201
|
Lee P, Abernethy A, Shaywitz D, Gundlapalli AV, Weinstein J, Doraiswamy PM, Schulman K, Madhavan S. Digital Health COVID-19 Impact Assessment: Lessons Learned and Compelling Needs. NAM Perspect 2022; 2022:202201c. [PMID: 35402858 PMCID: PMC8970223 DOI: 10.31478/202201c] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023]
|
202
|
Klompas AM, van Helmond N, Juskewitch JE, Pruthi RK, Sexton MA, Soto JCD, Klassen SA, Senese KA, van Buskirk CM, Winters JL, Stubbs JR, Hammel SA, Joyner MJ, Senefeld JW. Coagulation profile of human COVID-19 convalescent plasma. Sci Rep 2022; 12:637. [PMID: 35022488 PMCID: PMC8755772 DOI: 10.1038/s41598-021-04670-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 12/17/2021] [Indexed: 12/27/2022] Open
Abstract
Convalescent plasma is used to treat COVID-19. There are theoretical concerns about the impact of pro-coagulant factors in convalescent plasma on the coagulation cascade particularly among patients with severe COVID-19. The aim of this study was to evaluate the coagulation profile of COVID-19 convalescent plasma. Clotting times and coagulation factor assays were compared between fresh frozen plasma, COVID-19 convalescent plasma, and pathogen-reduced COVID-19 convalescent plasma. Measurements included prothrombin time, activated partial thromboplastin time, thrombin time, fibrinogen, D-dimer, von Willebrand factor activity, von Willebrand factor antigen, coagulation factors II, V, VII-XII, protein S activity, protein C antigen, and alpha-2 plasmin inhibitor. Clotting times and coagulation factor assays were not different between COVID-19 convalescent plasma and fresh frozen plasma, except for protein C antigen. When compared to fresh frozen plasma and regular convalescent plasma, pathogen reduction treatment increased activated partial thromboplastin time and thrombin time, while reducing fibrinogen, coagulation factor II, V, VIII, IX, X, XI, XII, protein S activity, and alpha-2 plasmin inhibitor. The coagulation profiles of human COVID-19 convalescent plasma and standard fresh frozen plasma are not different. Pathogen reduced COVID-19 convalescent plasma is associated with reduction of coagulation factors and a slight prolongation of coagulation times, as anticipated. A key limitation of the study is that the COVID-19 disease course of the convalesced donors was not characterized.
Collapse
Affiliation(s)
- Allan M Klompas
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Noud van Helmond
- Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Health Care, Camden, NJ, USA
| | - Justin E Juskewitch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Rajiv K Pruthi
- Division of Hematology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Matthew A Sexton
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Juan C Diaz Soto
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Stephen A Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Katherine A Senese
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | | | - Jeffrey L Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - James R Stubbs
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Scott A Hammel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
203
|
Focosi D, Franchini M, Joyner MJ, Casadevall A. Are convalescent plasma stocks collected during former COVID-19 waves still effective against current SARS-CoV-2 variants? Vox Sang 2022; 117:641-646. [PMID: 35023163 DOI: 10.1111/vox.13239] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 12/06/2021] [Accepted: 12/06/2021] [Indexed: 12/01/2022]
Abstract
COVID-19 convalescent plasma (CCP) was among the few frontline therapies used to treat COVID-19. After large randomized controlled trials (RCTs) relying on late use in hospitalized patients and/or low antibody titres failed to meet their predefined primary endpoint, the infectious disease community reduced usage of CCP in favour of monoclonal antibodies. Consequently, there are CCP stocks at most transfusion centres worldwide, although scattered usage continues. Further, better designed RCTs are also being launched. The urgent question here is: should we use CCP units collected months before given the largely changed viral variant landscape? We review here in vitro evidence that discourages usage of such CCP units against Delta and other variants of concern. CCP collections should be continued in order to update the armamentarium of therapeutics against vaccine breakthrough infections or in unvaccinated patients and is especially relevant in next-generation RCTs.
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
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
204
|
Concerns about estimating relative risk of death associated with convalescent plasma for COVID-19. Nat Med 2022; 28:51-52. [PMID: 35013614 DOI: 10.1038/s41591-021-01638-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/22/2021] [Indexed: 11/09/2022]
|
205
|
Park H, Tarpey T, Liu M, Goldfeld K, Wu Y, Wu D, Li Y, Zhang J, Ganguly D, Ray Y, Paul SR, Bhattacharya P, Belov A, Huang Y, Villa C, Forshee R, Verdun NC, Yoon HA, Agarwal A, Simonovich VA, Scibona P, Burgos Pratx L, Belloso W, Avendaño-Solá C, Bar KJ, Duarte RF, Hsue PY, Luetkemeyer AF, Meyfroidt G, Nicola AM, Mukherjee A, Ortigoza MB, Pirofski LA, Rijnders BJA, Troxel A, Antman EM, Petkova E. Development and Validation of a Treatment Benefit Index to Identify Hospitalized Patients With COVID-19 Who May Benefit From Convalescent Plasma. JAMA Netw Open 2022; 5:e2147375. [PMID: 35076698 PMCID: PMC8790670 DOI: 10.1001/jamanetworkopen.2021.47375] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/15/2021] [Indexed: 12/20/2022] Open
Abstract
Importance Identifying which patients with COVID-19 are likely to benefit from COVID-19 convalescent plasma (CCP) treatment may have a large public health impact. Objective To develop an index for predicting the expected relative treatment benefit from CCP compared with treatment without CCP for patients hospitalized for COVID-19 using patients' baseline characteristics. Design, Setting, and Participants This prognostic study used data from the COMPILE study, ie, a meta-analysis of pooled individual patient data from 8 randomized clinical trials (RCTs) evaluating CCP vs control in adults hospitalized for COVID-19 who were not receiving mechanical ventilation at randomization. A combination of baseline characteristics, termed the treatment benefit index (TBI), was developed based on 2287 patients in COMPILE using a proportional odds model, with baseline characteristics selected via cross-validation. The TBI was externally validated on 4 external data sets: the Expanded Access Program (1896 participants), a study conducted under Emergency Use Authorization (210 participants), and 2 RCTs (with 80 and 309 participants). Exposure Receipt of CCP. Main Outcomes and Measures World Health Organization (WHO) 11-point ordinal COVID-19 clinical status scale and 2 derivatives of it (ie, WHO score of 7-10, indicating mechanical ventilation to death, and WHO score of 10, indicating death) at day 14 and day 28 after randomization. Day 14 WHO 11-point ordinal scale was used as the primary outcome to develop the TBI. Results A total of 2287 patients were included in the derivation cohort, with a mean (SD) age of 60.3 (15.2) years and 815 (35.6%) women. The TBI provided a continuous gradation of benefit, and, for clinical utility, it was operationalized into groups of expected large clinical benefit (B1; 629 participants in the derivation cohort [27.5%]), moderate benefit (B2; 953 [41.7%]), and potential harm or no benefit (B3; 705 [30.8%]). Patients with preexisting conditions (diabetes, cardiovascular and pulmonary diseases), with blood type A or AB, and at an early COVID-19 stage (low baseline WHO scores) were expected to benefit most, while those without preexisting conditions and at more advanced stages of COVID-19 could potentially be harmed. In the derivation cohort, odds ratios for worse outcome, where smaller odds ratios indicate larger benefit from CCP, were 0.69 (95% credible interval [CrI], 0.48-1.06) for B1, 0.82 (95% CrI, 0.61-1.11) for B2, and 1.58 (95% CrI, 1.14-2.17) for B3. Testing on 4 external datasets supported the validation of the derived TBIs. Conclusions and Relevance The findings of this study suggest that the CCP TBI is a simple tool that can quantify the relative benefit from CCP treatment for an individual patient hospitalized with COVID-19 that can be used to guide treatment recommendations. The TBI precision medicine approach could be especially helpful in a pandemic.
Collapse
Affiliation(s)
- Hyung Park
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Thaddeus Tarpey
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Mengling Liu
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
- Department of Environmental Medicine, New York University Grossman School of Medicine, New York
| | - Keith Goldfeld
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Yinxiang Wu
- Department of Biostatistics, School of Public Health, University of Washington, Seattle
| | - Danni Wu
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Yi Li
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Jinchun Zhang
- Biostatistics and Research Decision Sciences, Merck Research Labortory, Merck & Co Inc, Rahway, New Jersey
| | - Dipyaman Ganguly
- Translational Research Unit of Excellence, Council Of Scientific And Industrial Research–Indian Institute of Chemical Biology, Kolkata, India
| | - Yogiraj Ray
- Infectious Disease, Beleghata General Hospital, Kolkata, India
- School of Tropical Medicine, Kolkata, India
| | | | | | - Artur Belov
- Center for Biologics Evaluation and Research, Office of Biostatistics and Epidemiology, Analytics and Benefit-Risk Assessment Team, US Food and Drug Administration, Silver Spring, Maryland
| | - Yin Huang
- Center for Biologics Evaluation and Research, Office of Biostatistics and Epidemiology, Analytics and Benefit-Risk Assessment Team, US Food and Drug Administration, Silver Spring, Maryland
| | - Carlos Villa
- Center for Biologics Evaluation and Research, Office of Biostatistics and Epidemiology, Analytics and Benefit-Risk Assessment Team, US Food and Drug Administration, Silver Spring, Maryland
| | - Richard Forshee
- Center for Biologics Evaluation and Research, Office of Biostatistics and Epidemiology, Analytics and Benefit-Risk Assessment Team, US Food and Drug Administration, Silver Spring, Maryland
| | - Nicole C. Verdun
- Office of Blood Research and Review, Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland
| | - Hyun ah Yoon
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
| | - Anup Agarwal
- Indian Council of Medical Research, New Delhi, India
| | - Ventura Alejandro Simonovich
- Clinical Pharmacology Section, Department of Internal Medicine and Department of Research, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Paula Scibona
- Clinical Pharmacology Section, Internal Medicine Service, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Leandro Burgos Pratx
- Transfusional Medicine Service, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Waldo Belloso
- Department of Research, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | | | - Katharine J Bar
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Rafael F. Duarte
- Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Priscilla Y. Hsue
- Zuckerberg San Francisco General, University of California, San Francisco
| | | | - Geert Meyfroidt
- Department of Intensive Care Medicine, University Hospitals Leuven, Leuven, Belgium
| | - André M. Nicola
- Hospital Universitário de Brasília, University of Brasília, Brasília, Brazil
| | | | - Mila B. Ortigoza
- Departments of Medicine and Microbiology, New York University Grossman School of Medicine, New York
| | - Liise-anne Pirofski
- Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York
| | - Bart J. A. Rijnders
- Department of Internal Medicine, Section of Infectious Diseases, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andrea Troxel
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
| | - Elliott M. Antman
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eva Petkova
- Division of Biostatistics, Department of Population Health, New York University Grossman School of Medicine, New York
- Department of Child and Adolescent Psychiatry, New York University Grossman School of Medicine
- Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
| |
Collapse
|
206
|
Troxel AB, Petkova E, Goldfeld K, Liu M, Tarpey T, Wu Y, Wu D, Agarwal A, Avendaño-Solá C, Bainbridge E, Bar KJ, Devos T, Duarte RF, Gharbharan A, Hsue PY, Kumar G, Luetkemeyer AF, Meyfroidt G, Nicola AM, Mukherjee A, Ortigoza MB, Pirofski LA, Rijnders BJA, Rokx C, Sancho-Lopez A, Shaw P, Tebas P, Yoon HA, Grudzen C, Hochman J, Antman EM. Association of Convalescent Plasma Treatment With Clinical Status in Patients Hospitalized With COVID-19: A Meta-analysis. JAMA Netw Open 2022; 5:e2147331. [PMID: 35076699 PMCID: PMC8790669 DOI: 10.1001/jamanetworkopen.2021.47331] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 12/15/2021] [Indexed: 12/15/2022] Open
Abstract
Importance COVID-19 convalescent plasma (CCP) is a potentially beneficial treatment for COVID-19 that requires rigorous testing. Objective To compile individual patient data from randomized clinical trials of CCP and to monitor the data until completion or until accumulated evidence enables reliable conclusions regarding the clinical outcomes associated with CCP. Data Sources From May to August 2020, a systematic search was performed for trials of CCP in the literature, clinical trial registry sites, and medRxiv. Domain experts at local, national, and international organizations were consulted regularly. Study Selection Eligible trials enrolled hospitalized patients with confirmed COVID-19, not receiving mechanical ventilation, and randomized them to CCP or control. The administered CCP was required to have measurable antibodies assessed locally. Data Extraction and Synthesis A minimal data set was submitted regularly via a secure portal, analyzed using a prespecified bayesian statistical plan, and reviewed frequently by a collective data and safety monitoring board. Main Outcomes and Measures Prespecified coprimary end points-the World Health Organization (WHO) 11-point ordinal scale analyzed using a proportional odds model and a binary indicator of WHO score of 7 or higher capturing the most severe outcomes including mechanical ventilation through death and analyzed using a logistic model-were assessed clinically at 14 days after randomization. Results Eight international trials collectively enrolled 2369 participants (1138 randomized to control and 1231 randomized to CCP). A total of 2341 participants (median [IQR] age, 60 [50-72] years; 845 women [35.7%]) had primary outcome data as of April 2021. The median (IQR) of the ordinal WHO scale was 3 (3-6); the cumulative OR was 0.94 (95% credible interval [CrI], 0.74-1.19; posterior probability of OR <1 of 71%). A total of 352 patients (15%) had WHO score greater than or equal to 7; the OR was 0.94 (95% CrI, 0.69-1.30; posterior probability of OR <1 of 65%). Adjusted for baseline covariates, the ORs for mortality were 0.88 at day 14 (95% CrI, 0.61-1.26; posterior probability of OR <1 of 77%) and 0.85 at day 28 (95% CrI, 0.62-1.18; posterior probability of OR <1 of 84%). Heterogeneity of treatment effect sizes was observed across an array of baseline characteristics. Conclusions and Relevance This meta-analysis found no association of CCP with better clinical outcomes for the typical patient. These findings suggest that real-time individual patient data pooling and meta-analysis during a pandemic are feasible, offering a model for future research and providing a rich data resource.
