1
|
Tan SSX, Tan TT. Convalescent plasma: is there still a role in the treatment of COVID-19? Singapore Med J 2025:00077293-990000000-00190. [PMID: 40205748 DOI: 10.4103/singaporemedj.smj-2024-197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/01/2024] [Indexed: 04/11/2025]
|
2
|
Fekrvand S, Saleki K, Abolhassani H, Almasi-Hashiani A, Hakimelahi A, Zargarzadeh N, Yekaninejad MS, Rezaei N. COVID-19 infection in inborn errors of immunity and their phenocopies: a systematic review and meta-analysis. Infect Dis (Lond) 2025:1-35. [PMID: 40178994 DOI: 10.1080/23744235.2025.2483339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 02/09/2025] [Accepted: 02/23/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Inborn errors of immunity (IEI) are congenital disorders of the immune system. Due to impaired immune system, they are at a higher risk to develop a more severe COVID-19 course compared to general population. OBJECTIVES Herein, we aimed to systematically review various aspects of IEI patients infected with SARS-CoV-2. Moreover, we performed a meta-analysis to determine the frequency of COVID-19 in patients with different IEI. METHODS Embase, Web of Science, PubMed, and Scopus were searched introducing terms related to IEI and COVID-19. RESULTS 3646 IEI cases with a history of COVID-19 infection were enrolled. The majority of patients had critical infections (1013 cases, 27.8%). The highest frequency of critical and severe cases was observed in phenocopies of IEI (95.2%), defects in intrinsic and innate immunity (69.4%) and immune dysregulation (23.9%). 446 cases (12.2%) succumbed to the disease and the highest mortality was observed in IEI phenocopies (34.6%). COVID-19 frequency in immunodeficient patients was 11.9% (95% CI: 8.3 to 15.5%) with innate immunodeficiency having the highest COVID-19 frequency [34.1% (12.1 to 56.0%)]. COVID-19 case fatality rate among IEI patients was estimated as 5.4% (95% CI: 3.5-8.3%, n = 8 studies, I2 = 17.5%). CONCLUSION IEI with underlying defects in specific branches of the immune system responding to RNA virus infection experience a higher frequency and mortality of COVID-19 infection. Increasing awareness about these entities and underlying genetic defects, adherence to prophylactic strategies and allocating more clinical attention to these patients could lead to a decrease in COVID-19 frequency and mortality in these patients.
Collapse
Affiliation(s)
- Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Kiarash Saleki
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Division of Clinical Immunology, Department of Biosciences and Nutrition, KarolinskaInstitutet, Karolinska University Hospital, Stockholm, Sweden
| | - Amir Almasi-Hashiani
- Department of Epidemiology, School of Health, Arak University of Medical Sciences, Arak, Iran
| | - Ali Hakimelahi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nikan Zargarzadeh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mir Saeed Yekaninejad
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Primary Immunodeficiency Diseases Network (PIDNet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
3
|
Hoffmann S, Schrezenmeier E, Desmarets M, Halleck F, Durrbach A, Peters L, Tremmel AT, Seidel A, Führer M, Bachmann F, Schrezenmeier J, Greiner J, Körper S, Hofmann H, Ludwig C, Vieweg C, Jahrsdörfer B, Budde K, Schmidt M, Münch J, Joher N, Daguindau E, Grüner B, Brunotte G, Vauchy C, Seifried E, Bradshaw D, Estcourt LJ, Roberts DJ, Toussirot E, Rijnders B, Tiberghien P, Schrezenmeier H. Early, very high-titre convalescent plasma therapy in clinically vulnerable individuals with mild COVID-19: an international, randomised, open-label trial. EBioMedicine 2025; 113:105613. [PMID: 40020259 PMCID: PMC11919330 DOI: 10.1016/j.ebiom.2025.105613] [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: 11/15/2024] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) is a treatment option for COVID-19. This study investigated the safety and efficacy of early, very high-titre CCP in immunocompromised individuals with mild COVID-19. METHODS This randomised, controlled, open-label trial assessed CCP in immunocompromised patients (n = 120) with mild COVID-19 in 10 clinical trial centres across Germany, France, and the Netherlands. Patients were randomised 1:1 to receive either standard