - Eva Petkova
- Department of Population Health, NYU Grossman School of Medicine, New York, New York.,Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York.,Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Alexander Bragat
- Clinical Research Information Technology and Informatics, NYU Grossman School of Medicine, New York, New York
| | - Reza Shaker
- Clinical and Translational Science Institute of Southern Wisconsin, Medical College of Wisconsin Milwaukee
| | - David D McPherson
- Center for Clinical and Translational Sciences, Division of Cardiovascular Medicine, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Ralph L Sacco
- Miami Clinical and Translational Science Institute, University of Miami Miller School of Medicine Miami, Florida
| | - Marla J Keller
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.,Harold and Muriel Block Institute for Clinical and Translational Research, Albert Einstein College of Medicine and Montefiore Medical Center Bronx, New York
| | - Corita R Grudzen
- Ronald O. Perelman Department of Emergency Medicine, NYU Grossman School of Medicine, New York, New York.,NYC Health and Hospitals Corporation Clinical and Translational Science Institute, NYU Grossman School of Medicine, New York, New York
| | - Judith S Hochman
- NYC Health and Hospitals Corporation Clinical and Translational Science Institute, NYU Grossman School of Medicine, New York, New York.,Leon H. Charney Division of Cardiology, Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Liise-Anne Pirofski
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.,Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | | | - Lalitha Parameswaran
- Division of Infectious Disease, Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Anthony T Corcoran
- Department of Urology, NYU Long Island School of Medicine, Mineola, New York
| | - Abhinav Rohatgi
- Department of Medicine, NYU Long Island School of Medicine, Mineola, New York
| | - Marta W Wronska
- Department of Medicine, NYU Long Island School of Medicine, Mineola, New York
| | - Xinyuan Wu
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Ranjini Srinivasan
- Department of Pediatrics, NYU Grossman School of Medicine, New York, New York
| | - Fang-Ming Deng
- Department of Pathology, NYU Grossman School of Medicine, New York, New York
| | - Thomas D Filardo
- Division of Infectious Disease, Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Jay Pendse
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Simone B Blaser
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Olga Whyte
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | | | - Ololade E Thomas
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Danibel Ramos
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | | | - Charlotte C Fong
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Ivy M Daus
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | | | - Joseph T Chiofolo
- Department of Pathology, NYU Long Island School of Medicine, Mineola, New York
| | - Mark T Friedman
- Department of Pathology, NYU Long Island School of Medicine, Mineola, New York
| | - Ding Wen Wu
- Department of Pathology, NYU Grossman School of Medicine, New York, New York
| | - Jessica L Jacobson
- Department of Pathology, NYU Grossman School of Medicine, New York, New York
| | - Jeffrey G Schneider
- Department of Medicine, NYU Long Island School of Medicine, Mineola, New York
| | - Uzma N Sarwar
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York.,Pfizer Vaccine Clinical Research and Development, Pfizer Inc, Pearl River, New York
| | - Henry E Wang
- Department of Emergency Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston.,Department of Emergency Medicine, The Ohio State University, Ohio
| | - Ryan M Huebinger
- Department of Emergency Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Goutham Dronavalli
- Division of Critical Care, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Yu Bai
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Carolyn Z Grimes
- Division of Infectious Disease, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Karen W Eldin
- Department of Pathology and Laboratory Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Virginia E Umana
- Division of Infectious Disease, Department of Internal Medicine, The University of Texas Health Science Center at Houston, McGovern Medical School, Houston
| | - Jessica G Martin
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg
| | - Timothy R Heath
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg
| | - Fatimah O Bello
- Department of Internal Medicine, The University of Texas Rio Grande Valley, Edinburg
| | - Daru Lane Ransford
- Miami Clinical and Translational Science Institute, University of Miami Miller School of Medicine Miami, Florida
| | - Maudry Laurent-Rolle
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Sheela V Shenoi
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Oscar Bate Akide-Ndunge
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Bipin Thapa
- Department of Medicine, Medical College of Wisconsin, Milwaukee
| | - Jennifer L Peterson
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Medical College of Wisconsin, Milwaukee
| | - Kelly Knauf
- Clinical and Translational Science Institute of Southern Wisconsin, Medical College of Wisconsin Milwaukee
| | - Shivani U Patel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Laura L Cheney
- Division of Infectious Disease, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York
| | - Christopher A Tormey
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Jeanne E Hendrickson
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut.,Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
8
|
Lasso G, Khan S, Allen SA, Mariano M, Florez C, Orner EP, Quiroz JA, Quevedo G, Massimi A, Hegde A, Wirchnianski AS, Bortz RH, Malonis RJ, Georgiev GI, Tong K, Herrera NG, Morano NC, Garforth SJ, Malaviya A, Khokhar A, Laudermilch E, Dieterle ME, Fels JM, Haslwanter D, Jangra RK, Barnhill J, Almo SC, Chandran K, Lai JR, Kelly L, Daily JP, Vergnolle O. Longitudinally monitored immune biomarkers predict the timing of COVID-19 outcomes. PLoS Comput Biol 2022; 18:e1009778. [PMID: 35041647 PMCID: PMC8812869 DOI: 10.1371/journal.pcbi.1009778] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 02/03/2022] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
The clinical outcome of SARS-CoV-2 infection varies widely between individuals. Machine learning models can support decision making in healthcare by assessing fatality risk in patients that do not yet show severe signs of COVID-19. Most predictive models rely on static demographic features and clinical values obtained upon hospitalization. However, time-dependent biomarkers associated with COVID-19 severity, such as antibody titers, can substantially contribute to the development of more accurate outcome models. Here we show that models trained on immune biomarkers, longitudinally monitored throughout hospitalization, predicted mortality and were more accurate than models based on demographic and clinical data upon hospital admission. Our best-performing predictive models were based on the temporal analysis of anti-SARS-CoV-2 Spike IgG titers, white blood cell (WBC), neutrophil and lymphocyte counts. These biomarkers, together with C-reactive protein and blood urea nitrogen levels, were found to correlate with severity of disease and mortality in a time-dependent manner. Shapley additive explanations of our model revealed the higher predictive value of day post-symptom onset (PSO) as hospitalization progresses and showed how immune biomarkers contribute to predict mortality. In sum, we demonstrate that the kinetics of immune biomarkers can inform clinical models to serve as a powerful monitoring tool for predicting fatality risk in hospitalized COVID-19 patients, underscoring the importance of contextualizing clinical parameters according to their time post-symptom onset.
