1
|
Dragotakes Q, Johnson PW, Buras MR, Carter RE, Joyner MJ, Bloch E, Gebo KA, Hanley DF, Henderson JP, Pirofski LA, Shoham S, Senefeld JW, Tobian AA, Wiggins CC, Wright RS, Paneth NS, Sullivan DJ, Casadevall A. Estimates of actual and potential lives saved in the United States from the use of COVID-19 convalescent plasma. Proc Natl Acad Sci U S A 2024; 121:e2414957121. [PMID: 39352932 PMCID: PMC11474081 DOI: 10.1073/pnas.2414957121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 08/23/2024] [Indexed: 10/04/2024] Open
Abstract
In the Spring of 2020, the United States of America (USA) deployed COVID-19 convalescent plasma (CCP) to treat hospitalized patients. Over 500,000 patients were treated with CCP during the first year of the pandemic. In this study, we estimated the number of actual inpatient lives saved by CCP treatment in the United States of America based on CCP weekly use, weekly national mortality data, and CCP mortality reduction data from meta-analyses of randomized controlled trials and real-world data. We also estimate the potential number of lives saved if CCP had been deployed for 100% of hospitalized patients or used in 15 to 75% of outpatients. Depending on the assumptions modeled in stratified analyses, we estimated that CCP saved between 16,476 and 66,296 lives. The CCP ideal use might have saved as many as 234,869 lives and prevented 1,136,133 hospitalizations. CCP deployment was a successful strategy for ameliorating the impact of the COVID-19 pandemic in the USA. This experience has important implications for convalescent plasma use in future infectious disease emergencies.
Collapse
Affiliation(s)
- Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Patrick W. Johnson
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Jacksonville, FL32224
| | - Matthew R. Buras
- Division of Biostatistics and Clinical Trials, Department of Quantitative Health Sciences, Scottsdale, AZ85259
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL32224
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN55905
| | - Evan Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Kelly A. Gebo
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniel F. Hanley
- Department of Neurology, Brain Injury Outcomes Division, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jeffrey P. Henderson
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Louis, St. Louis, MO63110
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine, New York, NY10461
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Jonathon W. Senefeld
- Department of Health and Kinesiology, University of Illinois Urbana-Champaign, Urbana, IL61801
| | - Aaron A.R. Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Chad C. Wiggins
- Department of Kinesiology, Michigan State University, East Lansing, MI48823
| | - R. Scott Wright
- Departments of Cardiovascular Medicine and Human Research Protection Program, Mayo Clinic, Rochester, MN55905
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, MI48823
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI48823
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD21205
| |
Collapse
|
2
|
Carroll TD, Wong T, Morris MK, Di Germanio C, Ma ZM, Stone M, Ball E, Fritts L, Rustagi A, Simmons G, Busch M, Miller CJ. Vaccine-Boosted CCP Decreases Virus Replication and Hastens Resolution of Infection Despite Transiently Enhancing Disease in SARS-CoV-2-Infected Hamsters. J Infect Dis 2024; 229:1702-1710. [PMID: 38213276 PMCID: PMC11175670 DOI: 10.1093/infdis/jiad568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 12/05/2023] [Indexed: 01/13/2024] Open
Abstract
Definitive data demonstrating the utility of coronavirus disease 2019 (COVID-19) convalescent plasma (CCP) for treating immunocompromised patients remains elusive. To better understand the mechanism of action of CCP, we studied viral replication and disease progression in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-infected hamsters treated with CCP obtained from recovered COVID-19 patients that were also vaccinated with an mRNA vaccine, hereafter referred to as Vaxplas. Vaxplas transiently enhanced disease severity and lung pathology in hamsters treated near peak viral replication due to immune complex and activated complement deposition in pulmonary endothelium, and recruitment of M1 proinflammatory macrophages into the lung parenchyma. However, aside from one report, transient enhanced disease has not been reported in CCP recipient patients, and the transient enhanced disease in Vaxplas hamsters may have been due to mismatched species IgG-FcR interactions, infusion timing, or other experimental factors. Despite transient disease enhancement, Vaxplas dramatically reduced virus replication in lungs and improved infection outcome in SARS-CoV-2-infected hamsters.