Collapse
Affiliation(s)
- Andrea B. Troxel
- Department of Population 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, New York
- The Nathan S. Kline Institute for Psychiatric Research, Orangeburg, New York
| | - Keith Goldfeld
- Department of Population Health, NYU Grossman School of Medicine, New York, 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
| | - Thaddeus Tarpey
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Yinxiang Wu
- Department of Biostatistics, University of Washington School of Public Health, Seattle
| | - Danni Wu
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
| | - Anup Agarwal
- Indian Council of Medical Research, New Delhi, Delhi, India
| | | | - Emma Bainbridge
- Zuckerberg San Francisco General, University of California San Francisco, San Francisco
| | - Katherine J. Bar
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Timothy Devos
- Department of Hematology, University Hospitals Leuven and Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium
| | - Rafael F. Duarte
- Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Arvind Gharbharan
- Section of Infectious Diseases, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Priscilla Y. Hsue
- Zuckerberg San Francisco General, University of California San Francisco, San Francisco
| | - Gunjan Kumar
- Indian Council of Medical Research, New Delhi, Delhi, India
| | - Annie F. Luetkemeyer
- Zuckerberg San Francisco General, University of California San Francisco, San Francisco
| | - Geert Meyfroidt
- Department of Intensive Care Medicine, University Hospitals Leuven, Leuven, Belgium
| | - André M. Nicola
- Hospital Universitário de Brasília, University of Brasília, Brasília, Brazil
| | | | - Mila B. Ortigoza
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
- Department of Microbiology, NYU Grossman School of Medicine, New York, New York
| | - Liise-anne Pirofski
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Bart J. A. Rijnders
- Section of Infectious Diseases, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Casper Rokx
- Section of Infectious Diseases, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Pamela Shaw
- Biostatistics Unit, Kaiser Permanente Washington Health Research Institute, Seattle
| | - Pablo Tebas
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Hyun-Ah Yoon
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | - Corita Grudzen
- Department of Population Health, NYU Grossman School of Medicine, New York, New York
- Department of Emergency Medicine, NYU Grossman School of Medicine, New York, New York
| | - Judith Hochman
- Department of Medicine, NYU Grossman School of Medicine, New York, New York
| | - Elliott M. Antman
- Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
207
|
Raman N, Kishore K, Rana S, Vardhan V, Thareja S, Padmaprakash K, Muthukrishnan J, Rajmohan K, Agarwal M, Ashta K, Anilkumar A. Propensity score-matched case–control study of convalescent plasma in treatment of patients with moderate and severe COVID-19. THE JOURNAL OF ASSOCIATION OF CHEST PHYSICIANS 2022. [DOI: 10.4103/jacp.jacp_44_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
208
|
Volk A, Covini-Souris C, Kuehnel D, De Mey C, Römisch J, Schmidt T. SARS-CoV-2 Neutralization in Convalescent Plasma and Commercial Lots of Plasma-Derived Immunoglobulin. BioDrugs 2022; 36:41-53. [PMID: 34843105 PMCID: PMC8628143 DOI: 10.1007/s40259-021-00511-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Patients with primary or secondary immunodeficiency (PID or SID) face increased insecurity and discomfort in the light of the COVID-19 pandemic, not knowing if and to what extent their comorbidities may impact the course of a potential SARS-CoV-2 infection. Furthermore, recently available vaccination options might not be amenable or effective for all patients in this heterogeneous population. Therefore, these patients often rely on passive immunization with plasma-derived, intravenous or subcutaneous immunoglobulin (IVIG/SCIG). Whether the ongoing COVID-19 pandemic and/or the progress in vaccination programs lead to increased and potentially protective titers in plasma-derived immunoglobulins (Ig) indicated (e.g., for humoral immunodeficiency) remains a pressing question for this patient population. PURPOSE We investigated SARS-CoV-2 reactivity of US plasma-derived IVIG/SCIG products from the end of 2020 until June 2021 as well as in convalescent plasma (CP) from May 2020 to August 2020 to determine whether potentially neutralizing antibody titers may be present. METHODS Final containers of IVIG/SCIG and CP donations were analyzed by commercial ELISA for anti-SARS-CoV-2 S1-receptor binding domain (RBD) IgG as well as microneutralization assay using a patient-derived SARS-CoV-2 (D614G) isolate. Neutralization capacities of 313 single plasma donations and 119 plasma-derived IVIG/SCIG lots were determined. Results obtained from both analytical methods were normalized against the WHO International Standard. Finally, based on dense pharmacokinetic profiles of an IVIG preparation from previously published investigations, possible steady-state plasma levels of SARS-CoV-2 neutralization capacities were approximated based on currently measured anti-SARS-CoV-2 potencies in IVIG/SCIG preparations. RESULTS CP donations presented with high variability with regards to anti-SARS-CoV-2 reactivity in ELISA as well as in neutralization testing. While approximately 50% of convalescent donations were not/low neutralizing, approximately 10% were at or above 600 IU/mL. IVIG/SCIG lots derived from pre-pandemic plasma donations did not show neutralizing capacities for SARS-CoV-2. Lots produced between December 2020 and June 2021 entailing plasma donations after the emergence of SARS-CoV-2 showed a rapid and constant increase in anti-SARS-CoV-2 reactivity and neutralization capacity over time. While lot-to-lot variability was substantial, neutralization capacity increased from a mean of 21 IU/mL in December 2020 to 506 IU/mL in June 2021 with a maximum of 864 IU/mL for the most recent lots. Pharmacokinetic extrapolations, based on non-compartmental superposition principles using steady-state reference profiles from previously published pharmacokinetic investigations on IVIG in PID, yielded potential steady-state trough plasma levels of 16 IU/mL of neutralizing SARS-CoV-2 IgG based on the average final container concentration from May 2021 of 216 IU/mL. Maximum extrapolated trough levels could reach 64 IU/mL based on the latest maximal final container potency tested in June 2021. CONCLUSIONS SARS-CoV-2 reactivity and neutralization capacity in IVIG/SCIG produced from US plasma rapidly and in part exponentially increased in the first half of 2021. The observed increase of final container potencies is likely trailing the serological status of the US donor population in terms of COVID-19 convalescence and vaccination by at least 5 months due to production lead times and should in principle continue at least until Fall 2021. In summary, the data support rapidly increasing levels of anti-SARS-CoV-2 antibodies in IVIG/SCIG products, implicating that a certain level of protection could be possible against COVID-19 for regularly substituted PID/SID patients. Nevertheless, more research is still needed to confirm which plasma levels are needed to provide protection against SARS-CoV-2 infection in immune-compromised patients.
Collapse
Affiliation(s)
- Andreas Volk
- Virus and Prion Validation, Octapharma Biopharmaceuticals GmbH, Frankfurt, Germany.
| | | | - Denis Kuehnel
- Virus and Prion Validation, Octapharma Biopharmaceuticals GmbH, Frankfurt, Germany
| | | | - Jürgen Römisch
- R&D Plasma, Octapharma Pharmazeutika Produktionsgesellschaft m.b.H., Vienna, Austria
| | - Torben Schmidt
- Virus and Prion Validation, Octapharma Biopharmaceuticals GmbH, Frankfurt, Germany
| |
Collapse
|
209
|
Dhawan M, Priyanka, Parmar M, Angural S, Choudhary OP. Convalescent plasma therapy against the emerging SARS-CoV-2 variants: Delineation of the potentialities and risks. Int J Surg 2022; 97:106204. [PMID: 34974199 PMCID: PMC8717699 DOI: 10.1016/j.ijsu.2021.106204] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has resulted in a catastrophic pandemic and severely impacted people's livelihoods worldwide. In addition, the emergence of SARS-CoV-2 variants has posed a severe threat to humankind. Due to the dearth of therapeutic options during the commencement of the pandemic, convalescent plasma therapy (CPT) played a significant part in the management of patients with severe form of COVID-19. Several recent studies have proposed various protective effects of CPT, such as antiviral, anti-inflammatory, anti-thrombotic, and immunomodulatory actions, curtailing the devastating consequences of the SARS-CoV-2 infection. On the contrary, several clinical studies have raised some serious concerns about the effectiveness and reliability of CPT in the management of patients with COVID-19. The protective effects of CPT in severely ill patients are yet to be proved. Moreover, the emergence of SARS-CoV-2 variants has raised concerns about the effectiveness of CPT against COVID-19. Therefore, to establish concrete evidence of the efficacy of CPT and adjudicate its inclusion in the management of COVID-19, an updated review of present literature is required, which could help in the development of an efficient therapeutic regimen to treat COVID-19 amid the emergence of new viral variants.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, 141004, Punjab, India,The Trafford Group of Colleges, Manchester, WA14 5PQ, UK
| | - Priyanka
- Independent Researcher, 07, Type IV Quarter, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, 796015, Mizoram, India
| | - Manisha Parmar
- Department of Veterinary Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, 141004, Punjab, India
| | - Steffy Angural
- Department of Medical Lab Technology, Faculty of Applied Health Sciences, GNA University, Phagwara-Hoshiarpur Road, Sri Hargobindgarh, 144401, Punjab, India,Corresponding author
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, 796015, Mizoram, India,Corresponding author
| |
Collapse
|
210
|
Sait AS, Chiang TPY, Marr KA, Massie AB, Cochran W, Shah P, Brennan DC, Thomas AG, Mehta Steinke S, Permpalung N, Shoham S, Merlo C, Jain T, Boyarsky B, Charnaya O, Gurakar A, Sharma K, Durand CM, Werbel WA, Huang CY, Ostrander D, Desai N, Kim MY, Alasfar S, Bloch EM, Tobian AA, Garonzik-Wang J, Segev DL, Avery RK. Outcomes of SOT Recipients With COVID-19 in Different Eras of COVID-19 Therapeutics. Transplant Direct 2022; 8:e1268. [PMID: 34966840 PMCID: PMC8710330 DOI: 10.1097/txd.0000000000001268] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 10/05/2021] [Accepted: 10/19/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Few reports have focused on newer coronavirus disease 2019 (COVID-19) therapies (remdesivir, dexamethasone, and convalescent plasma) in solid organ transplant recipients; concerns had been raised regarding possible adverse impact on allograft function or secondary infections. METHODS We studied 77 solid organ transplant inpatients with COVID-19 during 2 therapeutic eras (Era 1: March-May 2020, 21 patients; and Era 2: June-November 2020, 56 patients) and 52 solid organ transplant outpatients. RESULTS In Era 1, no patients received remdesivir or dexamethasone, and 4 of 21 (19.4%) received convalescent plasma, whereas in Era 2, remdesivir (24/56, 42.9%), dexamethasone (24/56, 42.9%), and convalescent plasma (40/56, 71.4%) were commonly used. Mortality was low across both eras, 4 of 77 (5.6%), and rejection occurred in only 2 of 77 (2.8%) inpatients; infections were similar in hypoxemic patients with or without dexamethasone. Preexisting graft dysfunction was associated with greater need for hospitalization, higher severity score, and lower survival. Acute kidney injury was present in 37.3% of inpatients; renal function improved more rapidly in patients who received remdesivir and convalescent plasma. Post-COVID-19 renal and liver function were comparable between eras, out to 90 d. CONCLUSIONS Newer COVID-19 therapies did not appear to have a deleterious effect on allograft function, and infectious complications were comparable.
Collapse
Affiliation(s)
- Afrah S. Sait
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Teresa Po-Yu Chiang
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kieren A. Marr
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allan B. Massie
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD
| | - Willa Cochran
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Pali Shah
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Daniel C. Brennan
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Comprehensive Transplant Center, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alvin G. Thomas
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Seema Mehta Steinke
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nitipong Permpalung
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christian Merlo
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Tania Jain
- Hematologic Malignancies and Bone Marrow Transplantation Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD
| | - Brian Boyarsky
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Olga Charnaya
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ahmet Gurakar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kavita Sharma
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christine M. Durand
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - William A. Werbel
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chiung-Yu Huang
- Department of Statistics, University of California at San Francisco, San Francisco, CA
| | - Darin Ostrander
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Niraj Desai
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Min Young Kim
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Sami Alasfar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Evan M. Bloch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Dorry L. Segev
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Epidemiology, Johns Hopkins University School of Public Health, Baltimore, MD
| | - Robin K. Avery
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
211
|
Tang J, Grubbs G, Lee Y, Wu H, Luke TC, Egland KA, Bausch CL, Sullivan EJ, Khurana S. OUP accepted manuscript. J Infect Dis 2022; 226:655-663. [PMID: 35106573 PMCID: PMC8903330 DOI: 10.1093/infdis/jiac031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 12/01/2022] Open
Abstract
Passive antibody immunotherapeutics directed against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are promising countermeasures for protection and treatment of coronavirus disease 2019 (COVID-19). SARS-CoV-2 variants of concern (VOCs) and variants of interest (VOIs) can impact the clinical efficacy of immunotherapeutics. A fully human polyclonal antibody immunotherapeutic purified from plasma of transchromosomic (Tc) bovines hyperimmunized with SARS-CoV-2 WA-1 spike (SAB-185) is being assessed for efficacy in a phase 2/3 clinical trial when different circulating SARS-CoV-2 variants predominated. We evaluated antibody binding, avidity maturation, and SARS-CoV-2 VOCs/VOIs virus-neutralizing capacity of convalescent plasma compared with different lots of SAB-185 and individual Tc bovine sera sequentially obtained after each vaccination against Alpha, Epsilon, Iota, Gamma, Beta, Kappa, and Delta variants. In contrast to convalescent plasma, sera and SAB-185 derived from hyperimmunized Tc bovines demonstrated higher antibody avidity and more potent cross-neutralizing activity of VOCs/VOIs. Thus, SAB-185 is a potential promising therapeutic candidate for the treatment of patients infected with SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Juanjie Tang
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Gabrielle Grubbs
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Youri Lee
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
| | - Hua Wu
- SAB Biotherapeutics, Sioux Falls, South Dakota, USA
| | | | | | | | | | - Surender Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research, United States Food and Drug Administration, Silver Spring, Maryland, USA
- Correspondence: Surender Khurana, PhD, Division of Viral Products, Center for Biologics Evaluation and Research, US Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA ()
| |
Collapse
|
212
|
Menichetti F, Falcone M, Tiseo G. Management of COVID patients with convalescent plasma: Do we have the final word? Eur J Intern Med 2022; 95:13-16. [PMID: 34895816 PMCID: PMC8630688 DOI: 10.1016/j.ejim.2021.10.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/24/2021] [Indexed: 11/24/2022]
Abstract
Immunotherapy with convalescent plasma (CP) has been used in the past in several different infectious diseases and proposed as a potential therapeutic option in patients with COVID-19. However, a clear benefit was never demonstrated and randomized clinical trials (RCTs) conducted in different populations of COVID-19 patients showed contrasting results. In general, current evidences suggest that CP in patients with moderate to severe COVID-19 does not reduce the progression to severe respiratory failure or death within 30 days. However, currently published RCTs have several limitations. The administration of plasma with low titer of neutralizing antibodies (NAbs), the use of suboptimal surrogate serological tests to determine NAbs titer, the delayed administration of CP from the onset of COVID-19 symptoms and the lack of information about antibody titer of recipients before CP infusion, are all limiting factors that may have affected the study results. Thus, a potential benefit of early (within the first 72 h from onset of symptoms), high titer CP in patients with mild COVID-19 (pO2/FiO2>300) cannot be definitively excluded. However, immunotherapy with monoclonal antibodies developed from CP demonstrated efficacy in reducing progression to severe COVID-19 and hospitalization and are today recommended in the early phase of COVID-19.
Collapse
Affiliation(s)
- Francesco Menichetti
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Marco Falcone
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giusy Tiseo
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
213
|
Abstract
The unprecedented public health and economic impact of the COVID-19 pandemic caused by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been met with an equally unprecedented scientific response. Much of this response has focused, appropriately, on the mechanisms of SARS-CoV-2 entry into host cells, and in particular the binding of the spike (S) protein to its receptor, angiotensin-converting enzyme 2 (ACE2), and subsequent membrane fusion. This Review provides the structural and cellular foundations for understanding the multistep SARS-CoV-2 entry process, including S protein synthesis, S protein structure, conformational transitions necessary for association of the S protein with ACE2, engagement of the receptor-binding domain of the S protein with ACE2, proteolytic activation of the S protein, endocytosis and membrane fusion. We define the roles of furin-like proteases, transmembrane protease, serine 2 (TMPRSS2) and cathepsin L in these processes, and delineate the features of ACE2 orthologues in reservoir animal species and S protein adaptations that facilitate efficient human transmission. We also examine the utility of vaccines, antibodies and other potential therapeutics targeting SARS-CoV-2 entry mechanisms. Finally, we present key outstanding questions associated with this critical process.
Collapse
Affiliation(s)
- Cody B Jackson
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Michael Farzan
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA
| | - Bing Chen
- Division of Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Hyeryun Choe
- Department of Immunology and Microbiology, Scripps Research, Jupiter, FL, USA.
| |
Collapse
|
214
|
Boggiano C, Eisinger RW, Lerner AM, Anderson JM, Woodcock J, Fauci AS, Collins FS. Update on and Future Directions for Use of Anti-SARS-CoV-2 Antibodies: National Institutes of Health Summit on Treatment and Prevention of COVID-19. Ann Intern Med 2022; 175:119-126. [PMID: 34724404 PMCID: PMC8559823 DOI: 10.7326/m21-3669] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
As the fourth wave of the SARS-CoV-2 pandemic encircles the globe, there remains an urgent challenge to identify safe and effective treatment and prevention strategies that can be implemented in a range of health care and clinical settings. Substantial advances have been made in the use of anti-SARS-CoV-2 antibodies to mitigate the morbidity and mortality associated with COVID-19. On 15 June 2021, the National Institutes of Health, in collaboration with the U.S. Food and Drug Administration, convened a virtual summit to summarize existing knowledge on anti-SARS-CoV-2 antibodies and to identify key unanswered scientific questions to further catalyze the clinical development and implementation of antibodies.