of care (SoC) alone (SoC group) or SoC and 2 units of CCP. Most patients (89.7%) had received ≥3 SARS-CoV-2 vaccinations. The primary endpoint was hospitalisation for progressive COVID-19 symptoms or death by day 28 after randomisation, analysed on a modified intention-to-treat basis (117 patients). The safety analysis included the full analysis set. The trial is registered with EudraCT 2021-006621-22, and ClinicalTrials.gov, NCT05271929. FINDINGS Between April 11, 2022 and November 27, 2023, 120 patients were enrolled. Patients in the CCP group received a median of 559 ml CCP from convalescent, vaccinated donors with very high levels of SARS-CoV-2 antibodies (median 81,810 IU/ml) at a median 4 days after symptom onset. The primary outcome occurred in 5/58 patients (8.6%) in the SoC group and in 0/59 patients (0%) in the CCP group, difference -8.6% (95% confidence interval of difference -19% to -0.80%; p-value 0.027; Fisher's exact test). The course of SARS-CoV-2 antibodies in the patients demonstrated a passive transfer of antibodies by the CCP, in particular neutralising effects against new SARS-CoV-2 variants. Whole genome sequencing of SARS-CoV-2 in patients during follow-up showed significant intra-host viral evolution, but without differences between groups. CCP was well tolerated. INTERPRETATION Early administration of high-titre CCP can prevent hospitalisation or death in immunocompromised patients with mild COVID-19. FUNDING Support-e project (European Union's Horizon 2020 Programme), German Federal Ministry of Education and Research, ZonMw, the Netherlands Organisation for Health Research and Development.
Collapse
Affiliation(s)
- Simone Hoffmann
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Maxime Desmarets
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Antoine Durrbach
- Department of Nephrology, AP-HP Hôpital Henri Mondor, Créteil, Île-de-France, France; INSERM UMR1186, Universite Paris Saclay, France
| | - Lynn Peters
- Division of Infectious Diseases, Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | | | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Centre, Ulm, Germany
| | - Marita Führer
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Friederike Bachmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jens Schrezenmeier
- Division of Haematology, Oncology, and Cancer Immunology, Medical Department, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jochen Greiner
- Department of Internal Medicine, Diakonie Hospital Stuttgart, Stuttgart, Germany
| | - Sixten Körper
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Henrike Hofmann
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Carolin Ludwig
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Christiane Vieweg
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen, Frankfurt, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Centre, Ulm, Germany
| | - Nizar Joher
- Department of Nephrology, AP-HP Hôpital Henri Mondor, Créteil, Île-de-France, France
| | - Etienne Daguindau
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; Haematology Department, CHU Besançon, Besançon, France
| | - Beate Grüner
- Division of Infectious Diseases, Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| | - Gaëlle Brunotte
- CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Charline Vauchy
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg - Hessen, Frankfurt, Germany
| | - Daniel Bradshaw
- Virus Reference Department, UK Health Security Agency, London, UK
| | - Lise J Estcourt
- NHS Blood and Transplant, Oxford, Oxfordshire, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - David J Roberts
- NHS Blood and Transplant, Oxford, Oxfordshire, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK
| | - Eric Toussirot
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; CHU Besançon, Inserm, Centre d'Investigation Clinique (CIC 1431), Besançon, France
| | - Bart Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Pierre Tiberghien
- Université Marie et Louis Pasteur, EFS, Inserm, RIGHT (UMR 1098), Besançon, France; Etablissement Francais du Sang, La Plaine Saint-Denis, Île-de-France, France
| | - Hubert Schrezenmeier
- Institute of Clinical Transfusion Medicine and Immunogenetics, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, University of Ulm, Ulm, Germany.