Collapse
Affiliation(s)
- Timothy D Carroll
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Talia Wong
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Mary Kate Morris
- Division of Viral and Rickettsial Diseases, California Department of Public Health, Richmond, California, USA
| | | | - Zhong-min Ma
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA
| | - Erin Ball
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Linda Fritts
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Arjun Rustagi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, USA
| | - Michael Busch
- Vitalant Research Institute, San Francisco, California, USA
| | - Christopher J Miller
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California, USA
| |
Collapse
|
3
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Streblow DN, Hirsch AJ, Stanton JJ, Lewis AD, Colgin L, Hessell AJ, Kreklywich CN, Smith JL, Sutton WF, Chauvin D, Woo J, Bimber BN, LeBlanc CN, Acharya SN, O'Roak BJ, Sardar H, Sajadi MM, Tehrani ZR, Walter MR, Martinez-Sobrido L, Kobie JJ, Reader RJ, Olstad KJ, Hobbs TR, Saphire EO, Schendel SL, Carnahan RH, Knoch J, Branco LM, Crowe JE, Van Rompay KKA, Lovalenti P, Vu Truong, Forthal DN, Haigwood NL. Aerosol delivery of SARS-CoV-2 human monoclonal antibodies in macaques limits viral replication and lung pathology. Nat Commun 2023; 14:7062. [PMID: 37923717 PMCID: PMC10624670 DOI: 10.1038/s41467-023-42440-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 10/11/2023] [Indexed: 11/06/2023] Open
Abstract
Passively administered monoclonal antibodies (mAbs) given before or after viral infection can prevent or blunt disease. Here, we examine the efficacy of aerosol mAb delivery to prevent infection and disease in rhesus macaques inoculated with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Delta variant via intranasal and intratracheal routes. SARS-CoV-2 human mAbs or a human mAb directed to respiratory syncytial virus (RSV) are nebulized and delivered using positive airflow via facemask to sedated macaques pre- and post-infection. Nebulized human mAbs are detectable in nasal, oropharyngeal, and bronchoalveolar lavage (BAL) samples. SARS-CoV-2 mAb treatment significantly reduces levels of SARS-CoV-2 viral RNA and infectious virus in the upper and lower respiratory tracts relative to controls. Reductions in lung and BAL virus levels correspond to reduced BAL inflammatory cytokines and lung pathology. Aerosolized antibody therapy for SARS-CoV-2 could be effective for reducing viral burden and limiting disease severity.
Collapse
Affiliation(s)
- Daniel N Streblow
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Alec J Hirsch
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Jeffrey J Stanton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Anne D Lewis
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Lois Colgin
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Ann J Hessell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Craig N Kreklywich
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - Jessica L Smith
- Vaccine & Gene Therapy Institute, Oregon Health & Science University, Beaverton, OR, USA
| | - William F Sutton
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | | | | | - Benjamin N Bimber
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Cierra N LeBlanc
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Sonia N Acharya
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Brian J O'Roak
- Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, OR, USA
| | - Harjinder Sardar
- Environmental Health & Safety, Oregon Health & Science University, Portland, OR, USA
| | - Mohammad M Sajadi
- Baltimore VA Medical Center, VA Maryland Health Care System, Baltimore, MD, USA
| | - Zahra R Tehrani
- Division of Clinical Care and Research, Institute of Human Virology, University of Maryland, Baltimore, MD, USA
| | - Mark R Walter
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - James J Kobie
- Department of Medicine, Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rachel J Reader
- California National Primate Research Center, University of California, Davis, CA, USA
| | - Katherine J Olstad
- California National Primate Research Center, University of California, Davis, CA, USA
| | - Theodore R Hobbs
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Sharon L Schendel
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | | | | | - James E Crowe
- Vanderbilt University Medical Center, Nashville, TN, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, CA, USA
| | | | - Vu Truong
- Aridis Pharmaceuticals, Los Gatos, CA, USA.
| | - Donald N Forthal
- University of California, Irvine, School of Medicine, Irvine, CA, USA.
| | - Nancy L Haigwood
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, USA.