Collapse
Affiliation(s)
- César Boggiano
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (C.B.)
| | - Robert W Eisinger
- Office of the Director, National Institutes of Health, Bethesda, Maryland (R.W.E., J.M.A., F.S.C.)
| | - Andrea M Lerner
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (A.M.L., A.S.F.)
| | - James M Anderson
- Office of the Director, National Institutes of Health, Bethesda, Maryland (R.W.E., J.M.A., F.S.C.)
| | - Janet Woodcock
- Office of the Commissioner, Food and Drug Administration, Silver Spring, Maryland (J.W.)
| | - Anthony S Fauci
- Office of the Director, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland (A.M.L., A.S.F.)
| | - Francis S Collins
- Office of the Director, National Institutes of Health, Bethesda, Maryland (R.W.E., J.M.A., F.S.C.)
| |
Collapse
|
215
|
Effectiveness and Controversy of Convalescent Plasma Therapy for Coronavirus Disease 2019 Patients. INFECTIOUS DISEASES & IMMUNITY 2022. [PMID: 37521156 PMCID: PMC8772051 DOI: 10.1097/id9.0000000000000033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Since the coronavirus disease 2019 (COVID-19) began to spread, it remains pandemic worldwide. The European Medicines Agency's human medicines committee and Food and Drug Administration have only granted a conditional marketing authorization for remdesivir to treat COVID-19. It is essential to apply other valuable treatments. Convalescent plasma (CP), donated by persons who have recovered from COVID-19, is the cellular component of blood that contains specific antibodies. Therefore, to determine the feasibility of CP for COVID-19, the effectiveness and controversy are discussed in depth here. It is suggested that CP plays a certain role in the treatment of COVID-19. As a treatment, it may have its own indications and contraindications, which need to be further discussed. Meanwhile, it is critical to establish a standard procedure for treatment from CP collection, preservation, transport, to transfusion, and conduct some large sample randomized controlled trials to confirm the transfusion dosage, appropriate time, frequency, and actively prevent adverse outcomes that may occur.
Collapse
|
216
|
Ling RR, Sim JJL, Tan FL, Tai BC, Syn N, Mucheli SS, Fan BE, Mitra S, Ramanathan K. Convalescent Plasma for Patients Hospitalized With Coronavirus Disease 2019: A Meta-Analysis With Trial Sequential Analysis of Randomized Controlled Trials. Transfus Med Rev 2022; 36:16-26. [PMID: 34782209 PMCID: PMC8502250 DOI: 10.1016/j.tmrv.2021.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/24/2021] [Accepted: 09/29/2021] [Indexed: 02/06/2023]
Abstract
Current evidence from randomized controlled trials (RCTs) and systematic reviews on the utility of convalescent plasma (CP) in patients with coronavirus disease 2019 (COVID-19) suggests a lack of benefit. We conducted an updated meta-analysis of RCTs with trial sequential analysis to investigate whether convalescent plasma is futile in reducing mortality in patients hospitalized with COVID-19. We searched 6 databases from December 1, 2019 to August 1, 2021 for RCTs comparing the use of CP with standard of care or transfusion of non-CP standard plasma in patients with COVID-19. The risk of bias was assessed using the Cochrane Risk-of-Bias 2 Tool. Random effects (DerSimonian and Laird) meta-analyses were conducted. The primary outcome was the aggregate risk for in-hospital mortality between both arms. We conducted a trial sequential analysis (TSA) based on the pooled relative risks (RRs) for in-hospital mortality. Secondary outcomes included the pooled RR for receipt of mechanical ventilation and mean difference in hospital length of stay. We included 18 RCTs (8702 CP, 7906 control). CP was not associated with a significant mortality benefit (RR: 0.95, 95%-CI: 0.86-1.04, P = .27, high certainty). Subgroup analysis did not find any significant differences (pinteraction = 0.30) between patients who received CP within 8 days of symptom onset (RR: 0.97, 95%-CI: 0.79-1.19, P = .80), or after 8 days (RR: 0.79, 95%-CI: 0.57-1.10, P = .16). TSA based on a RR reduction of 10% from a baseline mortality of 20% found that CP was not effective, with the pooled effect within the boundary for futility. CP did not significantly reduce the requirement for mechanical ventilation (RR: 1.00, 95%-CI: 0.91-1.10, P = .99, moderate certainty) or hospital length of stay (+1.32, 95%-CI: -1.86 to +4.52, P = .42, low certainty). CP does not improve relevant clinical outcomes in patients with COVID-19, especially in severe disease. The pooled effect of mortality was within the boundary of futility, suggesting the lack of benefit of CP in patients hospitalized with COVID-19.
Collapse
Affiliation(s)
- Ryan Ruiyang Ling
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Jackie Jia Lin Sim
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Felicia Liying Tan
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Bee Choo Tai
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore; Saw Swee Hock School of Public Health, National University of Singapore, National University Health System, Singapore
| | - Nicholas Syn
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore
| | - Sharavan Sadasiv Mucheli
- Department of Infectious Diseases, Tan Tock Seng Hospital, National Centre for Infectious Diseases, Singapore
| | | | - Saikat Mitra
- Lyell McEwin Hospital, Adelaide, South Australia, Australia
| | - Kollengode Ramanathan
- Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore; Cardiothoracic Intensive Care Unit, National University Heart Centre, National University Hospital, Singapore.
| |
Collapse
|
217
|
Epidemiological Characteristics of Hospitalized Patients with Moderate versus Severe COVID-19 Infection: A Retrospective Cohort Single Centre Study. Diseases 2021; 10:diseases10010001. [PMID: 35076497 PMCID: PMC8788538 DOI: 10.3390/diseases10010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/11/2021] [Accepted: 12/17/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 has a devastating impact worldwide. Recognizing factors that cause its progression is important for the utilization of appropriate resources and improving clinical outcomes. In this study, we aimed to identify the epidemiological and clinical characteristics of patients who were hospitalized with moderate versus severe COVID-19 illness. A single-center, retrospective cohort study was conducted between 3 March and 9 September 2020. Following the CDC guidelines, a two-category variable for COVID-19 severity (moderate versus severe) based on length of stay, need for intensive care or mechanical ventilation and mortality was developed. Data including demographic, clinical characteristics, laboratory parameters, therapeutic interventions and clinical outcomes were assessed using descriptive and inferential analysis. A total of 1002 patients were included, the majority were male (n = 646, 64.5%), Omani citizen (n = 770, 76.8%) and with an average age of 54.2 years. At the bivariate level, patients classified as severe were older (Mean = 55.2, SD = 16) than the moderate patients (Mean = 51.5, SD = 15.8). Diabetes mellitus was the only significant comorbidity potential factor that was more prevalent in severe patients than moderate (n = 321, 46.6%; versus n = 178, 42.4%; p < 0.001). Under the laboratory factors; total white cell count (WBC), C-reactive protein (CRP), Lactate dehydrogenase (LDH), D-dimer and corrected calcium were significant. All selected clinical characteristics and therapeutics were significant. At the multivariate level, under demographic factors, only nationality was significant and no significant comorbidity was identified. Three clinical factors were identified, including; sepsis, Acute respiratory disease syndrome (ARDS) and requirement of non-invasive ventilation (NIV). CRP and steroids were also identified under laboratory and therapeutic factors, respectively. Overall, our study identified only five factors from a total of eighteen proposed due to their significant values (p < 0.05) from the bivariate analysis. There are noticeable differences in levels of COVID-19 severity among nationalities. All the selected clinical and therapeutic factors were significant, implying that they should be a key priority when assessing severity in hospitalized COVID-19 patients. An elevated level of CRP may be a valuable early marker in predicting the progression in non-severe patients with COVID-19. Early recognition and intervention of these factors could ease the management of hospitalized COVID-19 patients and reduce case fatalities as well medical expenditure.
Collapse
|
218
|
Deere JD, Carroll TD, Dutra J, Fritts L, Sammak RL, Yee JL, Olstad KJ, Reader JR, Kistler A, Kamm J, Di Germanio C, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Simmons G, Watanabe J, Pollard RE, Usachenko J, Immareddy R, Schmidt BA, O’Connor SL, DeRisi J, Busch MP, Iyer SS, Van Rompay KKA, Hartigan-O’Connor DJ, Miller CJ. SARS-CoV-2 Infection of Rhesus Macaques Treated Early with Human COVID-19 Convalescent Plasma. Microbiol Spectr 2021; 9:e0139721. [PMID: 34817208 PMCID: PMC8612156 DOI: 10.1128/spectrum.01397-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
Human clinical studies investigating use of convalescent plasma (CP) for treatment of coronavirus disease 2019 (COVID-19) have produced conflicting results. Outcomes in these studies may vary at least partly due to different timing of CP administration relative to symptom onset. The mechanisms of action of CP include neutralizing antibodies but may extend beyond virus neutralization to include normalization of blood clotting and dampening of inflammation. Unresolved questions include the minimum therapeutic titer in the CP units or CP recipient as well as the optimal timing of administration. Here, we show that treatment of macaques with CP within 24 h of infection does not reduce viral shedding in nasal or lung secretions compared to controls and does not detectably improve any clinical endpoint. We also demonstrate that CP administration does not impact viral sequence diversity in vivo, although the selection of a viral sequence variant in both macaques receiving normal human plasma was suggestive of immune pressure. Our results suggest that CP, administered to medium titers, has limited efficacy, even when given very early after infection. Our findings also contribute information important for the continued development of the nonhuman primate model of COVID-19. These results should inform interpretation of clinical studies of CP in addition to providing insights useful for developing other passive immunotherapies and vaccine strategies. IMPORTANCE Antiviral treatment options for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) remain very limited. One treatment that was explored beginning early in the pandemic (and that is likely to be tested early in future pandemics) is plasma collected from people who have recovered from coronavirus disease 2019 (COVID-19), known as convalescent plasma (CP). We tested if CP reduces viral shedding or disease in a nonhuman primate model. Our results demonstrate that administration of CP 1 day after SARS-CoV-2 infection had no significant impact on viral loads, clinical disease, or sequence diversity, although treatment with normal human plasma resulted in selection of a specific viral variant. Our results demonstrate that passive immunization with CP, even during early infection, provided no significant benefit in a nonhuman primate model of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Jesse D. Deere
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Timothy D. Carroll
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Joseph Dutra
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
| | - Linda Fritts
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Rebecca Lee Sammak
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - JoAnn L. Yee
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Katherine J. Olstad
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - J. Rachel Reader
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Amy Kistler
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Jack Kamm
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | | | | | - Sonny R. Elizaldi
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Jamin W. Roh
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Rachel E. Pollard
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Jodie Usachenko
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Brian A. Schmidt
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph DeRisi
- Chan Zuckerberg Biohub, San Francisco, California, USA
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, USA
| | - Michael P. Busch
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Smita S. Iyer
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Dennis J. Hartigan-O’Connor
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Christopher J. Miller
- Center for Immunology and Infectious Diseases, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- Department of Internal Medicine, Division of Infectious Diseases, School of Medicine, University of California Davis, Davis, California, USA
| |
Collapse
|
219
|
Rando HM, Wellhausen N, Ghosh S, Lee AJ, Dattoli AA, Hu F, Byrd JB, Rafizadeh DN, Lordan R, Qi Y, Sun Y, Brueffer C, Field JM, Ben Guebila M, Jadavji NM, Skelly AN, Ramsundar B, Wang J, Goel RR, Park Y, Boca SM, Gitter A, Greene CS. Identification and Development of Therapeutics for COVID-19. mSystems 2021; 6:e0023321. [PMID: 34726496 PMCID: PMC8562484 DOI: 10.1128/msystems.00233-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
After emerging in China in late 2019, the novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spread worldwide, and as of mid-2021, it remains a significant threat globally. Only a few coronaviruses are known to infect humans, and only two cause infections similar in severity to SARS-CoV-2: Severe acute respiratory syndrome-related coronavirus, a species closely related to SARS-CoV-2 that emerged in 2002, and Middle East respiratory syndrome-related coronavirus, which emerged in 2012. Unlike the current pandemic, previous epidemics were controlled rapidly through public health measures, but the body of research investigating severe acute respiratory syndrome and Middle East respiratory syndrome has proven valuable for identifying approaches to treating and preventing novel coronavirus disease 2019 (COVID-19). Building on this research, the medical and scientific communities have responded rapidly to the COVID-19 crisis and identified many candidate therapeutics. The approaches used to identify candidates fall into four main categories: adaptation of clinical approaches to diseases with related pathologies, adaptation based on virological properties, adaptation based on host response, and data-driven identification (ID) of candidates based on physical properties or on pharmacological compendia. To date, a small number of therapeutics have already been authorized by regulatory agencies such as the Food and Drug Administration (FDA), while most remain under investigation. The scale of the COVID-19 crisis offers a rare opportunity to collect data on the effects of candidate therapeutics. This information provides insight not only into the management of coronavirus diseases but also into the relative success of different approaches to identifying candidate therapeutics against an emerging disease. IMPORTANCE The COVID-19 pandemic is a rapidly evolving crisis. With the worldwide scientific community shifting focus onto the SARS-CoV-2 virus and COVID-19, a large number of possible pharmaceutical approaches for treatment and prevention have been proposed. What was known about each of these potential interventions evolved rapidly throughout 2020 and 2021. This fast-paced area of research provides important insight into how the ongoing pandemic can be managed and also demonstrates the power of interdisciplinary collaboration to rapidly understand a virus and match its characteristics with existing or novel pharmaceuticals. As illustrated by the continued threat of viral epidemics during the current millennium, a rapid and strategic response to emerging viral threats can save lives. In this review, we explore how different modes of identifying candidate therapeutics have borne out during COVID-19.