Collapse
Affiliation(s)
- Gorka Lasso
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Saad Khan
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Stephanie A. Allen
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Margarette Mariano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Catalina Florez
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Chemistry and Life Science, United States Military Academy at West Point, West Point, New York, United States of America
| | - Erika P. Orner
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jose A. Quiroz
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Gregory Quevedo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Aldo Massimi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Aditi Hegde
- Eastchester High School, 2 Stewart Place, Eastchester, New York, United States of America
| | - Ariel S. Wirchnianski
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Robert H. Bortz
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Ryan J. Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - George I. Georgiev
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Karen Tong
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Natalia G. Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Nicholas C. Morano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Scott J. Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Avinash Malaviya
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Ahmed Khokhar
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Ethan Laudermilch
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - M. Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - J. Maximilian Fels
- Department of Cell Biology, Harvard Medical School, Boston, Cambridge, Massachusetts, United States of America
- Department of Microbiology, Harvard Medical School, Boston, Cambridge, Massachusetts, United States of America
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Cambridge, Massachusetts, United States of America
| | - Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jason Barnhill
- Department of Chemistry and Life Science, United States Military Academy at West Point, West Point, New York, United States of America
- Department of Radiology and Radiological Services, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Libusha Kelly
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Johanna P. Daily
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Olivia Vergnolle
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
9
|
Focosi D, Franchini M, Pirofski LA, Maggi F, Casadevall A. Is SARS-CoV-2 viral clearance in nasopharyngeal swabs an appropriate surrogate marker for clinical efficacy of neutralising antibody-based therapeutics? Rev Med Virol 2021; 32:e2314. [PMID: 34861088 DOI: 10.1002/rmv.2314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022]
Abstract
Viral clearance is likely the best way to assess the efficacy of antibody-based therapies. Although antibodies can mediate a variety of effects that include modulation of inflammation, the demonstration of viral clearance provides an accessible and measurable parameter that can be used to evaluate efficacy and determine dosing. Therefore, it is important to ascertain the ability of monoclonal antibodies and convalescent plasma to effect viral clearance. For COVID-19, which is caused by the respiratory virus SARS-CoV-2, the most common assay to assess viral clearance is via a nasopharyngeal swab (NPS). However, assessment of antibody efficacy by sampling this site may be misleading because it may not be as accessible to serum antibodies as respiratory secretions or circulating blood. Adding to the complexity of assessing the efficacy of administered antibody, particularly in randomised controlled trials (RCTs) that enroled patients at different times after the onset of COVID-19 symptoms, viral clearance may also be mediated by endogenous antibody. In this article we critically review available data on viral clearance in RCTs, matched control studies, case series and case reports of antibody therapies in an attempt to identify variables that contribute to antibody efficacy and suggest optimal strategies for future studies.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Departments of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York, USA
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.,Laboratory of Microbiology, ASST Sette Laghi, Varese, Italy
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Herman JD, Wang C, Loos C, Yoon H, Rivera J, Eugenia Dieterle M, Haslwanter D, Jangra RK, Bortz RH, Bar KJ, Julg B, Chandran K, Lauffenburger D, Pirofski LA, Alter G. Functional convalescent plasma antibodies and pre-infusion titers shape the early severe COVID-19 immune response. Nat Commun 2021; 12:6853. [PMID: 34824251 PMCID: PMC8617042 DOI: 10.1038/s41467-021-27201-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 10/28/2021] [Indexed: 01/10/2023] Open
Abstract
Transfer of convalescent plasma (CP) had been proposed early during the SARS-CoV-2 pandemic as an accessible therapy, yet trial results worldwide have been mixed, potentially due to the heterogeneous nature of CP. Here we perform deep profiling of SARS-CoV-2-specific antibody titer, Fc-receptor binding, and Fc-mediated functional assays in CP units, as well as in plasma from hospitalized COVID-19 patients before and after CP administration. The profiling results show that, although all recipients exhibit expanded SARS-CoV-2-specific humoral immune responses, CP units contain more functional antibodies than recipient plasma. Meanwhile, CP functional profiles influence the evolution of recipient humoral immunity in conjuncture with the recipient's pre-existing SARS-CoV2-specific antibody titers: CP-derived SARS-CoV-2 nucleocapsid-specific antibody functions are associated with muted humoral immune evolution in patients with high titer anti-spike IgG. Our data thus provide insights into the unexpected impact of CP-derived functional anti-spike and anti-nucleocapsid antibodies on the evolution of SARS-CoV-2-specific response following severe infection.