| |
Collapse
|
5
|
Carroll TD, Wong T, Morris MK, Di Germanio C, Ma ZM, Stone M, Ball E, Fritts L, Rustagi A, Simmons G, Busch M, Miller CJ. Administration of vaccine-boosted COVID-19 convalescent plasma to SARS-CoV-2 infected hamsters decreases virus replication in lungs and hastens resolution of the infection despite transiently enhancing disease and lung pathology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.22.553458. [PMID: 37662344 PMCID: PMC10473650 DOI: 10.1101/2023.08.22.553458] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The utility of COVID-19 convalescent plasma (CCP) for treatment of immunocompromised patients who are not able to mount a protective antibody response against SARS-CoV-2 and who have contraindications or adverse effects from currently available antivirals remains unclear. To better understand the mechanism of protection in CCP, we studied viral replication and disease progression in SARS-CoV-2 infected hamsters treated with CCP plasma obtained from recovered COVID patients that had also been vaccinated with an mRNA vaccine, hereafter referred to as Vaxplas. We found that Vaxplas dramatically reduced virus replication in the lungs and improved infection outcome in SARS-CoV-2 infected hamsters. However, we also found that Vaxplas transiently enhanced disease severity and lung pathology in treated animals likely due to the deposition of immune complexes, activation of complement and recruitment of increased numbers of macrophages with an M1 proinflammatory phenotype into the lung parenchyma.
Collapse
Affiliation(s)
- Timothy D. Carroll
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Talia Wong
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Mary Kate Morris
- Division of Viral and Rickettsial Diseases, California Department of Public Health, Richmond, California, USA
| | | | - Zhong-min Ma
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, CA, USA
| | - Erin Ball
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
| | - Linda Fritts
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
| | - Arjun Rustagi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Palo Alto, California, USA
| | | | | | - Christopher J. Miller
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California Davis, Davis, California, USA
- California National Primate Research Center, University of California Davis, Davis, California, USA
- Division of Infectious Diseases, Department of Internal Medicine, School of Medicine, University of California Davis, Sacramento, California, USA
| |
Collapse
|
6
|
Saito S, Kutsuna S, Akifumi I, Hase R, Oda R, Terada J, Shimizu Y, Uemura Y, Takamatsu Y, Yasuhara A, Shiratori K, Satake M, Sakamoto N, Miyazaki Y, Shimizu H, Togano T, Matsunaga A, Okuma K, Hamaguchi I, Fujisawa K, Nagashima M, Ashida S, Terada M, Kimura A, Morioka S, Matsubayashi K, Tsuno NH, Kojima M, Kuramitsu M, Tezuka K, Ikebe E, Ishizaka Y, Kenji M, Hangaishi A, Mikami A, Sugiura W, Ohmagari N, Mitsuya H. Efficacy of convalescent plasma therapy for COVID-19 in Japan: An open-label, randomized, controlled trial. J Infect Chemother 2023:S1341-321X(23)00122-8. [PMID: 37178973 PMCID: PMC10174342 DOI: 10.1016/j.jiac.2023.05.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 04/13/2023] [Accepted: 05/10/2023] [Indexed: 05/15/2023]
Abstract
BACKGROUND Convalescent plasma is a potential therapeutic option for patients with coronavirus disease 2019 (COVID-19). Despite its use for treating several viral infections, we lack comprehensive data on its efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS We conducted a multicenter, open-label, randomized controlled trial of convalescent plasma therapy with high neutralizing activity against SARS-CoV-2 in high-risk patients within five days after the onset of COVID-19 symptoms. The primary endpoint was the time-weighted average change in the SARS-CoV-2 viral load in nasopharyngeal swabs from days 0-5. RESULTS Between February 24, 2021, and November 30, 2021, 25 patients were randomly assigned to either convalescent plasma (n = 14) or standard of care (n = 11) groups. Four patients discontinued their allocated convalescent plasma, and 21 were included in the modified intention-to-treat analysis. The median interval between the symptom onset and plasma administration was 4.5 days (interquartile range, 3-5 days). The primary outcome of the time-weighted average change in the SARS-CoV-2 viral load in nasopharyngeal swabs did not significantly differ between days 0-5 (1.2 log10 copies/mL in the convalescent plasma vs. 1.2 log10 copies/mL in the standard of care (effect estimate, 0.0 [95% confidence interval, -0.8-0.7]; P = 0.94)). No deaths were observed in either group. CONCLUSIONS The early administration of convalescent plasma with high neutralizing activity did not contribute to a decrease in the viral load within five days compared with the standard of care alone.