Collapse
Affiliation(s)
- Halie M. Rando
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Nils Wellhausen
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Soumita Ghosh
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alexandra J. Lee
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Ada Dattoli
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fengling Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Brian Byrd
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Diane N. Rafizadeh
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Yanjun Qi
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | - Yuchen Sun
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
| | | | - Jeffrey M. Field
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marouen Ben Guebila
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
| | - Nafisa M. Jadavji
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Ashwin N. Skelly
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | | | - Jinhui Wang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rishi Raj Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - YoSon Park
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - COVID-19 Review Consortium
BansalVikasBartonJohn P.BocaSimina M.BoerckelJoel D.BruefferChristianByrdJames BrianCaponeStephenDasShiktaDattoliAnna AdaDziakJohn J.FieldJeffrey M.GhoshSoumitaGitterAnthonyGoelRishi RajGreeneCasey S.GuebilaMarouen BenHimmelsteinDaniel S.HuFenglingJadavjiNafisa M.KamilJeremy P.KnyazevSergeyKollaLikhithaLeeAlexandra J.LordanRonanLubianaTiagoLukanTemitayoMacLeanAdam L.MaiDavidMangulSergheiManheimDavidMcGowanLucy D’AgostinoNaikAmrutaParkYoSonPerrinDimitriQiYanjunRafizadehDiane N.RamsundarBharathRandoHalie M.RaySandipanRobsonMichael P.RubinettiVincentSellElizabethShinholsterLamonicaSkellyAshwin N.SunYuchenSunYushaSzetoGregory L.VelazquezRyanWangJinhuiWellhausenNils
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Institute of Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Computer Science, University of Virginia, Charlottesville, Virginia, USA
- Department of Clinical Sciences, Lund University, Lund, Sweden
- Department of Biostatistics, Harvard School of Public Health, Boston, Massachusetts, USA
- Biomedical Science, Midwestern University, Glendale, Arizona, USA
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- The DeepChem Project
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| | - Simina M. Boca
- Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC, USA
- Early Biometrics & Statistical Innovation, Data Science & Artificial Intelligence, R & D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Anthony Gitter
- Department of Biostatistics and Medical Informatics, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Casey S. Greene
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado, USA
- Center for Health AI, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Childhood Cancer Data Lab, Alex’s Lemonade Stand Foundation, Philadelphia, Pennsylvania, USA
| |
Collapse
|
220
|
Abstract
The development of effective antiviral therapy for COVID-19 is critical for those awaiting vaccination, as well as for those who do not respond robustly to vaccination. This review summarizes 1 year of progress in the race to develop antiviral therapies for COVID-19, including research spanning preclinical and clinical drug development efforts, with an emphasis on antiviral compounds that are in clinical development or that are high priorities for clinical development. The review is divided into sections on compounds that inhibit SARS-CoV-2 enzymes, including its polymerase and proteases; compounds that inhibit virus entry, including monoclonal antibodies; interferons; and repurposed drugs that inhibit host processes required for SARS-CoV-2 replication. The review concludes with a summary of the lessons to be learned from SARS-CoV-2 drug development efforts and the challenges to continued progress.
Collapse
Affiliation(s)
- Kaiming Tao
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Philip L. Tzou
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Janin Nouhin
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Hector Bonilla
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Prasanna Jagannathan
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| | - Robert W. Shafer
- Division of Infectious Diseases, Department of Medicine, Stanford University, Stanford, California, USA
| |
Collapse
|
221
|
Bar KJ, Shaw PA, Choi GH, Aqui N, Fesnak A, Yang JB, Soto-Calderon H, Grajales L, Starr J, Andronov M, Mastellone M, Amonu C, Feret G, DeMarshall M, Buchanan M, Caturla M, Gordon J, Wanicur A, Monroy MA, Mampe F, Lindemuth E, Gouma S, Mullin AM, Barilla H, Pronina A, Irwin L, Thomas R, Eichinger RA, Demuth F, Luning Prak ET, Pascual JL, Short WR, Elovitz MA, Baron J, Meyer NJ, Degnan KO, Frank I, Hensley SE, Siegel DL, Tebas P. A randomized controlled study of convalescent plasma for individuals hospitalized with COVID-19 pneumonia. J Clin Invest 2021; 131:e155114. [PMID: 34788233 PMCID: PMC8670841 DOI: 10.1172/jci155114] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 11/01/2021] [Indexed: 11/17/2022] Open
Abstract
BackgroundAntibody-based strategies for COVID-19 have shown promise in prevention and treatment of early disease. COVID-19 convalescent plasma (CCP) has been widely used but results from randomized trials supporting its benefit in hospitalized patients with pneumonia are limited. Here, we assess the efficacy of CCP in severely ill, hospitalized adults with COVID-19 pneumonia.MethodsWe performed a randomized control trial (PennCCP2), with 80 adults hospitalized with COVID-19 pneumonia, comparing up to 2 units of locally sourced CCP plus standard care versus standard care alone. The primary efficacy endpoint was comparison of a clinical severity score. Key secondary outcomes include 14- and 28-day mortality, 14- and 28-day maximum 8-point WHO ordinal score (WHO8) score, duration of supplemental oxygenation or mechanical ventilation, respiratory SARS-CoV-2 RNA, and anti-SARS-CoV-2 antibodies.ResultsEighty hospitalized adults with confirmed COVID-19 pneumonia were enrolled at median day 6 of symptoms and day 1 of hospitalization; 60% were anti-SARS-CoV-2 antibody seronegative. Participants had a median of 3 comorbidities, including risk factors for severe COVID-19 and immunosuppression. CCP treatment was safe and conferred significant benefit by clinical severity score (median [MED] and interquartile range [IQR] 10 [5.5-30] vs. 7 [2.75-12.25], P = 0.037) and 28-day mortality (n = 10, 26% vs. n = 2, 5%; P = 0.013). All other prespecified outcome measures showed weak evidence toward benefit of CCP.ConclusionTwo units of locally sourced CCP administered early in hospitalization to majority seronegative participants conferred a significant benefit in clinical severity score and 28-day mortality. Results suggest CCP may benefit select populations, especially those with comorbidities who are treated early.Trial RegistrationClinicalTrials.gov NCT04397757.FundingUniversity of Pennsylvania.
Collapse
Affiliation(s)
- Katharine J. Bar
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pamela A. Shaw
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Kaiser Permanente Washington Health Research Group, Seattle, Washington, USA
| | - Grace H. Choi
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nicole Aqui
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Andrew Fesnak
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jasper B. Yang
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Kaiser Permanente Washington Health Research Group, Seattle, Washington, USA
| | | | - Lizette Grajales
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julie Starr
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michelle Andronov
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Miranda Mastellone
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Chigozie Amonu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Geoff Feret
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maureen DeMarshall
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marie Buchanan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria Caturla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James Gordon
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan Wanicur
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M. Alexandra Monroy
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Felicity Mampe
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Emily Lindemuth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sigrid Gouma
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anne M. Mullin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Holly Barilla
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anastasiya Pronina
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leah Irwin
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Raeann Thomas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Risa A. Eichinger
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Faye Demuth
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eline T. Luning Prak
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jose L. Pascual
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William R. Short
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Michal A. Elovitz
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jillian Baron
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nuala J. Meyer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kathleen O. Degnan
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ian Frank
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Scott E. Hensley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Donald L. Siegel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Pablo Tebas
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
222
|
Gallian P, Le Cam S, Brisbarre N, Pastorino B, Amroun A, Malard L, de Lamballerie X, Bliem C, Richard P, Morel P, Tiberghien P. COVID-19 convalescent plasma: Evolving strategies for serological screening in France. Vox Sang 2021; 117:606-610. [PMID: 34897745 DOI: 10.1111/vox.13228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 12/28/2022]
Abstract
Quantitation of anti-SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) neutralizing antibodies (Nabs) is a key parameter in determining the effective dose for treatment with COVID-19 convalescent plasma (CCP). Interpretation of results from clinical trials conducted worldwide requires comparison of Nabs titres obtained from different methods. As virus neutralization tests (VNTs) are not standardized scalable or commercially available, strategies based on intensity of ELISA (Enzyme Linked Immunosorbent Assay) or chemiluminescent binding serological tests were implemented to allow comparisons and establish criteria for determining 'high-titres' of anti-SARS-CoV-2 antibodies (Abs). To this end, the FDA (Food and Drug Administration) has proposed criteria to define high-titre plasmas using different serological assays, including the one used in France for the CCP SARS-CoV-2 Abs screening (Euroimmun anti-S1 IgG). A retrospective study revealed that when using the FDA criteria (ELISA signal-to-cut-off [S/C ratio] ≥3.5), 91% of CCP had Nabs titres ≥40 as assessed with an in-house VNT. French strategy to ensure sufficient stocks of CCP of increasing titre has evolved over time. Recently, we improved our strategy by collecting only plasma from vaccinated convalescent donors as we confirmed that the mean IgG antibody level (ELISA S/C ratio) was significantly higher in plasma from vaccinated convalescent donors compared to donations from unvaccinated convalescent donors: 9.31 (CI 95%: 8.46-10.16) versus 3.22 (CI 95%: 3.05-3.39) (p < 0.001).
Collapse
Affiliation(s)
- Pierre Gallian
- Etablissement français du Sang, La Plaine Saint Denis, France.,Unité des Virus Émergents (UVE), Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Sophie Le Cam
- Etablissement français du Sang, La Plaine Saint Denis, France
| | - Nadège Brisbarre
- Unité des Virus Émergents (UVE), Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France.,Etablissement français du Sang Provence Alpes Côte d'Azur et Corse, Marseille, France
| | - Boris Pastorino
- Unité des Virus Émergents (UVE), Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Abdennour Amroun
- Unité des Virus Émergents (UVE), Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Lucile Malard
- Etablissement français du Sang, La Plaine Saint Denis, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE), Aix-Marseille Univ, IRD 190, Inserm 1207, IHU Méditerranée Infection, Marseille, France
| | - Cathy Bliem
- Etablissement français du Sang, La Plaine Saint Denis, France
| | - Pascale Richard
- Etablissement français du Sang, La Plaine Saint Denis, France
| | - Pascal Morel
- Etablissement français du Sang, La Plaine Saint Denis, France.,UMR RIGHT 1098, Inserm, EFS, Université de Franche Comté, Besançon, France
| | - Pierre Tiberghien
- Etablissement français du Sang, La Plaine Saint Denis, France.,UMR RIGHT 1098, Inserm, EFS, Université de Franche Comté, Besançon, France
| |
Collapse
|
223
|
Golnabi EY, Sanders JM, Johns ML, Lin K, Ortwine JK, Wei W, Mang NS, Cutrell JB. Therapeutic Options for Coronavirus Disease 2019 (COVID-19): Where Are We Now? Curr Infect Dis Rep 2021; 23:28. [PMID: 34924819 PMCID: PMC8665318 DOI: 10.1007/s11908-021-00769-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE OF REVIEW Rapidly evolving treatment paradigms of coronavirus disease 2019 (COVID-19) introduce challenges for clinicians to keep up with the pace of published literature and to critically appraise the voluminous data produced. This review summarizes the clinical evidence from key studies examining the place of therapy of recommended drugs and management strategies for COVID-19. RECENT FINDINGS The global magnitude and duration of the pandemic have resulted in a flurry of interventional treatment trials evaluating both novel and repurposed drugs targeting various aspects of the viral life cycle. Additionally, clinical observations have documented various stages or phases of COVID-19 and underscored the importance of timing for the efficacy of studied therapies. Since the start of the COVID-19 pandemic, many observational, retrospective, and randomized controlled studies have been conducted to guide management of COVID-19 using drug therapies and other management strategies. Large, randomized, or adaptive platform trials have proven the most informative to guide recommended treatments to-date. Antimicrobial stewardship programs can play a pivotal role in ensuring appropriate use of COVID-19 therapies based on evolving clinical data and limiting unnecessary antibiotics given low rates of co-infection. SUMMARY Given the rapidly evolving medical literature and treatment paradigms, it is recommended to reference continuously updated, curated guidelines from national and international sources. While the drugs and management strategies mentioned in this review represent the current state of recommendations, many therapies are still under investigation to further define optimal COVID-19 treatment. SUPPLEMENTARY INFORMATION The online version contains supplementary material available at 10.1007/s11908-021-00769-8.
Collapse
Affiliation(s)
- Esther Y. Golnabi
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, US
| | - James M. Sanders
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, US
| | - Meagan L. Johns
- Department of Pharmacy, University of Texas Southwestern Medical Center, Dallas, US
| | - Kevin Lin
- Department of Pharmacy, Ochsner Medical Center, New Orleans, US
| | | | - Wenjing Wei
- Department of Pharmacy, Parkland Hospital, Dallas, US
| | | | - James B. Cutrell
- Department of Medicine, Division of Infectious Diseases and Geographic
Medicine, University of Texas Southwestern Medical Center, Dallas, US
| |
Collapse
|
224
|
Aksoyalp ZŞ, Nemutlu-Samur D. Sex-related susceptibility in coronavirus disease 2019 (COVID-19): Proposed mechanisms. Eur J Pharmacol 2021; 912:174548. [PMID: 34606834 PMCID: PMC8486578 DOI: 10.1016/j.ejphar.2021.174548] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/07/2021] [Accepted: 09/29/2021] [Indexed: 12/23/2022]
Abstract
The importance of sex differences is increasingly acknowledged in the incidence and treatment of disease. Accumulating clinical evidence demonstrates that sex differences are noticeable in COVID-19, and the prevalence, severity, and mortality rate of COVID-19 are higher among males than females. Sex-related genetic and hormonal factors and immunological responses may underlie the sex bias in COVID-19 patients. Angiotensin-converting enzyme 2 (ACE2) and transmembrane protease/serine subfamily member 2 (TMPRSS2) are essential proteins involved in the cell entry of SARS-CoV-2. Since ACE2 is encoded on the X-chromosome, a double copy of ACE2 in females may compensate for virus-mediated downregulation of ACE2, and thus ACE2-mediated cellular protection is greater in females. The X chromosome also contains the largest immune-related genes leading females to develop more robust immune responses than males. Toll-like receptor-7 (TLR-7), one of the key players in innate immunity, is linked to sex differences in autoimmunity and vaccine efficacy, and its expression is greater in females. Sex steroids also affect immune cell function. Estrogen contributes to higher CD4+ and CD8+ T cell activation levels, and females have more B cells than males. Sex differences not only affect the severity and progression of the disease, but also alter the efficacy of pharmacological treatment and adverse events related to the drugs/vaccines used against COVID-19. Administration of different drugs/vaccines in different doses or intervals may be useful to eliminate sex differences in efficacy and side/adverse effects. It should be noted that studies should include sex-specific analyses to develop further sex-specific treatments for COVID-19.
Collapse
Affiliation(s)
- Zinnet Şevval Aksoyalp
- Izmir Katip Celebi University, Faculty of Pharmacy, Department of Pharmacology, 35620, Izmir, Turkey.
| | - Dilara Nemutlu-Samur
- Alanya Alaaddin Keykubat University, Faculty of Medicine, Department of Pharmacology, 07450, Antalya, Turkey.
| |
Collapse
|
225
|
Lindahl H, Smith CIE, Bergman P. COVID-19 in a patient with Good's syndrome and in 13 patients with common variable immunodeficiency. CLINICAL IMMUNOLOGY COMMUNICATIONS 2021; 1:20-24. [PMID: 38620775 PMCID: PMC8497938 DOI: 10.1016/j.clicom.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 12/18/2022]
Abstract
Antibody deficiencies constitute the majority of primary immunodeficiencies in adults. These patients have a well-established increased risk of bacterial infections but there is a lack of knowledge regarding the relative risks upon contracting COVID-19. In this monocentric study the disease course of COVID-19 in 1 patient with Good's syndrome and in 13 patients with common variable immunodeficiency (CVID) is described. The severity of disease ranged from very mild to severe. Several patients required hospitalization and immunomodulatory treatment but all survived. Although viral infections are not a typical feature of humoral immunodeficiencies we recommend that vigilance is increased in the management of patients with Good's syndrome and CVID during the COVID-19 pandemic.
Collapse
Affiliation(s)
- Hannes Lindahl
- Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - C I Edvard Smith
- Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Immunodeficiency Unit, Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
226
|
Garcia-Muñoz R, Farfán-Quiroga G, Ruiz-de-Lobera N, Feliu J, Anton-Remirez J, Nájera Irazu MJ, Lisa Catón V, Oteo-Revuelta JA. Serology-based therapeutic strategy in SARS-CoV-2-infected patients. Int Immunopharmacol 2021; 101:108214. [PMID: 34649116 PMCID: PMC8491979 DOI: 10.1016/j.intimp.2021.108214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 12/22/2022]
Abstract
SARS-CoV-2 infection can be a life-threatening disease. The optimal treatment of patients is not yet standardized. We use a serology-based therapeutic strategy based on the presence of antibodies against the SARS-CoV-2 virus, in which patients with positive serology receive aggressive anti-inflammatory treatment with high-dose dexamethasone and/or tocilizumab and patients with negative serology receive early convalescent plasma therapy. We also analyze the immunological impact of this therapy in the recovery of T cells, B cells and NK cells during hospitalization in a COVID-19 infectious ward. Our results suggest that aggressive therapy with early administration of convalescent plasma and high-dose dexamethasone may be of benefit in patients with SARS-CoV-2 infection and might avoid progression of lung damage or need of admission in intensive care. This strategy did not impair immune responses against SARS-CoV-2, as 93% of the patients generated antibodies against the virus. Independently of previous immunological status of the patients, serology-guided therapy might benefit even patients with a high CIRS-G score, immunosuppressed or medically debilitated individuals and elderly patients. T cell disturbances were most frequent in patients who required high-dose dexamethasone, and B cell depletion was most frequent in patients who received tocilizumab. Early passive immunotherapy with convalescent plasma does not affect lymphoid recovery.