Collapse
Affiliation(s)
- Jonathan D Herman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Brigham and Women's Hospital, Boston, MA, USA
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Loos
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hyunah Yoon
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Johanna Rivera
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - M Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rohit K Jangra
- Department of Microbiology and Immunology, Louisiana State University Health Science Center-Shreveport, Shreveport, LA, USA
| | - Robert H Bortz
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Katharine J Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Sarif J, Raychaudhuri D, D'Rozario R, Bandopadhyay P, Singh P, Mehta P, Hoque MA, Sinha BP, Kushwaha M, Sahni S, Devi P, Chattopadhyay P, Paul SR, Ray Y, Chaudhuri K, Banerjee S, Majumdar D, Saha B, Sarkar BS, Bhattacharya P, Chatterjee S, Paul S, Ghosh P, Pandey R, Sengupta S, Ganguly D. Plasma Gradient of Soluble Urokinase-Type Plasminogen Activator Receptor Is Linked to Pathogenic Plasma Proteome and Immune Transcriptome and Stratifies Outcomes in Severe COVID-19. Front Immunol 2021; 12:738093. [PMID: 34777349 PMCID: PMC8581406 DOI: 10.3389/fimmu.2021.738093] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/06/2021] [Indexed: 12/11/2022] Open
Abstract
Disease caused by SARS-CoV-2 coronavirus (COVID-19) led to significant morbidity and mortality worldwide. A systemic hyper-inflammation characterizes severe COVID-19 disease, often associated with acute respiratory distress syndrome (ARDS). Blood biomarkers capable of risk stratification are of great importance in effective triage and critical care of severe COVID-19 patients. Flow cytometry and next-generation sequencing were done on peripheral blood cells and urokinase-type plasminogen activator receptor (suPAR), and cytokines were measured from and mass spectrometry-based proteomics was done on plasma samples from an Indian cohort of COVID-19 patients. Publicly available single-cell RNA sequencing data were analyzed for validation of primary data. Statistical analyses were performed to validate risk stratification. We report here higher plasma abundance of suPAR, expressed by an abnormally expanded myeloid cell population, in severe COVID-19 patients with ARDS. The plasma suPAR level was found to be linked to a characteristic plasma proteome, associated with coagulation disorders and complement activation. Receiver operator characteristic curve analysis to predict mortality identified a cutoff value of suPAR at 1,996.809 pg/ml (odds ratio: 2.9286, 95% confidence interval 1.0427-8.2257). Lower-than-cutoff suPAR levels were associated with a differential expression of the immune transcriptome as well as favorable clinical outcomes, in terms of both survival benefit (hazard ratio: 0.3615, 95% confidence interval 0.1433-0.912) and faster disease remission in our patient cohort. Thus, we identified suPAR as a key pathogenic circulating molecule linking systemic hyperinflammation to the hypercoagulable state and stratifying clinical outcomes in severe COVID-19 patients with ARDS.
Collapse
Affiliation(s)
- Jafar Sarif
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Deblina Raychaudhuri
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Ranit D'Rozario
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Purbita Bandopadhyay
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Praveen Singh
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Priyanka Mehta
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Md Asmaul Hoque
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Bishnu Prasad Sinha
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Manoj Kushwaha
- Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shweta Sahni
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Priti Devi
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Partha Chattopadhyay
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shekhar Ranjan Paul
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Yogiraj Ray
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India.,Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Kausik Chaudhuri
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Sayantan Banerjee
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Debajyoti Majumdar
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India.,Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Bibhuti Saha
- Department of Tropical Medicine, School of Tropical Medicine, Kolkata, India
| | - Biswanath Sharma Sarkar
- Department of Medicine, Infectious Diseases and Beliaghata General (ID & BG) Hospital, Kolkata, India
| | - Prasun Bhattacharya
- Department of Immunohematology and Blood Transfusion, Medical College, Kolkata, India
| | - Shilpak Chatterjee
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Sandip Paul
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Pramit Ghosh
- Department of Community Medicine, Deben Mahata Government Medical College & Hospital, Purulia, India
| | - Rajesh Pandey
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Shantanu Sengupta
- Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.,Cardiorespiratory Disease Biology, Council of Scientific & Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Dipyaman Ganguly
- Indian Institute of Chemical Biology (IICB)-Translational Research Unit of Excellence, Council of Scientific & Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India.,Department of Biological Sciences, Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
12
|
Körper S, Appl T, Jahrsdörfer B, Lotfi R, Rojewski M, Wuchter P, Tonn T, Bakchoul T, Karatas M, Schmidt M, Klüter H, Seifried E, Schrezenmeier H. Randomisierte Studien zum Einsatz von Rekonvaleszentenplasma bei COVID-19: eine Standortbestimmung. TRANSFUSIONSMEDIZIN 2021. [DOI: 10.1055/a-1521-7884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungPlasma von genesenen Spendern mit COVID-19 (COVID-19 Convalescent Plasma, CCP) wurde als Behandlungsoption für Patienten mit COVID-19 in Betracht gezogen. In der ersten Phase der Pandemie wurden mehrere Fallberichte und Fallkontrollstudien mit Hinweisen auf eine therapeutische Wirkung veröffentlicht. Inzwischen liegen die Ergebnisse zahlreicher randomisierter Studien vor. Die Studien unterscheiden sich in vielen Aspekten, u. a. in den Patientenpopulationen, die von ambulanten Patienten mit mildem COVID-19 bis zu kritisch Kranken reichten, wie auch den Endpunkten. Ebenso war der Behandlungsstandard innerhalb der klinischen Studien sehr unterschiedlich. Vor allem aber unterschied sich das Prüfpräparat CCP erheblich in Bezug auf das Behandlungsschema, das Volumen und den Gehalt an Antikörpern. Im Folgenden werden wir die Ergebnisse der bisher publizierten randomisierten Studien diskutieren. Aus den bisher veröffentlichten Ergebnissen lässt sich eine Wirksamkeit von CCP
ableiten, sofern es sehr hohe Titer neutralisierender Antikörper enthält und früh im Krankheitsverlauf verabreicht wird. COVID-19-Rekonvaleszenten-Plasma ist noch keine Routinebehandlung und sollte möglichst weiter in klinischen Studien untersucht werden. Neu konzipierte Studien sollten sich auf die frühe Anwendung von CCP mit einem hohen Gehalt an neutralisierenden Antikörpern bei Patienten mit hohem Risiko für eine Progression zu einer schweren COVID-19-Erkrankung konzentrieren und wichtige Begleitmedikationen kontrollieren.