Collapse
Affiliation(s)
- Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Satoshi Kutsuna
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; Department of Infection Control, Graduate School of Medicine Faculty of Medicine, Osaka University, Japan
| | - Imamura Akifumi
- Department of Infectious Diseases, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Ryota Hase
- Department of Infectious Diseases, Japanese Red Cross Narita Hospital, Chiba, Japan
| | - Rentaro Oda
- Division of Infectious Diseases, Tokyo Bay Urayasu Ichikawa Medical Center, Chiba, Japan
| | - Junko Terada
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yosuke Shimizu
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukari Uemura
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuki Takamatsu
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Akemi Yasuhara
- Joint Center for Researchers, Associates and Clinicians Data Center, Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Katsuyuki Shiratori
- Clinical Laboratory Department, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Naoya Sakamoto
- Department of Infectious Diseases, Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hidefumi Shimizu
- Department of Respiratory Medicine, JCHO Tokyo Shinjuku Medical Center, Tokyo, Japan
| | - Tomiteru Togano
- Department of Hematology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akihiro Matsunaga
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kazu Okuma
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan; Department of Microbiology, Faculty of Medicine, Kansai Medical University, Osaka, Japan
| | - Isao Hamaguchi
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kyoko Fujisawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Maki Nagashima
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinobu Ashida
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mari Terada
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Akiko Kimura
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinichiro Morioka
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | | | | | - Makiko Kojima
- Japanese Red Cross Central Blood Institute, Tokyo, Japan; Japanese Red Cross Kanto-Koshin-etsu Block Blood Center, Tokyo, Japan
| | - Madoka Kuramitsu
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kenta Tezuka
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Emi Ikebe
- Department of Safety Research on Blood and Biological Products, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yukihito Ishizaka
- Department of Intractable Diseases, National Center for Global Health and Medicine, Tokyo, Japan
| | - Maeda Kenji
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Akira Hangaishi
- Department of Hematology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ayako Mikami
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Wataru Sugiura
- Center for Clinical Science, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| |
Collapse
|
7
|
Casadevall A, Joyner MJ, Pirofski LA, Senefeld JW, Shoham S, Sullivan D, Paneth N, Focosi D. Convalescent plasma therapy in COVID-19: Unravelling the data using the principles of antibody therapy. Expert Rev Respir Med 2023:1-15. [PMID: 37129285 DOI: 10.1080/17476348.2023.2208349] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
INTRODUCTION When the COVID-19 pandemic struck no specific therapies were available and many turned to COVID-19 convalescent plasma (CCP), a form of antibody therapy. The literature provides mixed evidence for CCP efficacy. AREAS COVERED PubMed was searched using the words COVID-19 and convalescent plasma and individual study designs were evaluated for adherence to the three principles of antibody therapy, i.e. that plasma 1) contain specific antibody; 2) have enough specific antibody to mediate a biological effect; and 3) be administered early in the course of disease. Using this approach, a diverse and seemingly contradictory collection of clinical findings was distilled into a consistent picture whereby CCP was effective when used according to the above principles of antibody therapy. In addition, CCP therapy in immunocompromised patients is useful at any time in the course of disease. EXPERT OPINION CCP is safe and effective when used appropriately. Today, most of humanity has some immunity to SARS-CoV-2 from vaccines and infection, which has lessened the need for CCP in the general population. However, COVID-19 in immunocompromised patients is a major therapeutic challenge, and with the deauthorization of all SARS-CoV-2-spike protein-directed monoclonal antibodies, CCP is the only antibody therapy available for this population.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nigel Paneth
- Departments of Epidemiology & Biostatistics and Pediatrics & Human Development, Michigan State University, East Lansing, MI, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
8
|
Milligan EC, Olstad K, Williams CA, Mallory M, Cano P, Cross KA, Munt JE, Garrido C, Lindesmith L, Watanabe J, Usachenko JL, Hopkins L, Immareddy R, Shaan Lakshmanappa Y, Elizaldi SR, Roh JW, Sammak RL, Pollard RE, Yee JL, Herbek S, Scobey T, Miehlke D, Fouda G, Ferrari G, Gao H, Shen X, Kozlowski PA, Montefiori D, Hudgens MG, Edwards DK, Carfi A, Corbett KS, Graham BS, Fox CB, Tomai M, Iyer SS, Baric R, Reader R, Dittmer DP, Van Rompay KKA, Permar SR, De Paris K. Infant rhesus macaques immunized against SARS-CoV-2 are protected against heterologous virus challenge 1 year later. Sci Transl Med 2023; 15:eadd6383. [PMID: 36454813 PMCID: PMC9765459 DOI: 10.1126/scitranslmed.add6383] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