Collapse
Affiliation(s)
- Ricardo Garcia-Muñoz
- Hematology Department, Hospital San Pedro, Logroño, La Rioja. C/Piqueras 98, PC 26006, Spain.
| | - Giovanna Farfán-Quiroga
- Hematology Department, Hospital San Pedro, Logroño, La Rioja. C/Piqueras 98, PC 26006, Spain.
| | - Noemí Ruiz-de-Lobera
- Emergency Department, Hospital San Pedro, Logroño, La Rioja. C/Piqueras 98, PC 26006, Spain.
| | - Jesus Feliu
- Hematology Department, Hospital San Pedro, Logroño, La Rioja. C/Piqueras 98, PC 26006, Spain.
| | - Judith Anton-Remirez
- Department of Physical Medicine and Rehabilitation. Complejo Hospitalario de Navarra, Pamplona, Spain.
| | - Maria José Nájera Irazu
- Hematology Department, Hospital San Pedro, Logroño, La Rioja. C/Piqueras 98, PC 26006, Spain.
| | | | | |
Collapse
|
227
|
Senefeld JW, Johnson PW, Kunze KL, Bloch EM, van Helmond N, Golafshar MA, Klassen SA, Klompas AM, Sexton MA, Diaz Soto JC, Grossman BJ, Tobian AAR, Goel R, Wiggins CC, Bruno KA, van Buskirk CM, Stubbs JR, Winters JL, Casadevall A, Paneth NS, Shaz BH, Petersen MM, Sachais BS, Buras MR, Wieczorek MA, Russoniello B, Dumont LJ, Baker SE, Vassallo RR, Shepherd JRA, Young PP, Verdun NC, Marks P, Haley NR, Rea RF, Katz L, Herasevich V, Waxman DA, Whelan ER, Bergman A, Clayburn AJ, Grabowski MK, Larson KF, Ripoll JG, Andersen KJ, Vogt MNP, Dennis JJ, Regimbal RJ, Bauer PR, Blair JE, Buchholtz ZA, Pletsch MC, Wright K, Greenshields JT, Joyner MJ, Wright RS, Carter RE, Fairweather D. Access to and safety of COVID-19 convalescent plasma in the United States Expanded Access Program: A national registry study. PLoS Med 2021; 18:e1003872. [PMID: 34928960 PMCID: PMC8730442 DOI: 10.1371/journal.pmed.1003872] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 01/05/2022] [Accepted: 11/18/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The United States (US) Expanded Access Program (EAP) to coronavirus disease 2019 (COVID-19) convalescent plasma was initiated in response to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19. While randomized clinical trials were in various stages of development and enrollment, there was an urgent need for widespread access to potential therapeutic agents. The objective of this study is to report on the demographic, geographical, and chronological characteristics of patients in the EAP, and key safety metrics following transfusion of COVID-19 convalescent plasma. METHODS AND FINDINGS Mayo Clinic served as the central institutional review board for all participating facilities, and any US physician could participate as a local physician-principal investigator. Eligible patients were hospitalized, were aged 18 years or older, and had-or were at risk of progression to-severe or life-threatening COVID-19; eligible patients were enrolled through the EAP central website. Blood collection facilities rapidly implemented programs to collect convalescent plasma for hospitalized patients with COVID-19. Demographic and clinical characteristics of all enrolled patients in the EAP were summarized. Temporal patterns in access to COVID-19 convalescent plasma were investigated by comparing daily and weekly changes in EAP enrollment in response to changes in infection rate at the state level. Geographical analyses on access to convalescent plasma included assessing EAP enrollment in all national hospital referral regions, as well as assessing enrollment in metropolitan areas and less populated areas that did not have access to COVID-19 clinical trials. From April 3 to August 23, 2020, 105,717 hospitalized patients with severe or life-threatening COVID-19 were enrolled in the EAP. The majority of patients were 60 years of age or older (57.8%), were male (58.4%), and had overweight or obesity (83.8%). There was substantial inclusion of minorities and underserved populations: 46.4% of patients were of a race other than white, and 37.2% of patients were of Hispanic ethnicity. Chronologically and geographically, increases in the number of both enrollments and transfusions in the EAP closely followed confirmed infections across all 50 states. Nearly all national hospital referral regions enrolled and transfused patients in the EAP, including both in metropolitan and in less populated areas. The incidence of serious adverse events was objectively low (<1%), and the overall crude 30-day mortality rate was 25.2% (95% CI, 25.0% to 25.5%). This registry study was limited by the observational and pragmatic study design that did not include a control or comparator group; thus, the data should not be used to infer definitive treatment effects. CONCLUSIONS These results suggest that the EAP provided widespread access to COVID-19 convalescent plasma in all 50 states, including for underserved racial and ethnic minority populations. The study design of the EAP may serve as a model for future efforts when broad access to a treatment is needed in response to an emerging infectious disease. TRIAL REGISTRATION ClinicalTrials.gov NCT#: NCT04338360.
Collapse
Affiliation(s)
- Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Patrick W. Johnson
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Katie L. Kunze
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Evan M. Bloch
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Noud van Helmond
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Health Care, Camden, New Jersey, United States of America
| | - Michael A. Golafshar
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Stephen A. Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Allan M. Klompas
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthew A. Sexton
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Juan C. Diaz Soto
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Aaron A. R. Tobian
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ruchika Goel
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
- ImpactLife, Davenport, Iowa, United States of America
| | - Chad C. Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Camille M. van Buskirk
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - James R. Stubbs
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Jeffrey L. Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, College of Human Medicine, Michigan State University, East Lansing, Michigan, United States of America
- Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, United States of America
| | - Beth H. Shaz
- Department of Pathology, Duke University, Durham, North Carolina, United States of America
| | - Molly M. Petersen
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Bruce S. Sachais
- New York Blood Center Enterprises, New York City, New York, United States of America
| | - Matthew R. Buras
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, Arizona, United States of America
| | - Mikolaj A. Wieczorek
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Benjamin Russoniello
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Larry J. Dumont
- Vitalant Research Institute, Denver, Colorado, United States of America
- University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, United States of America
| | - Sarah E. Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | | | - John R. A. Shepherd
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Pampee P. Young
- American Red Cross, Washington, District of Columbia, United States of America
| | - Nicole C. Verdun
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - Peter Marks
- Center for Biologics Evaluation and Research, U. S. Food and Drug Administration, Silver Spring, Maryland, United States of America
| | - N. Rebecca Haley
- Bloodworks Northwest, Seattle, Washington, United States of America
| | - Robert F. Rea
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Louis Katz
- ImpactLife, Davenport, Iowa, United States of America
| | - Vitaly Herasevich
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Dan A. Waxman
- Versiti, Indianapolis, Indiana, United States of America
| | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, Florida, United States of America
| | - Aviv Bergman
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, New York City, New York, United States of America
| | - Andrew J. Clayburn
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Mary Kathryn Grabowski
- Division of Transfusion Medicine, Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Kathryn F. Larson
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Juan G. Ripoll
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Kylie J. Andersen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Matthew N. P. Vogt
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Joshua J. Dennis
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Riley J. Regimbal
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Philippe R. Bauer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Janis E. Blair
- Division of Infectious Diseases, Department of Internal Medicine, Mayo Clinic, Phoenix, Arizona, United States of America
| | - Zachary A. Buchholtz
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Michaela C. Pletsch
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Katherine Wright
- School of Sustainability, Arizona State University, Tempe, Arizona, United States of America
| | - Joel T. Greenshields
- Department of Kinesiology, Indiana University, Bloomington, Indiana, United States of America
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - R. Scott Wright
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, Florida, United States of America
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, Florida, United States of America
| |
Collapse
|
228
|
Harvala H, Gopal R, Patel M, Zambon M, Roberts D, Lamikanra A, Ploeg R, Hoogerwerf M, Zaaijer H, Hogema B, Reusken C, van der Schoot E, Reimerink J. Comparison of SARS-CoV-2 neutralizing antibody testing of convalescent plasma donations in the Netherlands and England: A pilot study. Health Sci Rep 2021; 4:e439. [PMID: 34888419 PMCID: PMC8637097 DOI: 10.1002/hsr2.439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/12/2021] [Accepted: 10/14/2021] [Indexed: 11/21/2022] Open
Affiliation(s)
- Heli Harvala
- National Microbiology ServicesNHS Blood and TransplantLondonUK
| | - Robin Gopal
- Virology Reference DepartmentNational Infection Service, Public Health EnglandLondonUK
| | - Monika Patel
- Virology Reference DepartmentNational Infection Service, Public Health EnglandLondonUK
| | - Maria Zambon
- Virology Reference DepartmentNational Infection Service, Public Health EnglandLondonUK
| | - David Roberts
- NHS Blood and TransplantJohn Radcliffe HospitalOxfordUK
- Radcliffe Department of Medicine and BRC Haematology ThemeUniversity of Oxford, John Radcliffe HospitalOxfordUK
| | - Abigail Lamikanra
- NHS Blood and TransplantJohn Radcliffe HospitalOxfordUK
- Radcliffe Department of Medicine and BRC Haematology ThemeUniversity of Oxford, John Radcliffe HospitalOxfordUK
| | - Rutger Ploeg
- Nuffield Department of Surgical SciencesUniversity of OxfordOxfordUK
- Organ Transplant CentreOxford University Hospitals NHS Foundation TrustOxfordUK
| | - Marieke Hoogerwerf
- Centre for Infectious Disease Control, WHO COVID‐19 Reference LaboratoryNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Hans Zaaijer
- Deparment of ImmunopathologySanquin Research and Landsteiner Laboratory Academic Medical CentreAmsterdamThe Netherlands
| | - Boris Hogema
- Deparment of ImmunopathologySanquin Research and Landsteiner Laboratory Academic Medical CentreAmsterdamThe Netherlands
| | - Chantal Reusken
- Centre for Infectious Disease Control, WHO COVID‐19 Reference LaboratoryNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| | - Ellen van der Schoot
- Deparment of ImmunopathologySanquin Research and Landsteiner Laboratory Academic Medical CentreAmsterdamThe Netherlands
| | - Johan Reimerink
- Centre for Infectious Disease Control, WHO COVID‐19 Reference LaboratoryNational Institute for Public Health and the EnvironmentBilthovenThe Netherlands
| |
Collapse
|
229
|
Stinca S, Barnes TW, Vogel P, Meyers W, Schulte-Pelkum J, Filchtinski D, Steller L, Hauser T, Manni S, Gardiner DF, Popik S, Roth NJ, Schuetz P. Modelling the concentration of anti-SARS-CoV-2 immunoglobulin G in intravenous immunoglobulin product batches. PLoS One 2021; 16:e0259731. [PMID: 34843493 PMCID: PMC8629175 DOI: 10.1371/journal.pone.0259731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 10/24/2021] [Indexed: 11/18/2022] Open
Abstract
Background Plasma-derived intravenous immunoglobulin (IVIg) products contain a dynamic spectrum of immunoglobulin (Ig) G reactivities reflective of the donor population from which they are derived. We sought to model the concentration of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) IgG which could be expected in future plasma pool and final-product batches of CSL Behring’s immunoglobulin product Privigen. Study design and methods Data was extracted from accessible databases, including the incidence of coronavirus disease 2019 and SARS-CoV-2 vaccination status, antibody titre in convalescent and vaccinated groups and antibody half-life. Together, these parameters were used to create an integrated mathematical model that could be used to predict anti-SARS-CoV-2 antibody levels in future IVIg preparations. Results We predict that anti-SARS-CoV-2 IgG concentration will peak in batches produced in mid-October 2021, containing levels in the vicinity of 190-fold that of the mean convalescent (unvaccinated) plasma concentration. An elevated concentration (approximately 35-fold convalescent plasma) is anticipated to be retained in batches produced well into 2022. Measurement of several Privigen batches using the Phadia™ EliA™ SARS-CoV-2-Sp1 IgG binding assay confirmed the early phase of this model. Conclusion The work presented in this paper may have important implications for physicians and patients who use Privigen for indicated diseases.
Collapse
Affiliation(s)
- Sara Stinca
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Thomas W. Barnes
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Peter Vogel
- Global Digital Core, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, Marburg, Germany
| | - Wilfried Meyers
- Global Digital Core, Plasma Product Development, Research & Development, CSL Behring Innovation GmbH, Marburg, Germany
| | | | - Daniel Filchtinski
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Freiburg, Germany
| | - Laura Steller
- Assay Design, Thermo Fisher Scientific ImmunoDiagnostics Phadia GmbH, Freiburg, Germany
| | - Thomas Hauser
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Sandro Manni
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - David F. Gardiner
- Immunology, CSL Behring, King of Prussia, Pennsylvania, United States of America
| | - Sharon Popik
- Immunology, CSL Behring, King of Prussia, Pennsylvania, United States of America
| | - Nathan J. Roth
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
| | - Patrick Schuetz
- Department of Bioanalytical Sciences, Plasma Product Development, Research & Development, CSL Behring AG, Bern, Switzerland
- * E-mail:
| |
Collapse
|
230
|
Lanza F, Agostini V, Monaco F, Passamonti F, Seghatchian J. Therapeutic Use of Convalescent Plasma in COVID-19 Infected Patients with Concomitant Hematological Disorders. Clin Hematol Int 2021; 3:77-82. [PMID: 34820612 PMCID: PMC8486975 DOI: 10.2991/chi.k.210403.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 03/17/2021] [Indexed: 12/27/2022] Open
Abstract
The use of convalescent plasma (CP) from individuals recovered from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a promising therapeutic modality for the coronavirus disease 2019 (COVID-19). CP has been in use for at least a century to provide passive immunity against a number of diseases, and was recently proposed by the World Health Organization for human Ebola virus infection. Only a few small studies have so far been published on patients with COVID-19 and concomitant hematological malignancies (HM). The Italian Hematology Alliance on HM and COVID-19 has found that HM patients with COVID-19 clinically perform more poorly than those with either HM or COVID-19 alone. A COVID-19 infection in patients with B-cell lymphoma is associated with impaired generation of neutralizing antibody titers and lowered clearance of SARS-CoV-2. Treatment with CP was seen to increase antibody titers in all patients and to improve clinical response in 80% of patients examined. However, a recent study has reported impaired production of SARS-CoV-2-neutralizing antibodies in an immunosuppressed individual treated with CP, possibly supporting the notion of virus escape, particularly in immunocompromised individuals where prolonged viral replication occurs. This may limit the efficacy of CP treatment in at least some HM patients. More recently, it has been shown that CP may provide a neutralising effect against B.1.1.7 and other SARS-CoV-2 variants, thus expanding its application in clinical practice. More extensive studies are needed to further assess the use of CP in COVID-19-infected HM patients.