Collapse
Affiliation(s)
- Sixten Körper
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Thomas Appl
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Bernd Jahrsdörfer
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Ramin Lotfi
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Markus Rojewski
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Patrick Wuchter
- Institut für Transfusionsmedizin und Immunologie, Medizinische Fakultät Mannheim, Universität Heidelberg; DRK-Blutspendedienst Baden-Württemberg – Hessen, Deutschland
| | - Torsten Tonn
- Experimentelle Transfusionsmedizin, Technische Universität Dresden, Deutsches Rotes Kreuz Bluttransfusionsdienst Nord-Ost gGmbH, Dresden, Deutschland
| | - Tamam Bakchoul
- Institut für klinische und experimentelle Transfusionsmedizin, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - Mesut Karatas
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Michael Schmidt
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Harald Klüter
- Institut für Transfusionsmedizin und Immunologie, Medizinische Fakultät Mannheim, Universität Heidelberg; DRK-Blutspendedienst Baden-Württemberg – Hessen, Deutschland
| | - Erhard Seifried
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Hubert Schrezenmeier
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| |
Collapse
|
13
|
Arnold Egloff SA, Junglen A, Restivo JS, Wongskhaluang M, Martin C, Doshi P, Schlauch D, Fromell G, Sears LE, Correll M, Burris HA, LeMaistre CF. Convalescent plasma associates with reduced mortality and improved clinical trajectory in patients hospitalized with COVID-19. J Clin Invest 2021; 131:e151788. [PMID: 34464352 DOI: 10.1172/jci151788] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUNDEvidence supporting convalescent plasma (CP), one of the first investigational treatments for coronavirus disease 2019 (COVID-19), has been inconclusive, leading to conflicting recommendations. The primary objective was to perform a comparative effectiveness study of CP for all-cause, in-hospital mortality in patients with COVID-19.METHODSThe multicenter, electronic health records-based, retrospective study included 44,770 patients hospitalized with COVID-19 in one of 176 HCA Healthcare-affiliated community hospitals. Coarsened exact matching (1:k) was employed, resulting in a sample of 3774 CP and 10,687 comparison patients.RESULTSExamination of mortality using a shared frailty model, controlling for concomitant medications, date of admission, and days from admission to transfusion, demonstrated a significant association of CP with lower mortality risk relative to the comparison group (adjusted hazard ratio [aHR] = 0.71; 95% CI, 0.59-0.86; P < 0.001). Examination of patient risk trajectories, represented by 400 clinico-demographic features from our real-time risk model (RTRM), indicated that patients who received CP recovered more quickly. The stratification of days to transfusion revealed that CP within 3 days after admission, but not within 4 to 7 days, was associated with a significantly lower mortality risk (aHR = 0.53; 95% CI, 0.47-0.60; P < 0.001). CP serology level was inversely associated with mortality when controlling for its interaction with days to transfusion (HR = 0.998; 95% CI, 0.997-0.999; P = 0.013), yet it did not reach univariable significance.CONCLUSIONSThis large, diverse, multicenter cohort study demonstrated that CP, compared with matched controls, is significantly associated with reduced risk of in-hospital mortality. These observations highlight the utility of real-world evidence and suggest the need for further evaluation prior to abandoning CP as a viable therapy for COVID-19.FUNDINGThis research was supported in whole by HCA Healthcare and/or an HCA Healthcare-affiliated entity, including Sarah Cannon and Genospace.