The U.S. Food and Drug Administration only gave emergency use authorization of the BNT162b2 and mRNA-1273 SARS-CoV-2 vaccines for infants 6 months and older in June 2022. Yet questions regarding the durability of vaccine efficacy, especially against emerging variants, in this age group remain. We demonstrated previously that a two-dose regimen of stabilized prefusion Washington SARS-CoV-2 S-2P spike (S) protein encoded by mRNA encapsulated in lipid nanoparticles (mRNA-LNP) or purified S-2P mixed with 3M-052, a synthetic Toll-like receptor (TLR) 7/8 agonist, in a squalene emulsion (Protein+3M-052-SE) was safe and immunogenic in infant rhesus macaques. Here, we demonstrate that broadly neutralizing and spike-binding antibodies against variants of concern (VOCs), as well as T cell responses, persisted for 12 months. At 1 year, corresponding to human toddler age, we challenged vaccinated rhesus macaques and age-matched nonvaccinated controls intranasally and intratracheally with a high dose of heterologous SARS-CoV-2 B.1.617.2 (Delta). Seven of eight control rhesus macaques exhibited severe interstitial pneumonia and high virus replication in the upper and lower respiratory tract. In contrast, vaccinated rhesus macaques had faster viral clearance with mild to no pneumonia. Neutralizing and binding antibody responses to the B.1.617.2 variant at the day of challenge correlated with lung pathology and reduced virus replication. Overall, the Protein+3M-052-SE vaccine provided superior protection to the mRNA-LNP vaccine, emphasizing opportunities for optimization of current vaccine platforms. The observed efficacy of both vaccines 1 year after vaccination supports the implementation of an early-life SARS-CoV-2 vaccine.
Collapse
Affiliation(s)
- Emma C Milligan
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Katherine Olstad
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Caitlin A Williams
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Michael Mallory
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patricio Cano
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaitlyn A Cross
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer E Munt
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Carolina Garrido
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Lisa Lindesmith
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jennifer Watanabe
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Jodie L Usachenko
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Lincoln Hopkins
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Ramya Immareddy
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | | | - Sonny R Elizaldi
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Jamin W Roh
- Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA.,Graduate Group in Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Rebecca L Sammak
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Rachel E Pollard
- School of Veterinary Medicine, University of California at Davis, Davis, CA 95616, USA
| | - JoAnn L Yee
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Savannah Herbek
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Trevor Scobey
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Dieter Miehlke
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Genevieve Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Guido Ferrari
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC 27710, USA.,Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA.,Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Hongmei Gao
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela A Kozlowski
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David Montefiori
- Department of Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Michael G Hudgens
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | | - Kizzmekia S Corbett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20852, USA
| | - Christopher B Fox
- Access to Advanced Health Institute, Seattle, WA 98102, USA.,Department of Global Health, University of Washington, Seattle, WA 98105, USA
| | - Mark Tomai
- 3M Corporate Research Materials Laboratory, Saint Paul, MN 55144, USA
| | - Smita S Iyer
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Center for Immunology and Infectious Diseases, University of California at Davis, Davis, CA 95616, USA
| | - Ralph Baric
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Rachel Reader
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA
| | - Dirk P Dittmer
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Koen K A Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, CA 95616, USA.,Department of Pathology, Microbiology and Immunology, University of California at Davis, Davis, CA 95616, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medical College, New York, NY 10065, USA
| | - Kristina De Paris
- Department of Microbiology and Immunology, Children's Research Institute, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|