Collapse
Affiliation(s)
- Francesco Lanza
- Hematology Unit & Romagna Transplant Network, Ravenna, Italy
| | - Vanessa Agostini
- Transfusion Medicine Department, IRCCS- Ospedale Policlinico San Martino, Genova, Italy
| | - Federica Monaco
- Hematology Unit & Romagna Transplant Network, Ravenna, Italy
| | | | - Jerard Seghatchian
- International Consultancy in Innovative Manufacturing and Quality/Safety of Blood-Derived Bioproducts, London, England, UK
| |
Collapse
|
231
|
Fazeli A, Sharifi S, Behdad F, Okati S, Esmaielifar G, Jelveh N, Eshghi P, Mohammadi S. Early high-titer convalescent plasma therapy in patients with moderate and severe COVID-19. Transfus Apher Sci 2021; 61:103321. [PMID: 34836825 PMCID: PMC8612443 DOI: 10.1016/j.transci.2021.103321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/06/2021] [Accepted: 11/13/2021] [Indexed: 12/27/2022]
Abstract
BACKGROUND AND OBJECTIVES The use of COVID-19 convalescent plasma (CCP) has been approved by the FDA. We assessed the outcome of patients with moderate and severe COVID-19 following convalescent plasma therapy and the association with variables such as antibody titer in CCP units and transfusion time. MATERIALS AND METHODS In this prospective cohort study, 3097 patients with moderate and severe COVID-19 (according to WHO Progression Scale) had heterogeneous demographic and clinical characteristics received plasma with an unknown titer at the transfusion time. Firstly, information about age, sex, blood group, the time interval from hospitalization to CCP transfusion, underlying disease, and antibody titer with the outcome were investigated. Then, multivariate logistic regression and area under the curve (AUC) were performed for the association between disease severity and intubation variables with transfusion time and outcome. RESULTS Patients with younger age receiving CCP in the first five days of hospitalization had lower mortality (P < 0.0001). Moreover, patients without the underlying disease had lower mortality (P < 0.001). The mortality rate also decreased in severe patients who were intubated receiving CCP for less than five days (P < 0.001). In patients with moderate severity (score less than 5) who received IgG antibody levels above 1:320 in less than five days had lower mortality (P < 0.0001). CONCLUSION Our findings suggested that COVID-19 patients with the moderate type of disease receiving CCP units with high antibody titers in the early stages of the disease experienced greater effectiveness of CCP therapy.
Collapse
Affiliation(s)
- Alieh Fazeli
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shahin Sharifi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Fatemeh Behdad
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Shamsi Okati
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Gilda Esmaielifar
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Nooshin Jelveh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Peyman Eshghi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Saeed Mohammadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran; Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran; Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
232
|
Korley FK, Durkalski-Mauldin V, Yeatts SD, Schulman K, Davenport RD, Dumont LJ, El Kassar N, Foster LD, Hah JM, Jaiswal S, Kaplan A, Lowell E, McDyer JF, Quinn J, Triulzi DJ, Van Huysen C, Stevenson VLW, Yadav K, Jones CW, Kea B, Burnett A, Reynolds JC, Greineder CF, Haas NL, Beiser DG, Silbergleit R, Barsan W, Callaway CW. Early Convalescent Plasma for High-Risk Outpatients with Covid-19. N Engl J Med 2021; 385:1951-1960. [PMID: 34407339 PMCID: PMC8385553 DOI: 10.1056/nejmoa2103784] [Citation(s) in RCA: 151] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Early administration of convalescent plasma obtained from blood donors who have recovered from coronavirus disease 2019 (Covid-19) may prevent disease progression in acutely ill, high-risk patients with Covid-19. METHODS In this randomized, multicenter, single-blind trial, we assigned patients who were being treated in an emergency department for Covid-19 symptoms to receive either one unit of convalescent plasma with a high titer of antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or placebo. All the patients were either 50 years of age or older or had one or more risk factors for disease progression. In addition, all the patients presented to the emergency department within 7 days after symptom onset and were in stable condition for outpatient management. The primary outcome was disease progression within 15 days after randomization, which was a composite of hospital admission for any reason, seeking emergency or urgent care, or death without hospitalization. Secondary outcomes included the worst severity of illness on an 8-category ordinal scale, hospital-free days within 30 days after randomization, and death from any cause. RESULTS A total of 511 patients were enrolled in the trial (257 in the convalescent-plasma group and 254 in the placebo group). The median age of the patients was 54 years; the median symptom duration was 4 days. In the donor plasma samples, the median titer of SARS-CoV-2 neutralizing antibodies was 1:641. Disease progression occurred in 77 patients (30.0%) in the convalescent-plasma group and in 81 patients (31.9%) in the placebo group (risk difference, 1.9 percentage points; 95% credible interval, -6.0 to 9.8; posterior probability of superiority of convalescent plasma, 0.68). Five patients in the plasma group and 1 patient in the placebo group died. Outcomes regarding worst illness severity and hospital-free days were similar in the two groups. CONCLUSIONS The administration of Covid-19 convalescent plasma to high-risk outpatients within 1 week after the onset of symptoms of Covid-19 did not prevent disease progression. (SIREN-C3PO ClinicalTrials.gov number, NCT04355767.).
Collapse
Affiliation(s)
- Frederick K Korley
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Valerie Durkalski-Mauldin
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Sharon D Yeatts
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Kevin Schulman
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Robertson D Davenport
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Larry J Dumont
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Nahed El Kassar
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Lydia D Foster
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Jennifer M Hah
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Siddartha Jaiswal
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Alesia Kaplan
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Ezekiel Lowell
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - John F McDyer
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - James Quinn
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Darrell J Triulzi
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Carol Van Huysen
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Valerie L W Stevenson
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Kabir Yadav
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Christopher W Jones
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Bory Kea
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Aaron Burnett
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Joshua C Reynolds
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Colin F Greineder
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Nathan L Haas
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - David G Beiser
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Robert Silbergleit
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - William Barsan
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| | - Clifton W Callaway
- From the University of Michigan, Ann Arbor (F.K.K., R.D.D., C.V.H., V.L.W.S., C.F.G., N.L.H., R.S., W.B.), Spectrum Health, Grand Rapids (J.C.R.), and Michigan State University, East Lansing (J.C.R.) - all in Michigan; the Medical University of South Carolina, Charleston (V.D.-M., S.D.Y., L.D.F., E.L.); Stanford University, Palo Alto, CA (K.S., J.M.H., S.J., J.Q.); Vitalant Research Institute, Scottsdale, AZ (L.J.D.); the National Heart, Lung, and Blood Institute, Bethesda, MD (N.E.K.); the University of Pittsburgh, Pittsburgh (A.K., J.F.M., D.J.T., C.W.C.); Harbor-UCLA Medical Center, Los Angeles (K.Y.); Cooper University Hospital, Camden, NJ (C.W.J.); Oregon Health and Science University, Portland (B.K.); Health Partners Methodist Hospital, St. Louis Park, MN (A.B.); and the University of Chicago, Chicago (D.G.B.)
| |
Collapse
|
233
|
Skorek A, Jaźwińska-Curyłło A, Romanowicz A, Kwaśniewski K, Narożny W, Tretiakow D. Assessment of anti-SARS-CoV-2 antibodies level in convalescents plasma. J Med Virol 2021; 94:1130-1137. [PMID: 34738646 PMCID: PMC8661642 DOI: 10.1002/jmv.27433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 10/18/2021] [Accepted: 11/02/2021] [Indexed: 11/10/2022]
Abstract
Despite extensive vaccination, the quantity of patients infected with the SARS-CoV-2 virus and its variants continues to grow worldwide. Treating patients with a severe course of COVID-19 is a difficult challenge. One of the generally accepted and specific therapy methods is the use of plasma rich in anti-SARS-CoV-2 antibodies. On the other hand, assessing the antibodies level depending on the time after infection allows for vaccine-decision. The study marked the level of anti-SARS-CoV-2 IgG antibodies in 351 COVID-19 convalescent residents of one geographical region in Poland. The study group included blood donors. The studies were cross-sectional and extended to a questionnaire to determine infection severity. These data were compiled statistically. The study considered epidemiological factors, the time from the end of the infection, and infection severity. The fastest increase of the antibodies level was observed up to 59 days after COVID-19, and it was statistically significantly higher among men. Higher levels of antibodies were found among people above the average age in both men and women. There was an increase in the level of antibodies since the onset of the disease in men, while in women, it decreased. The antibodies level was also found to depend on the severity of the course of COVID-19 infection. The optimal group of plasma donors in the studied geographical region is men and women above 39 years old. after a more severe infection. The titer of antibodies increases with time from the disease.
Collapse
Affiliation(s)
- Andrzej Skorek
- Department of Otolaryngology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Aleksandra Romanowicz
- Student Scientific Club at the Department of Otolaryngology, Medical University of Gdańsk, Gdańsk, Poland
| | - Krzysztof Kwaśniewski
- Vascular Surgery and Angiology Department, Antoni Jurasz University Hospital No. 1, Bydgoszcz, Poland
| | - Waldemar Narożny
- Department of Otolaryngology, Medical University of Gdańsk, Gdańsk, Poland
| | - Dmitry Tretiakow
- Department of Otolaryngology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
234
|
Prudente TP, Castro RG, Candido MA, Rodrigues RL, de Souza LM, Roberti MDRF. Antibody response against SARS-CoV-2 in convalescent plasma donors: can we predict subjects' eligibility? Hematol Transfus Cell Ther 2021; 44:1-6. [PMID: 34751255 PMCID: PMC8566374 DOI: 10.1016/j.htct.2021.07.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/13/2021] [Indexed: 01/10/2023] Open
Abstract
Introduction As the Coronavirus Disease 2019 (COVID-19) pandemic unfolds around the world; answers related to the antibody response against the virus are necessary to develop treatment and prophylactic strategies. We attempted to understand part of the immune response of convalescent plasma donation candidates. Method We carried out a cross-sectional, observational, non-intervention study, testing 102 convalescent plasma donation candidates for antibodies against the virus, relating these data to the time interval between symptom onset and sample collection, age, disease severity, and gender. Results In our sample, the individuals who developed a greater antibody response were the ones who had a longer time interval between symptom onset and sample collection, the ones who had been hospitalized and the subjects above 35 years old. Moreover, 17 individuals did not present any reactive antibodies. Conclusion These results are important in that they raise questions about the role of the humoral response against the virus, as some individuals do not develop antibodies to fight it. In addition, they help develop recruitment strategies for convalescent plasma donors, who should be asymptomatic for at least 21 days and are possibly more likely to have reactive antibodies after 35 days without symptoms.
Collapse
Affiliation(s)
- Tiago Paiva Prudente
- Faculdade de Medicina da Universidade Federal de Goiás (FM-UFG), Goiânia, GO, Brazil
| | - Renato Gomes Castro
- Faculdade de Medicina da Universidade Federal de Goiás (FM-UFG), Goiânia, GO, Brazil
| | | | | | | | - Maria do Rosario Ferraz Roberti
- Faculdade de Medicina da Universidade Federal de Goiás (FM-UFG), Goiânia, GO, Brazil.,Hemocentro de Goiás (HEMOGO), Goiânia, GO, Brazil
| |
Collapse
|
235
|
Gunn BM, Bai S. Building a better antibody through the Fc: advances and challenges in harnessing antibody Fc effector functions for antiviral protection. Hum Vaccin Immunother 2021; 17:4328-4344. [PMID: 34613865 PMCID: PMC8827636 DOI: 10.1080/21645515.2021.1976580] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 08/23/2021] [Accepted: 08/30/2021] [Indexed: 12/15/2022] Open
Abstract
Antibodies can provide antiviral protection through neutralization and recruitment of innate effector functions through the Fc domain. While neutralization has long been appreciated for its role in antibody-mediated protection, a growing body of work indicates that the antibody Fc domain also significantly contributes to antiviral protection. Recruitment of innate immune cells such as natural killer cells, neutrophils, monocytes, macrophages, dendritic cells and the complement system by antibodies can lead to direct restriction of viral infection as well as promoting long-term antiviral immunity. Monoclonal antibody therapeutics against viruses are increasingly incorporating Fc-enhancing features to take advantage of the Fc domain, uncovering a surprising breadth of mechanisms through which antibodies can control viral infection. Here, we review the recent advances in our understanding of antibody-mediated innate immune effector functions in protection from viral infection and review the current approaches and challenges to effectively leverage innate immune cells via antibodies.
Collapse
Affiliation(s)
- Bronwyn M. Gunn
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| | - Shuangyi Bai
- Paul G. Allen School of Global Health, College of Veterinary Medicine, Washington State University, Pullman, WA, USA
| |
Collapse
|
236
|
Brüssow H. Clinical trials with antiviral drugs against COVID-19: some progress and many shattered hopes. Environ Microbiol 2021; 23:6364-6376. [PMID: 34519154 PMCID: PMC8652531 DOI: 10.1111/1462-2920.15769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/27/2022]
Abstract
Vaccines and drugs are the cornerstones in the fight against the SARS-CoV-2 pandemic. While vaccines were a success story, the development of antiviral drugs against SARS-CoV-2 turned out to be difficult. For an accelerated use of antivirals in the clinic, most SARS-CoV-2 antivirals represented repurposed drugs. The present article summarizes the outcomes of clinical trials with antiviral drugs in COVID-19 patients. Many antiviral drugs failed to demonstrate beneficial effects or showed mixed results. One reason for the low success rate of clinical trials was shortcomings of antiviral tests in cell culture systems and another reason was the abundance of ill-coordinated and underpowered clinical trials. However, large pragmatic clinical trials particularly of the British RECOVERY trial series demonstrated that even under emergency situation drug trials can be conducted in a timely way such that the therapy of COVID-19 patients can be based on evidence basis instead on expert opinion or even worse on political pressure.
Collapse
Affiliation(s)
- Harald Brüssow
- Department of Biosystems, Laboratory of Gene TechnologyKU LeuvenLeuvenBelgium
| |
Collapse
|
237
|
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
|
238
|
Calvo C, Tagarro A, Méndez Echevarría A, Fernández Colomer B, Albañil Ballesteros MR, Bassat Q, Mellado Peña MJ. COVID-19 pandemic. What have we learned? An Pediatr (Barc) 2021; 95:382.e1-382.e8. [PMID: 34728170 PMCID: PMC8529266 DOI: 10.1016/j.anpede.2021.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/20/2021] [Indexed: 02/05/2023] Open
Abstract
Since the COVID-19 pandemic was declared in March 2020, we have learned a lot about the SARS-CoV-2 coronavirus, and its role in pediatric pathology. Children are infected in a rate quite similar to adults, although in most cases they suffer mild or asymptomatic symptoms. Around 1% of those infected require hospitalization, less than 0.02% require intensive care, and mortality is very low and generally in children with comorbidities. The most common clinical diagnoses are upper or lower respiratory infections, gastrointestinal infection and, more seriously, multisystemic inflammatory syndrome (MIS-C). Most episodes do not require treatment, except for MIS-C. Remdesivir has been widely used as a compassionate treatment and its role has yet to be defined. The newborn can become infected, although vertical transmission is very low (<1%) and it has been shown that the baby can safely cohabit with its mother and be breastfed. In general, neonatal infections have been mild. Primary care has supported a very important part of the management of the pandemic in pediatrics. There has been numerous collateral damage derived from the difficulty of access to care and the isolation suffered by children. The mental health of the pediatric population has been seriously affected. Although it has been shown that schooling has not led to an increase in infections, but rather the opposite. It is essential to continue maintaining the security measures that make schools a safe place, so necessary not only for children's education, but for their health in general.