Collapse
Affiliation(s)
- Shanna A Arnold Egloff
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Angela Junglen
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Joseph Sa Restivo
- HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | | | - Casey Martin
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Pratik Doshi
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Daniel Schlauch
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Gregg Fromell
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Lindsay E Sears
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Mick Correll
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Howard A Burris
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Charles F LeMaistre
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| |
Collapse
|
14
|
Cao H, Ming L, Chen L, Zhu X, Shi Y. The Effectiveness of Convalescent Plasma for the Treatment of Novel Corona Virus Disease 2019: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:641429. [PMID: 34646833 PMCID: PMC8502818 DOI: 10.3389/fmed.2021.641429] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Coronavirus disease 2019 (COVID-19), sweeping across the world, has created a worldwide pandemic. Effective treatments of COVID-19 are extremely urgent. Objective: To analyze the efficacy and safety of convalescent plasma (CCP) on patients with COVID-19. Methods: All the relevant studies were searched from PubMed, EMBASE,Cochrane library, Scopus, Web of Science, CBM, CNKI, Wan fang, VIP, Medrxiv, Biorxiv, and SSRN on July 19, 2021. PICOS criteria were as follows: (P) the study interests were human subjects with the infection of COVID-19; (I) the intervention of interest was CCP; (C) comparator treatments contained placebo, sham therapy, and standard treatment; (O) the primary outcome was mortality rates by the novel coronavirus. The secondary outcomes included the incidence of serious adverse events, the rate of ICU admission and mechanical ventilation (MV); the length of hospital stay; the duration of MV and ICU stay; the antibody levels, inflammatory factor levels, and viral loads. (S) Only randomized controlled trials (RCTs) of CCP were included. Subanalysis, quality assessment, sensitive analysis, and publication bias were conducted by two reviewers independently. Results: Sixteen RCTs were included and enrolled a total of 16,296 participants in this meta-analysis. The pooled data showed that no significant difference was observed in reducing the rate of overall mortality between CCP treatment group and placebo group (OR 0.96; 95% CI 0.90 to 1.03; p = 0.30; I 2 = 6%). According to the results of subgroup analysis, severe or critical patients with CCP showed significant difference in reducing the 28-day mortality of compared with placebo (OR 0.58, 95% CI 0.36 to 0.93, p = 0.02, I 2 = 0%). CCP groups have a significantly shorter duration of MV compared with the control group (weighted MD -1.00, 95% CI -1.86 to -0.14 d p = 0.02, I 2 = 0%). No significant difference was observed in the length of hospital stay, the duration of ICU, and the rate of ICU and MV. There is no conclusive evidence about the safety of CCP. Conclusion: Convalescent plasma can significantly reduce the 28-day mortality of severe or critical COVID-19 patients and the duration of MV. However, more evidence was needed to prove the safety of convalescent plasma.
Collapse
Affiliation(s)
- Huiling Cao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Ming
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Long Chen
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xingwang Zhu
- Department of Pediatrics, Jiulongpo People's Hospital, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
15
|
de Candia P, Prattichizzo F, Garavelli S, La Grotta R, De Rosa A, Pontarelli A, Parrella R, Ceriello A, Matarese G. Effect of time and titer in convalescent plasma therapy for COVID-19. iScience 2021; 24:102898. [PMID: 34316549 PMCID: PMC8297982 DOI: 10.1016/j.isci.2021.102898] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/18/2021] [Accepted: 07/19/2021] [Indexed: 12/15/2022] Open
Abstract
The clinical benefit of convalescent plasma (CP) for patients with coronavirus disease (COVID)-19 is still debated. In this systematic review and meta-analysis, we selected 10 randomized clinical trials (RCTs) and 15 non-randomized studies (total number of patients = 22,591) of CP treatment and evaluated two different scenarios: (1) disease stage of plasma recipients and (2) donated plasma antibody titer, considering all-cause mortality at the latest follow-up. Our results show that, when provided at early stages of the disease, CP significantly reduced mortality: risk ratio (RR) 0.72 (0.68, 0.77), p < 0.00001, while provided in severe or critical conditions, it did not (RR: 0.94 [0.86, 1.04], p = 0.22). On the other hand, the benefit on mortality was not increased by using plasma with a high-antibody titer compared with unselected plasma. This meta-analysis might promote CP usage in patients with early-stage COVID-19 in further RCTs to maximize its benefit in decreasing mortality, especially in less affluent countries. The benefit of convalescent plasma (CP) for patients with COVID-19 is still debated Only when provided at early disease stages, CP reduced COVID-19 mortality CP benefit on mortality does not increase when selecting high-antibody titers Early treatment with CP may maximize its clinical benefit
Collapse
Affiliation(s)
| | | | - Silvia Garavelli
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Naples, Italy
| | | | - Annunziata De Rosa
- Dipartimento di Malattie Infettive ed Emergenze Infettive, Divisione di Malattie Infettive Respiratorie, Ospedale Cotugno, AORN dei Colli, 80131 Naples, Italy
| | - Agostina Pontarelli
- Dipartimento di Malattie Infettive ed Emergenze Infettive, Divisione di Malattie Infettive Respiratorie, Ospedale Cotugno, AORN dei Colli, 80131 Naples, Italy
| | - Roberto Parrella
- Dipartimento di Malattie Infettive ed Emergenze Infettive, Divisione di Malattie Infettive Respiratorie, Ospedale Cotugno, AORN dei Colli, 80131 Naples, Italy
| | | | - Giuseppe Matarese