Collapse
Affiliation(s)
- Cristina Calvo
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales, Hospital Universitario La Paz, Madrid, Spain; Red de Investigación traslacional en infectología Pediátrica (RITIP), Spain.
| | - Alfredo Tagarro
- Red de Investigación traslacional en infectología Pediátrica (RITIP), Spain; Unidad de Pediatría Investigación y Ensayos Clínicos (UPIC), Instituto de Investigación Sanitaria Hospital 12 de Octubre (IMAS12), Madrid; Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid; Servicio de Pediatría, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, Madrid, Spain
| | - Ana Méndez Echevarría
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales, Hospital Universitario La Paz, Madrid, Spain; Red de Investigación traslacional en infectología Pediátrica (RITIP), Spain
| | | | - M Rosa Albañil Ballesteros
- Centro de Salud Cuzco, Fuenlabrada, Madrid; Grupo de Patología Infecciosa de la Asociación Española de Pediatría de Atención Primaria (AEPap), Spain
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; ICREA, Barcelona, Spain; Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues de Llobregat, Barcelona, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - M José Mellado Peña
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales, Hospital Universitario La Paz, Madrid, Spain; Red de Investigación traslacional en infectología Pediátrica (RITIP), Spain
| |
Collapse
|
239
|
Calvo C, Tagarro A, Méndez Echevarría A, Fernández Colomer B, Albañil Ballesteros MR, Bassat Q, Mellado Peña MJ. [COVID-19 pandemic. What have we learned?]. An Pediatr (Barc) 2021; 95:382.e1-382.e8. [PMID: 34580593 PMCID: PMC8457926 DOI: 10.1016/j.anpedi.2021.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 09/20/2021] [Indexed: 12/30/2022] Open
Abstract
Since the COVID-19 pandemic was declared in March 2020, we have learned a lot about the SARS-CoV-2 coronavirus, and its role in pediatric pathology.Children are infected in a rate quite similar to adults, although in most cases they suffer mild or asymptomatic symptoms. Around 1% of those infected require hospitalization, less than 0.02% require intensive care, and mortality is very low and generally in children with comorbidities. The most common clinical diagnoses are upper or lower respiratory infections, gastrointestinal infection and, more seriously, multisystemic inflammatory syndrome (MIS-C). Most episodes do not require treatment, except for MIS-C. Remdesivir has been widely used as a compassionate treatment and its role has yet to be defined.The newborn can become infected, although vertical transmission is very low (<1%) and it has been shown that the baby can safely cohabit with its mother and be breastfed. In general, neonatal infections have been mild.Primary care has supported a very important part of the management of the pandemic in pediatrics. There has been numerous collateral damage derived from the difficulty of access to care and the isolation suffered by children. The mental health of the pediatric population has been seriously affected. Although it has been shown that schooling has not led to an increase in infections, but rather the opposite. It is essential to continue maintaining the security measures that make schools a safe place, so necessary not only for children's education, but for their health in general.
Collapse
Affiliation(s)
- Cristina Calvo
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales. Hospital Universitario La Paz, Madrid, España
- Red de Investigación traslacional en infectología Pediátrica (RITIP), España
| | - Alfredo Tagarro
- Red de Investigación traslacional en infectología Pediátrica (RITIP), España
- Unidad de Pediatría Investigación y Ensayos Clínicos (UPIC), Instituto de Investigación Sanitaria Hospital 12 de Octubre (IMAS12), Madrid; Fundación para la Investigación Biomédica del Hospital 12 de Octubre, Madrid; Servicio de Pediatría, Hospital Universitario Infanta Sofía, Universidad Europea de Madrid, Madrid, España
| | - Ana Méndez Echevarría
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales. Hospital Universitario La Paz, Madrid, España
- Red de Investigación traslacional en infectología Pediátrica (RITIP), España
| | | | - María Rosa Albañil Ballesteros
- Centro de Salud Cuzco, Fuenlabrada, Madrid; Grupo de Patología Infecciosa de la Asociación Española de Pediatría de Atención Primaria (AEPap), España
| | - Quique Bassat
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, España
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
- ICREA, Barcelona, España
- Pediatrics Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Esplugues de Llobregat, Barcelona, España
- Consorcio de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), Madrid, España
| | - Maria José Mellado Peña
- Servicio de Pediatría, Enfermedades Infecciosas y Tropicales. Hospital Universitario La Paz, Madrid, España
- Red de Investigación traslacional en infectología Pediátrica (RITIP), España
| |
Collapse
|
240
|
Felsenstein S, Reiff AO. A hitchhiker's guide through the COVID-19 galaxy. Clin Immunol 2021; 232:108849. [PMID: 34563684 PMCID: PMC8461017 DOI: 10.1016/j.clim.2021.108849] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 09/04/2021] [Indexed: 01/08/2023]
Abstract
Numerous reviews have summarized the epidemiology, pathophysiology and the various therapeutic aspects of Coronavirus disease 2019 (COVID-19), but a practical guide on "how to treat whom with what and when" based on an understanding of the immunological background of the disease stages remains missing. This review attempts to combine the current knowledge about the immunopathology of COVID-19 with published evidence of available and emerging treatment options. We recognize that the information about COVID-19 and its treatment is rapidly changing, but hope that this guide offers those on the frontline of this pandemic an understanding of the host response in COVID-19 patients and supports their ongoing efforts to select the best treatments tailored to their patient's clinical status.
Collapse
Affiliation(s)
- Susanna Felsenstein
- University of Liverpool, Faculty of Health and Life Sciences, Brownlow Hill, Liverpool, L69 3GB, United Kingdom.
| | - Andreas Otto Reiff
- Arthritis & Rheumatic Diseases, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, United States.
| |
Collapse
|
241
|
Menichetti F, Popoli P, Puopolo M, Spila Alegiani S, Tiseo G, Bartoloni A, De Socio GV, Luchi S, Blanc P, Puoti M, Toschi E, Massari M, Palmisano L, Marano G, Chiamenti M, Martinelli L, Franchi S, Pallotto C, Suardi LR, Luciani Pasqua B, Merli M, Fabiani P, Bertolucci L, Borchi B, Modica S, Moneta S, Marchetti G, d’Arminio Monforte A, Stoppini L, Ferracchiato N, Piconi S, Fabbri C, Beccastrini E, Saccardi R, Giacometti A, Esperti S, Pierotti P, Bernini L, Bianco C, Benedetti S, Lanzi A, Bonfanti P, Massari M, Sani S, Saracino A, Castagna A, Trabace L, Lanza M, Focosi D, Mazzoni A, Pistello M, Falcone M. Effect of High-Titer Convalescent Plasma on Progression to Severe Respiratory Failure or Death in Hospitalized Patients With COVID-19 Pneumonia: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2136246. [PMID: 34842924 PMCID: PMC8630572 DOI: 10.1001/jamanetworkopen.2021.36246] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
IMPORTANCE Convalescent plasma (CP) has been generally unsuccessful in preventing worsening of respiratory failure or death in hospitalized patients with COVID-19 pneumonia. OBJECTIVE To evaluate the efficacy of CP plus standard therapy (ST) vs ST alone in preventing worsening respiratory failure or death in patients with COVID-19 pneumonia. DESIGN, SETTING, AND PARTICIPANTS This prospective, open-label, randomized clinical trial enrolled (1:1 ratio) hospitalized patients with COVID-19 pneumonia to receive CP plus ST or ST alone between July 15 and December 8, 2020, at 27 clinical sites in Italy. Hospitalized adults with COVID-19 pneumonia and a partial pressure of oxygen-to-fraction of inspired oxygen (Pao2/Fio2) ratio between 350 and 200 mm Hg were eligible. INTERVENTIONS Patients in the experimental group received intravenous high-titer CP (≥1:160, by microneutralization test) plus ST. The volume of infused CP was 200 mL given from 1 to a maximum of 3 infusions. Patients in the control group received ST, represented by remdesivir, glucocorticoids, and low-molecular weight heparin, according to the Agenzia Italiana del Farmaco recommendations. MAIN OUTCOMES AND MEASURES The primary outcome was a composite of worsening respiratory failure (Pao2/Fio2 ratio <150 mm Hg) or death within 30 days from randomization. RESULTS Of the 487 randomized patients (241 to CP plus ST; 246 to ST alone), 312 (64.1%) were men; the median (IQR) age was 64 (54.0-74.0) years. The modified intention-to-treat population included 473 patients. The primary end point occurred in 59 of 231 patients (25.5%) treated with CP and ST and in 67 of 239 patients (28.0%) who received ST (odds ratio, 0.88; 95% CI, 0.59-1.33; P = .54). Adverse events occurred more frequently in the CP group (12 of 241 [5.0%]) compared with the control group (4 of 246 [1.6%]; P = .04). CONCLUSIONS AND RELEVANCE In patients with moderate to severe COVID-19 pneumonia, high-titer anti-SARS-CoV-2 CP did not reduce the progression to severe respiratory failure or death within 30 days. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04716556.
Collapse
Affiliation(s)
- Francesco Menichetti
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Puopolo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | - Giusy Tiseo
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Alessandro Bartoloni
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Giuseppe Vittorio De Socio
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Sauro Luchi
- Infectious Disease Unit, Hospital of Lucca, Lucca, Italy
| | - Pierluigi Blanc
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Massimo Puoti
- University of Milano-Bicocca School of Medicine, Milan, Italy
- Azienda socio sanitaria territorial (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elena Toschi
- Research Coordination and Support Service (CoRi), Istituto Superiore di Sanità, Rome, Italy
| | - Marco Massari
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Palmisano
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Laura Martinelli
- Internal Medicine, Unità Sanitaria Locale (USL)–Umbria 1, Ospedale Città di Castello, Città di Castello, Italy
| | - Silvia Franchi
- Internal Medicine, Unità Sanitaria Locale (USL)–Umbria 1, Ospedale Città di Castello, Città di Castello, Italy
| | - Carlo Pallotto
- Infectious Diseases Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, Empoli, Italy
| | - Lorenzo Roberto Suardi
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
- Infectious Diseases Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, Empoli, Italy
| | - Barbara Luciani Pasqua
- Centro Regionale Sangue, Servizio Immunotrasfusionale, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Marco Merli
- Azienda socio sanitaria territorial (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Plinio Fabiani
- Internal Medicine, Ospedale Unico della Versilia, Lido di Camaiore, Italy
| | - Luca Bertolucci
- Internal Medicine, Ospedale Unico della Versilia, Lido di Camaiore, Italy
| | - Beatrice Borchi
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Sara Modica
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Sara Moneta
- Infectious Disease Unit, Hospital of Lucca, Lucca, Italy
| | - Giulia Marchetti
- Infectious Diseases Unit, Department of Health Sciences, ASST Santi Paolo e Carlo University Hospital, Milan, Italy
| | - Antonella d’Arminio Monforte
- Infectious Diseases Unit, Department of Health Sciences, ASST Santi Paolo e Carlo University Hospital, Milan, Italy
| | | | | | - Stefania Piconi
- Infectious Diseases, Azienda Ospedaliera di Lecco, Lecco, Italy
| | - Claudio Fabbri
- Infectious Diseases, Ospedale San Jacopo, Pistoia, Italy
| | - Enrico Beccastrini
- Cell Therapy and Transfusion Medicine, Careggi University Hospital, Florence, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine, Careggi University Hospital, Florence, Italy
| | - Andrea Giacometti
- Azienda Ospedaliera Universitaria, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Sara Esperti
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Piera Pierotti
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Laura Bernini
- Division of Infectious Diseases, Arezzo Hospital, Arezzo, Italy
| | - Claudia Bianco
- Division of Infectious Diseases, Arezzo Hospital, Arezzo, Italy
| | - Sara Benedetti
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Alessandra Lanzi
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, ASST Monza, University of Milano-Bicocca, Milan, Italy
| | - Marco Massari
- Infectious Disease Unit, Azienda USL–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) di Reggio Emilia, Reggio Emilia, Italy
| | - Spartaco Sani
- Infectious Diseases, Livorno Hospital, Livorno, Italy
| | - Annalisa Saracino
- Division of Infectious Diseases, Bari University Hospital, Bari, Italy
| | - Antonella Castagna
- Infectious Diseases, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Luigia Trabace
- Department of Experimental and Clinical Medicine, University of Foggia, Foggia, Italy
| | - Maria Lanza
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Alessandro Mazzoni
- Division of Transfusion Medicine and Transplant Biology, Pisa University Hospital, Pisa, Italy
| | - Mauro Pistello
- Division of Virology, University Hospital of Pisa, Retrovirus Center, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Marco Falcone
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| |
Collapse
|
242
|
Structural Basis of a Human Neutralizing Antibody Specific to the SARS-CoV-2 Spike Protein Receptor-Binding Domain. Microbiol Spectr 2021; 9:e0135221. [PMID: 34643438 PMCID: PMC8515945 DOI: 10.1128/spectrum.01352-21] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The emerging new lineages of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) have marked a new phase of coronavirus disease 2019 (COVID-19). Understanding the recognition mechanisms of potent neutralizing monoclonal antibodies (NAbs) against the spike protein is pivotal for developing new vaccines and antibody drugs. Here, we isolated several monoclonal antibodies (MAbs) against the SARS-CoV-2 spike protein receptor-binding domain (S-RBD) from the B cell receptor repertoires of a SARS-CoV-2 convalescent. Among these MAbs, the antibody nCoV617 demonstrates the most potent neutralizing activity against authentic SARS-CoV-2 infection, as well as prophylactic and therapeutic efficacies against the human angiotensin-converting enzyme 2 (ACE2) transgenic mouse model in vivo. The crystal structure of S-RBD in complex with nCoV617 reveals that nCoV617 mainly binds to the back of the "ridge" of RBD and shares limited binding residues with ACE2. Under the background of the S-trimer model, it potentially binds to both "up" and "down" conformations of S-RBD. In vitro mutagenesis assays show that mutant residues found in the emerging new lineage B.1.1.7 of SARS-CoV-2 do not affect nCoV617 binding to the S-RBD. These results provide a new human-sourced neutralizing antibody against the S-RBD and assist vaccine development. IMPORTANCE COVID-19 is a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The COVID-19 pandemic has posed a serious threat to global health and the economy, so it is necessary to find safe and effective antibody drugs and treatments. The receptor-binding domain (RBD) in the SARS-CoV-2 spike protein is responsible for binding to the angiotensin-converting enzyme 2 (ACE2) receptor. It contains a variety of dominant neutralizing epitopes and is an important antigen for the development of new coronavirus antibodies. The significance of our research lies in the determination of new epitopes, the discovery of antibodies against RBD, and the evaluation of the antibodies' neutralizing effect. The identified antibodies here may be drug candidates for the development of clinical interventions for SARS-CoV-2.
Collapse
|
243
|
Elevated cytokines and chemokines in peripheral blood of patients with SARS-CoV-2 pneumonia treated with high-titer convalescent plasma. PLoS Pathog 2021; 17:e1010025. [PMID: 34714894 PMCID: PMC8580259 DOI: 10.1371/journal.ppat.1010025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 11/10/2021] [Accepted: 10/11/2021] [Indexed: 12/28/2022] Open
Abstract
The global SARS-CoV-2 coronavirus pandemic continues to be devastating in many areas. Treatment options have been limited and convalescent donor plasma has been used by many centers to transfer passive neutralizing antibodies to patients with respiratory involvement. The results often vary by institution and are complicated by the nature and quality of the donor plasma itself, the timing of administration and the clinical aspects of the recipients. SARS-CoV-2 infection is known to be associated with an increase in the blood concentrations of several inflammatory cytokines/chemokines, as part of the overall immune response to the virus and consequential to mediated lung pathology. Some of these correlates contribute to the cytokine storm syndrome and acute respiratory distress syndrome, often resulting in fatality. A Phase IIa clinical trial at our institution using high neutralizing titer convalescent plasma transfer gave us the unique opportunity to study the elevations of correlates in the first 10 days after infusion. Plasma recipients were divided into hospitalized COVID-19 pneumonia patients who did not (Track 2) or did (Track 3) require mechanical ventilation. Several cytokines were elevated in the patients of each Track and some continued to rise through Day 10, while others initially increased and then subsided. Furthermore, elevations in MIP-1α, MIP-1β and CRP correlated with disease progression of Track 2 recipients. Overall, our observations serve as a foundation for further study of these correlates and the identification of potential biomarkers to improve upon convalescent plasma therapy and to drive more successful patient outcomes. COVID-19, the disease caused by the SARS-CoV-2 virus, has a varied clinical course with limited treatment options. While some patients mount a productive immune response leading to recovery, others progress to rapid respiratory deterioration that may require hospitalization and mechanical ventilation. Our institution conducted a clinical trial to evaluate the efficacy of convalescent plasma therapy (CPT) to treat patients hospitalized with COVID-19 pneumonia. In this arm of the study, we sought to examine immune analytes in donor plasma as well as evaluate the recipients’ plasma before CPT infusion, and at Day 3 and Day 10 post-CPT infusion. We found some analytes to be elevated in plasma donors, compared to healthy controls, even after recovery. Plasma composition in CPT recipients prior to infusion showed elevations in several analytes associated with immune activation. Some significant differences were seen in plasma composition in patients in our Track 2 cohort (hospitalized without mechanical ventilation) compared to the Track 3 cohort (hospitalized with mechanical ventilation). In addition, we obtained plasma samples for hospitalized COVID-19 patients that did not receive CPT and noted several differences in the course of immune analyte production over time compared to the CPT-treated patients.