- Istituto per l'Endocrinologia e l'Oncologia Sperimentale, Consiglio Nazionale delle Ricerche (IEOS-CNR), 80131 Naples, Italy.,Treg Cell Lab, Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Naples, Italy
| |
Collapse
|
16
|
Klassen SA, Senefeld JW, Senese KA, Johnson PW, Wiggins CC, Baker SE, van Helmond N, Bruno KA, Pirofski LA, Shoham S, Grossman BJ, Henderson JP, Wright RS, Fairweather D, Paneth NS, Carter RE, Casadevall A, Joyner MJ. Convalescent Plasma Therapy for COVID-19: A Graphical Mosaic of the Worldwide Evidence. Front Med (Lausanne) 2021; 8:684151. [PMID: 34164419 PMCID: PMC8215127 DOI: 10.3389/fmed.2021.684151] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Convalescent plasma has been used worldwide to treat patients hospitalized with coronavirus disease 2019 (COVID-19) and prevent disease progression. Despite global usage, uncertainty remains regarding plasma efficacy, as randomized controlled trials (RCTs) have provided divergent evidence regarding the survival benefit of convalescent plasma. Here, we argue that during a global health emergency, the mosaic of evidence originating from multiple levels of the epistemic hierarchy should inform contemporary policy and healthcare decisions. Indeed, worldwide matched-control studies have generally found convalescent plasma to improve COVID-19 patient survival, and RCTs have demonstrated a survival benefit when transfused early in the disease course but limited or no benefit later in the disease course when patients required greater supportive therapies. RCTs have also revealed that convalescent plasma transfusion contributes to improved symptomatology and viral clearance. To further investigate the effect of convalescent plasma on patient mortality, we performed a meta-analytical approach to pool daily survival data from all controlled studies that reported Kaplan-Meier survival plots. Qualitative inspection of all available Kaplan-Meier survival data and an aggregate Kaplan-Meier survival plot revealed a directionally consistent pattern among studies arising from multiple levels of the epistemic hierarchy, whereby convalescent plasma transfusion was generally associated with greater patient survival. Given that convalescent plasma has a similar safety profile as standard plasma, convalescent plasma should be implemented within weeks of the onset of future infectious disease outbreaks.
Collapse
Affiliation(s)
- Stephen A. Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Katherine A. Senese
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Patrick W. Johnson
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | - Chad C. Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Sarah E. Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Noud van Helmond
- Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Health Care, Camden, NJ, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY, United States
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Jeffrey P. Henderson
- Division of Infectious Diseases, Department of Medicine, Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - R. Scott Wright
- Department of Cardiovascular Medicine, Human Research Protection Program, Mayo Clinic, Rochester, MN, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Rickey E. Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | - Arturo Casadevall
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
17
|
Klassen SA, Senefeld JW, Johnson PW, Carter RE, Wiggins CC, Shoham S, Grossman BJ, Henderson JP, Musser J, Salazar E, Hartman WR, Bouvier NM, Liu STH, Pirofski LA, Baker SE, van Helmond N, Wright RS, Fairweather D, Bruno KA, Wang Z, Paneth NS, Casadevall A, Joyner MJ. The Effect of Convalescent Plasma Therapy on Mortality Among Patients With COVID-19: Systematic Review and Meta-analysis. Mayo Clin Proc 2021; 96:1262-1275. [PMID: 33958057 PMCID: PMC7888247 DOI: 10.1016/j.mayocp.2021.02.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 02/01/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
To determine the effect of COVID-19 convalescent plasma on mortality, we aggregated patient outcome data from 10 randomized clinical trials, 20 matched control studies, 2 dose-response studies, and 96 case reports or case series. Studies published between January 1, 2020, and January 16, 2021, were identified through a systematic search of online PubMed and MEDLINE databases. Random effects analyses of randomized clinical trials and matched control data demonstrated that patients with COVID-19 transfused with convalescent plasma exhibited a lower mortality rate compared with patients receiving standard treatments. Additional analyses showed that early transfusion (within 3 days of hospital admission) of higher titer plasma is associated with lower patient mortality. These data provide evidence favoring the efficacy of human convalescent plasma as a therapeutic agent in hospitalized patients with COVID-19.
Collapse
Affiliation(s)
- Stephen A Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Patrick W Johnson
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL
| | - Rickey E Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL
| | - Chad C Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Brenda J Grossman
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - Jeffrey P Henderson
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO; Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO
| | - James Musser
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX; Center for Molecular and Translational Human Infectious Diseases, Houston Methodist Research Institute, Houston, TX; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Eric Salazar
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - William R Hartman
- Department of Anesthesiology, University of Wisconsin-Madison School of Medicine and Public Health, Madison, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nicole M Bouvier
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sean T H Liu
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY; Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - Sarah E Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Noud van Helmond
- Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Health Care, Camden, NJ
| | - R Scott Wright
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN; Director, Human Research Protection Program, Mayo Clinic, Rochester, MN
| | | | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL
| | - Zhen Wang
- Evidence-Based Practice Center, Robert D. and Patricia E. Kern Center for Science of Health Care Delivery, Mayo Clinic, Rochester, MN
| | - Nigel S Paneth
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing; Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
18
|
Casadevall A, Dragotakes Q, Johnson PW, Senefeld JW, Klassen SA, Wright RS, Joyner MJ, Paneth N, Carter RE. Convalescent Plasma Use in the United States was inversely correlated with COVID-19 Mortality: Did Plasma Hesitancy cost lives? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.04.07.21255089. [PMID: 33851186 PMCID: PMC8043483 DOI: 10.1101/2021.04.07.21255089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The US Food and Drug Administration authorized Convalescent Plasma (CCP) therapy for hospitalized COVID-19 patients via the Expanded Access Program (EAP) and the Emergency Use Authorization (EUA), leading to use in about 500,000 patients during the first year of the pandemic for the US. METHODS We tracked the number of CCP units dispensed to hospitals by blood banking organizations and correlated that usage with hospital admission and mortality data. RESULTS CCP usage per admission peaked in Fall 2020, with more than 40% of inpatients estimated to have received CCP between late September and early November 2020. However, after randomized controlled trials failed to show a reduction in mortality, CCP usage per admission declined steadily to a nadir of less than 10% in March 2021. We found a strong inverse correlation (r = -0.52, P = 0.002) between CCP usage per hospital admission and deaths occurring two weeks after admission, and this finding was robust to examination of deaths taking place one, two or three weeks after admission. Changes in the number of hospital admissions, SARS-CoV-2 variants, and age of patients could not explain these findings. The retreat from CCP usage might have resulted in as many as 29,000 excess deaths from mid-November 2020 to February 2021. CONCLUSIONS A strong inverse correlation between CCP use and mortality per admission in the USA provides population level evidence consistent with the notion that CCP reduces mortality in COVID-19 and suggests that the recent decline in usage could have resulted in excess deaths.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD
| | - Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD
| | - Patrick W. Johnson
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL
| | - Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Steven A. Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | | | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Nigel Paneth
- Department of Epidemiology and Biostatistics and Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL
| |
Collapse
|
19
|
Bansal V, Mahapure KS, Mehra I, Bhurwal A, Tekin A, Singh R, Gupta I, Rathore SS, Khan H, Deshpande S, Gulati S, Armaly P, Sheraton M, Kashyap R. Mortality Benefit of Convalescent Plasma in COVID-19: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:624924. [PMID: 33898477 PMCID: PMC8062901 DOI: 10.3389/fmed.2021.624924] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 02/17/2021] [Indexed: 02/06/2023] Open
Abstract
Importance/Background: With a scarcity of high-grade evidence for COVID-19 treatment, researchers and health care providers across the world have resorted to classical and historical interventions. Immunotherapy with convalescent plasma (CPT) is one such therapeutic option. Methods: A systematized search was conducted for articles published between December 2019 and 18th January 2021 focusing on convalescent plasma efficacy and safety in COVID-19. The primary outcomes were defined as mortality benefit in patients treated with convalescent plasma compared to standard therapy/placebo. The secondary outcome was pooled mortality rate and the adverse event rate in convalescent plasma-treated patients. Results: A total of 27,706 patients were included in the qualitative analysis, and a total of 3,262 (2,127 in convalescent plasma-treated patients and 1,135 in the non-convalescent plasma/control group) patients died. The quantitative synthesis in 23 studies showed that the odds of mortality in patients who received plasma therapy were significantly lower than those in patients who did not receive plasma therapy [odds ratio (OR) 0.65, 95% confidence interval (CI) 0.53-0.80, p < 0.0001, I 2 = 15%). The mortality benefit remains the same even for 14 trials/prospective studies (OR 0.59, 95% CI 0.43-0.81, p = 0.001, I 2 = 22%) as well as for nine case series/retrospective observational studies (OR 0.78, 95% CI 0.65-0.94, p = 0.01, I 2 = 0%). However, in a subgroup analysis for 10 randomized controlled trials (RCTs), there was no statistically significant reduction in mortality between the CPT group compared to the non-CPT group (OR 0.76, 95% CI 0.53-1.08, p = 0.13, I 2 = 7%). Furthermore, the sensitivity analysis of 10 RCTs, excluding the study with the highest statistical weight, displayed a lower mortality rate compared to that of non-CPT COVID-19 patients (OR 0.64, 95% CI 0.42-0.97, p = 0.04, I 2 = 0%). The observed pooled mortality rate was 12.9% (95% CI 9.7-16.9%), and the pooled adverse event rate was 6.1% (95% CI 3.2-11.6), with significant heterogeneity. Conclusions and Relevance: Our systemic review and meta-analysis suggests that CPT could be an effective therapeutic option with promising evidence on the safety and reduced mortality in concomitant treatment for COVID-19 along with antiviral/antimicrobial drugs, steroids, and other supportive care. Future exploratory studies could benefit from more standardized reporting, especially in terms of the timing of interventions and clinically relevant outcomes, like days until discharge from the hospital and improvement of clinical symptoms.