Collapse
|
244
|
Fouka E, Kalomenidis I, Gianniou N, Gida S, Steiropoulos P. COVID-19 Advanced Care. J Pers Med 2021; 11:1082. [PMID: 34834434 PMCID: PMC8621119 DOI: 10.3390/jpm11111082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/10/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, related to the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a worldwide sudden and substantial burden in public health due to an enormous increase in hospitalizations for pneumonia with the multiorgan disease. Treatment for individuals with COVID-19 includes best practices for supportive management of acute hypoxic respiratory failure. Emerging data indicate that dexamethasone therapy reduces 28-day mortality in patients requiring supplemental oxygen compared with usual care, and ongoing trials are testing the efficacy of antiviral therapies, immune modulators and anticoagulants in the prevention of disease progression and complications, while monoclonal antibodies and hyperimmune globulin may provide additional preventive strategies. Consensus suggestions can standardize care, thereby improving outcomes and facilitating future research. This review discusses current evidence regarding the pharmacotherapy of COVID-19.
Collapse
Affiliation(s)
- Evangelia Fouka
- Pulmonary Department, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, 57010 Thessaloniki, Greece
| | - Ioannis Kalomenidis
- 1st Department of Critical Care and Pulmonary Medicine, Evaggelismos Hospital, National and Kapodistrian University of Athens, 10679 Athens, Greece; (I.K.); (N.G.)
| | - Niki Gianniou
- 1st Department of Critical Care and Pulmonary Medicine, Evaggelismos Hospital, National and Kapodistrian University of Athens, 10679 Athens, Greece; (I.K.); (N.G.)
| | - Sofia Gida
- Intensive Care Unit, General Hospital of Trikala, 42100 Trikala, Greece;
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, 67100 Alexandroupolis, Greece;
| |
Collapse
|
245
|
Abstract
As the coronavirus disease (COVID-19) pandemic led to a global health crisis, there were limited treatment options and no prophylactic therapies for those exposed to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Convalescent plasma is quick to implement, potentially provides benefits, and has a good safety profile. The therapeutic potential of COVID-19 convalescent plasma (CCP) is likely mediated by antibodies through direct viral neutralization and Fc-dependent functions such as a phagocytosis, complement activation, and antibody-dependent cellular cytotoxicity. In the United States, CCP became one of the most common treatments with over half million units transfused despite limited efficacy data. More than a dozen randomized trials now demonstrate that CCP does not provide benefit for those with moderate to severe disease. However, similar to other passive antibody therapies, CCP is beneficial for early disease, when provided to elderly outpatients within 72 hours after symptom onset. Only high-titer CCP should be transfused. CCP should also be considered for immunosuppressed COVID-19 patients. CCP collected in proximity, by time and location, to the patient may be more beneficial due to SARS-CoV-2 variants. Additional randomized trial data are still accruing and should be incorporated with other trial data to optimize CCP indications.
Collapse
|
246
|
Swenson E, Wong LK, Jhaveri P, Weng Y, Kappagoda S, Pandey S, Pritchard A, Rogers A, Ruoss S, Subramanian A, Shan H, Hollenhorst M. Active surveillance of serious adverse events following transfusion of COVID-19 convalescent plasma. Transfusion 2021; 62:28-36. [PMID: 34677830 PMCID: PMC8661846 DOI: 10.1111/trf.16711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/07/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Background The reported incidence of adverse reactions following Coronavirus disease 2019 (COVID‐19) convalescent plasma (CCP) transfusion has generally been lower than expected based on the incidence of transfusion reactions that have been observed in studies of conventional plasma transfusion. This raises the concern for under‐reporting of adverse events in studies of CCP that rely on passive surveillance strategies. Materials and Methods Our institution implemented a protocol to actively identify possible adverse reactions to CCP transfusion. In addition, we retrospectively reviewed the charts of inpatients who received CCP at Stanford Hospital between May 13, 2020 and January 31, 2021. We determined the incidence of adverse events following CCP transfusion. Results A total of 49 patients received CCP. Seven patients (14%) had an increased supplemental oxygen requirement within 4 h of transfusion completion, including one patient who was intubated during the transfusion. An additional 11 patients (total of 18, 37%) had increased oxygen requirements within 24 h of transfusion, including 3 patients who were intubated. Six patients (12%) fulfilled criteria for transfusion‐associated circulatory overload (TACO). Conclusion Using an active surveillance strategy, we commonly observed adverse events following the transfusion of CCP to hospitalized patients. It was not possible to definitively determine whether or not these adverse events are related to CCP transfusion. TACO was likely over‐diagnosed given overlap with the manifestations of COVID‐19. Nevertheless, these results suggest that the potential adverse effects of CCP transfusion may be underestimated by reports from passive surveillance studies.
Collapse
Affiliation(s)
- Erica Swenson
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Lisa Kanata Wong
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Perrin Jhaveri
- Department of Hematology, Kaiser Permanente, Renton, Washington, USA
| | - Yingjie Weng
- Department of Medicine, Quantitative Sciences Unit, Stanford University, Stanford, California, USA
| | - Shanthi Kappagoda
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - Suchitra Pandey
- Department of Pathology, Stanford University, Stanford, California, USA.,Stanford Blood Center, Stanford, California, USA
| | - Angelica Pritchard
- Department of Emergency Medicine, Stanford University, Stanford, California, USA
| | - Angela Rogers
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Stephen Ruoss
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, California, USA
| | - Aruna Subramanian
- Department of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford University, Stanford, California, USA
| | - Hua Shan
- Department of Pathology, Stanford University, Stanford, California, USA
| | - Marie Hollenhorst
- Department of Pathology, Stanford University, Stanford, California, USA.,Department of Medicine, Division of Hematology, Stanford University, Stanford, California, USA.,Chemistry, Engineering, & Medicine for Human Health Institute (ChEM-H), Stanford University, Stanford, California, USA
| |
Collapse
|
247
|
Zhou Y, Xu X, Wei H. Complex Pathophysiological Mechanisms and the Propose of the Three-Dimensional Schedule For Future COVID-19 Treatment. Front Immunol 2021; 12:716940. [PMID: 34745094 PMCID: PMC8564179 DOI: 10.3389/fimmu.2021.716940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 10/05/2021] [Indexed: 11/13/2022] Open
Abstract
At present, the global COVID-19 epidemic is still in a state of anxiety, and increasing the cure rate of critically ill patients is an important means to defeat the virus. From an immune perspective, ARDS driven by an inflammatory storm is still the direct cause of death in severe COVID-19 patients. Although some experience has been gained in the treatment of COVID-19, and intensive COVID-19 vaccination has been carried out recently, it is still effective to save lives to develop more effective programs to alleviate the inflammatory storm and ARDS in patients with SARS-CoV-2 or emerging variants of SARS-CoV-2. In reorganizing the ARDS-related inflammatory storm formation program in COVID-19 patients, we highlighted the importance of the vicious circle of inflammatory cytokines and inflammatory cell death, which is aggravated by blood circulation to form multi-system inflammation. Summarizes the interlocking and crisscrossing of inflammatory response and inflammatory cell death mechanisms including NETs, pyrolysis, apoptosis and PANoptosis in severe COVID-19. More importantly, in response to the inflammatory storm formation program we described, and on the premise of following ethical and clinical experimental norms, we propose a three-dimensional integrated program for future research based on boosting antiviral immune response at the initial stage, inhibiting inflammatory cytokine signaling at the exacerbation stage and inhibiting cell death before it's worse to prevent and alleviate ARDS.
Collapse
Affiliation(s)
- Yonggang Zhou
- Institute of Gerontology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiuxiu Xu
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- Institute of Gerontology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Hefei National Laboratory for Physical Sciences at Microscale, The Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| |
Collapse
|
248
|
Romon I, Dominguez-Garcia JJ, Arroyo JL, Suberviola B, Cabezón I, Abascal B, Baldeón C, Cuesta A, Portilla R, Casuso E, Ocio E, Briz M. Convalescent plasma treatment for patients of 80 years and older with COVID-19 pneumonia. BMC Geriatr 2021; 21:566. [PMID: 34663227 PMCID: PMC8521266 DOI: 10.1186/s12877-021-02447-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Older patients, frequently with multiple comorbidities, have a high mortality from COVID-19 infection. Convalescent plasma (CP) is a therapeutic option for these patients. Our objective is to retrospectively evaluate the efficacy and adverse events of CP treatment in this population group. METHODS Forty one patients over 80 years old with COVID-19 pneumonia received CP added to standard treatment, 51.2% with high anti-SARS-CoV-2 IgG titers and 48.8% with low titers. Median time between the onset of symptoms and the infusion of plasma was 7 days (IQR 4-10). A similar group of 82 patients who received only standard treatment, during a period in which CP was not available, were selected as a control group. RESULTS In-hospital mortality was 26.8% for controls and 14.6% for CP patients (P = 0.131) and ICU admission was 8.5% for controls and 4.9% for CP patients (P = 0.467). Mortality tended to be lower in the high-titer group (9.5%) than in the low-titer group (20%), and in patients transfused within the first 7 days of symptom onset (10%) than in patients transfused later (19.1%), although the differences were not statistically significant (P = 0.307 and P = 0.355 respectively). There was no difference in the length of hospitalization. No significant adverse events were associated with CP treatment. CONCLUSIONS Convalescent plasma treatment in patients over 80 years old with COVID-19 pneumonia was well tolerated but did not present a statistically significant difference in hospital mortality, ICU admission, or length of hospitalization. The results should be interpreted with caution as only half the patients received high-titer CP and the small number of patients included in the study limits the statistical power to detect significant differences. TRIAL REGISTRATION CEIm Cantabria # 2020.127.
Collapse
Affiliation(s)
- Iñigo Romon
- Hematology and Hemotherapy Service, Hospital Universitario Marqués de Valdecilla, Avenida Valdecilla, s/n, 39008, Santander, Spain.
| | - Juan J Dominguez-Garcia
- Hematology and Hemotherapy Service, Hospital Universitario Marqués de Valdecilla, Avenida Valdecilla, s/n, 39008, Santander, Spain
| | - Jose L Arroyo
- Banco de Sangre y Tejidos de Cantabria, 39121, Liencres, Spain
| | - Borja Suberviola
- Intensive Care Service, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Itxasne Cabezón
- Infectious Diseases Service, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Beatriz Abascal
- Pneumology Service, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Cristina Baldeón
- Internal Medicine Service, Hospital Universitario Marqués de Valdecilla, 39008, Santander, Spain
| | - Amalia Cuesta
- Haematology Service, Hospital de Sierrallana, 39300, Torrelavega, Spain
| | - Raquel Portilla
- Internal Medicine Service, Hospital de Sierrallana, 39300, Torrelavega, Spain
| | - Elena Casuso
- Internal Medicine Service, Hospital de Laredo, 39770, Laredo, Spain
| | - Enrique Ocio
- Hematology and Hemotherapy Service, Hospital Universitario Marqués de Valdecilla, Avenida Valdecilla, s/n, 39008, Santander, Spain
| | - Montserrat Briz
- Hematology and Hemotherapy Service, Hospital Universitario Marqués de Valdecilla, Avenida Valdecilla, s/n, 39008, Santander, Spain
| |
Collapse
|
249
|
Körper S, Weiss M, Zickler D, Wiesmann T, Zacharowski K, Corman VM, Grüner B, Ernst L, Spieth P, Lepper PM, Bentz M, Zinn S, Paul G, Kalbhenn J, Dollinger MM, Rosenberger P, Kirschning T, Thiele T, Appl T, Mayer B, Schmidt M, Drosten C, Wulf H, Kruse JM, Jungwirth B, Seifried E, Schrezenmeier H. Results of the CAPSID randomized trial for high-dose convalescent plasma in patients with severe COVID-19. J Clin Invest 2021; 131:e152264. [PMID: 34464358 DOI: 10.1172/jci152264] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUNDCOVID-19 convalescent plasma (CCP) has been considered a treatment option for COVID-19. This trial assessed the efficacy of a neutralizing antibody containing high-dose CCP in hospitalized adults with COVID-19 requiring respiratory support or intensive care treatment.METHODSPatients (n = 105) were randomized 1:1 to either receive standard treatment and 3 units of CCP or standard treatment alone. Control group patients with progress on day 14 could cross over to the CCP group. The primary outcome was a dichotomous composite outcome of survival and no longer fulfilling criteria for severe COVID-19 on day 21.ResultsThe primary outcome occurred in 43.4% of patients in the CCP group and 32.7% in the control group (P = 0.32). The median time to clinical improvement was 26 days in the CCP group and 66 days in the control group (P = 0.27). The median time to discharge from the hospital was 31 days in the CCP group and 51 days in the control group (P = 0.24). In the subgroup that received a higher cumulative amount of neutralizing antibodies, the primary outcome occurred in 56.0% of the patients (vs. 32.1%), with significantly shorter intervals to clinical improvement (20 vs. 66 days, P < 0.05) and to hospital discharge (21 vs. 51 days, P = 0.03) and better survival (day-60 probability of survival 91.6% vs. 68.1%, P = 0.02) in comparison with the control group.ConclusionCCP added to standard treatment was not associated with a significant improvement in the primary and secondary outcomes. A predefined subgroup analysis showed a significant benefit of CCP among patients who received a larger amount of neutralizing antibodies.Trial registrationClinicalTrials.gov NCT04433910.FundingBundesministerium für Gesundheit (German Federal Ministry of Health): ZMVI1-2520COR802.
Collapse
Affiliation(s)
- Sixten Körper
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Manfred Weiss
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Wiesmann
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Kai Zacharowski
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Beate Grüner
- Division of Infectious Diseases, University Hospital and Medical Center Ulm, Ulm, Germany
| | - Lucas Ernst
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Peter Spieth
- Department of Anesthesiology and Critical Care Medicine, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Philipp M Lepper
- Department of Internal Medicine V - Pneumology, Allergology, Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Martin Bentz
- Department of Internal Medicine III, Hospital of Karlsruhe, Karlsruhe, Germany
| | - Sebastian Zinn
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Gregor Paul
- Department of Gastroenterology, Hepatology, Pneumology and Infectious Diseases, Klinikum Stuttgart, Stuttgart, Germany
| | - Johannes Kalbhenn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Kirschning
- Department of Anaesthesiology and Surgical Intensive Care Medicine, University of Heidelberg, University Medical Centre Mannheim, Mannheim, Germany
| | - Thomas Thiele
- Institute of Immunology and Transfusion Medicine, University Hospital Greifswald, Greifswald, Germany
| | - Thomas Appl
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Hinnerk Wulf
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Jan Matthias Kruse
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bettina Jungwirth
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | | |
Collapse
|
250
|
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: 10.3] [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
|