Collapse
Affiliation(s)
- Vikas Bansal
- Department of Anaesthesiology and Critical Care Medicine, Mayo Clinic, Rochester, MN, United States
| | - Kiran S. Mahapure
- Senior Resident, Department of Plastic Surgery, KAHER J. N. Medical College, Belgaum, India
| | - Ishita Mehra
- Department of Internal Medicine, North Alabama Medical Center, Florence, AL, United States
| | - Abhishek Bhurwal
- Department of Gastroenterology and Hepatology, Rutgers Robert Wood Johnson School of Medicine, New Brunswick, NJ, United States
| | - Aysun Tekin
- Department of Anaesthesiology and Critical Care Medicine, Mayo Clinic, Rochester, MN, United States
| | - Romil Singh
- Department of Internal Medicine, Metropolitan Hospital, Jaipur, India
| | - Ishita Gupta
- Department of Internal Medicine, Dr. Rajendra Prasad Government Medical College, Tanda, India
| | - Sawai Singh Rathore
- Department of Internal Medicine, Dr. Sampurnanand Medical College, Jodhpur, India
| | - Hira Khan
- Department of Internal Medicine, Riphah International University Islamic International Medical College, Rawalpindi, Pakistan
| | - Sohiel Deshpande
- Department of Internal Medicine, Maharashtra Institute of Medical Education and Research, Pune, India
| | - Shivam Gulati
- Department of Internal Medicine, Adesh Institute of Medical Sciences and Research, Bathinda, India
| | - Paige Armaly
- Department of Internal Medicine, University of the West Indies, Nassau, Bahamas
| | - Mack Sheraton
- Department of Emergency Medicine, Trinity West Medical Center MSOPTI EM Program, Steubenville, OH, United States
| | - Rahul Kashyap
- Department of Anaesthesiology and Critical Care Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
20
|
Focosi D, Franchini M. COVID-19 convalescent plasma therapy: hit fast, hit hard! Vox Sang 2021; 116:935-942. [PMID: 33794556 PMCID: PMC8251446 DOI: 10.1111/vox.13091] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022]
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| |
Collapse
|
21
|
Herman JD, Wang C, Loos C, Yoon H, Rivera J, Dieterle ME, Haslwanter D, Jangra RK, Bortz RH, Bar KJ, Julg B, Chandran K, Lauffenburger D, Pirofski LA, Alter G. Functional Antibodies in COVID-19 Convalescent Plasma. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.08.21253157. [PMID: 33758875 PMCID: PMC7987034 DOI: 10.1101/2021.03.08.21253157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
In the absence of an effective vaccine or monoclonal therapeutic, transfer of convalescent plasma (CCP) was proposed early in the SARS-CoV-2 pandemic as an easily accessible therapy. However, despite the global excitement around this historically valuable therapeutic approach, results from CCP trials have been mixed and highly debated. Unlike other therapeutic interventions, CCP represents a heterogeneous drug. Each CCP unit is unique and collected from an individual recovered COVID-19 patient, making the interpretation of therapeutic benefit more complicated. While the prevailing view in the field would suggest that it is administration of neutralizing antibodies via CCP that centrally provides therapeutic benefit to newly infected COVID-19 patients, many hospitalized COVID-19 patients already possess neutralizing antibodies. Importantly, the therapeutic benefit of antibodies can extend far beyond their simple ability to bind and block infection, especially related to their ability to interact with the innate immune system. In our work we deeply profiled the SARS-CoV-2-specific Fc-response in CCP donors, along with the recipients prior to and after CCP transfer, revealing striking SARS-CoV-2 specific Fc-heterogeneity across CCP units and their recipients. However, CCP units possessed more functional antibodies than acute COVID-19 patients, that shaped the evolution of COVID-19 patient humoral profiles via distinct immunomodulatory effects that varied by pre-existing SARS-CoV-2 Spike (S)-specific IgG titers in the patients. Our analysis identified surprising influence of both S and Nucleocapsid (N) specific antibody functions not only in direct antiviral activity but also in anti-inflammatory effects. These findings offer insights for more comprehensive interpretation of correlates of immunity in ongoing large scale CCP trials and for the design of next generation therapeutic design.
Collapse
Affiliation(s)
- Jonathan D. Herman
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Infectious Disease, Brigham and Women’s Hospital, Boston, MA, USA
| | - Chuangqi Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Loos
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hyunah Yoon
- Division of Infectious Diseases, Department of Medicine. Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY,USA
| | - Johanna Rivera
- Division of Infectious Diseases, Department of Medicine. Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY,USA
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - M. Eugenia Dieterle
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Denise Haslwanter
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Rohit K. Jangra
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Robert H. Bortz
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Katharine J. Bar
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Department of Medicine. Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY,USA
- Department of Microbiology and Immunology. Albert Einstein College of Medicine, Bronx, NY, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
22
|
Casadevall A, Pirofski LA, Joyner MJ. The Principles of Antibody Therapy for Infectious Diseases with Relevance for COVID-19. mBio 2021; 12:e03372-20. [PMID: 33653885 PMCID: PMC8092292 DOI: 10.1128/mbio.03372-20] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Antibody therapies such as convalescent plasma and monoclonal antibodies have emerged as major potential therapeutics for coronavirus disease 2019 (COVID-19). Immunoglobulins differ from conventional antimicrobial agents in that they mediate direct and indirect antimicrobial effects that work in concert with other components of the immune system. The field of infectious diseases pioneered antibody therapies in the first half of the 20th century but largely abandoned them with the arrival of conventional antimicrobial therapy. Consequently, much of the knowledge gained from the historical development and use of immunoglobulins such as serum and convalescent antibody therapies was forgotten; principles and practice governing their use were not taught to new generations of medical practitioners, and further development of this modality stalled. This became apparent during the COVID-19 pandemic in the spring of 2020 when convalescent plasma was initially deployed as salvage therapy in patients with severe disease. In retrospect, this was a stage of disease when it was less likely to be effective. Lessons of the past tell us that antibody therapy is most likely to be effective when used early in respiratory diseases. This article puts forth three principles of antibody therapy, namely, specificity, temporal, and quantitative principles, connoting that antibody efficacy requires the administration of specific antibody, given early in course of disease in sufficient amount. These principles are traced to the history of serum therapy for infectious diseases. The application of the specificity, temporal, and quantitative principles to COVID-19 is discussed in the context of current use of antibody therapy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Liise-Anne Pirofski
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York, USA
- Montefiore Medical Center, Bronx, New York, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|