| |
Collapse
|
4
|
Ana-Sosa-Batiz F, Verma SK, Shafee N, Miller R, Conner C, Hastie KM, Timis J, Maule E, Nguyen MN, Tran L, Varghese K, Madany H, Street AE, Zandonatti M, Moi ML, Jarnagin K, Webb DR, Saphire EO, Kim K, Shresta S. A humanised ACE2, TMPRSS2, and FCGRT mouse model reveals the protective efficacy of anti-receptor binding domain antibodies elicited by SARS-CoV-2 hybrid immunity. EBioMedicine 2025; 113:105619. [PMID: 40020261 PMCID: PMC11910679 DOI: 10.1016/j.ebiom.2025.105619] [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: 01/26/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Despite the importance of vaccination- and infection-elicited antibodies (Abs) to SARS-CoV-2 immunity, current mouse models do not fully capture the dynamics of Ab-mediated immunity in vivo, including potential contributions of the neonatal Fc receptor, encoded by FCGRT. METHODS We generated triple knock-in (TKI) mice expressing human ACE2, TMPRSS2, and FCGRT; and evaluated the protective efficacy of anti-SARS-CoV-2 monoclonal Abs (mAbs) and plasma from individuals with immunity elicited by vaccination alone plus SARS-CoV-2 infection-induced (hybrid) immunity. FINDINGS A human anti-SARS-CoV-2 mAb harbouring a half-life-extending mutation, but not the wild-type mAb, exhibited prolonged half-life in TKI mice and protected against lung infection with Omicron BA.2, validating the utility of these mice for evaluating therapeutic Abs. Pooled plasma from individuals with hybrid immunity to Delta, but not from vaccinated-only individuals, cleared infectious Delta from the lungs of TKI mice (P < 0.01), even though the two plasma pools had similar Delta-binding and -neutralising Ab titres in vitro. Similarly, plasma from individuals with hybrid Omicron BA.1/2 immunity, but not hybrid Delta immunity, decreased lung infection (P < 0.05) with BA.5 in TKI mice, despite the plasma pools having comparable BA.5-binding and -neutralising titres in vitro. Depletion of receptor-binding domain-targeting Abs from hybrid immune plasma abrogated their protection against infection. INTERPRETATION These results demonstrate the utility of TKI mice as a tool for the development of anti-SARS-CoV-2 mAb therapeutics, show that in vitro neutralisation assays do not accurately predict in vivo protection, and highlight the importance of hybrid immunity for eliciting protective anti-receptor-binding domain Abs. FUNDING This work was funded by grants from the e-Asia Joint Research Program (N10A650706 and N10A660577 to MLM, in collaboration with SS); the NIH (U19 AI142790-02S1 to EOS and SS and R44 AI157900 to KJ); the GHR Foundation (to SS and EOS); the Overton family (to SS and EOS); the Arvin Gottlieb Foundation (to SS and EOS), the Prebys Foundation (to SS); and the American Association of Immunologists Fellowship Program for Career Reentry (to FASB).
Collapse
Affiliation(s)
| | - Shailendra Kumar Verma
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Norazizah Shafee
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA; Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Robyn Miller
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Chris Conner
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Kathryn M Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Michael N Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Linda Tran
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Krithik Varghese
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Henry Madany
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | | | - Michelle Zandonatti
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Meng Ling Moi
- School of International Health, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
| | - Kurt Jarnagin
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - David R Webb
- Synbal Inc., 1759 Yorktown Rd., San Mateo, CA, 94402, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Kenneth Kim
- Histopathology Core Facility, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA, 92037, USA; Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, UC San Diego School of Medicine, La Jolla, CA, 92037, USA.
| |
Collapse
|
5
|
Dequidt T, Richier Q, Louapre C, Ader F, Merad Y, Lauwerier N, Jacomet C, Carles M, Biron C, Gendrin V, Marlat C, Danion F, Lepage TM, Sotto A, Bourdellon L, Mania A, Martinot M, Falher GL, Ferre A, Pilmis B, Gondran G, Simeone P, Henry M, Kamel T, Ray S, Ancellin S, Mélé N, Camou F, Destremau M, Sellenet J, Zucman N, Le Maréchal M, Mellouki K, Langlois ME, Luque Paz D, Mousset M, Leclerc C, Sommet A, Lacombe K, Martin-Blondel G. Convalescent plasma in patients receiving rituximab or ocrelizumab for multiple sclerosis or neuromyelitis Optica spectrum disorder with Covid-19: A multicenter retrospective study. Int J Infect Dis 2025; 151:107323. [PMID: 39643155 DOI: 10.1016/j.ijid.2024.107323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/09/2024] Open
Abstract
BACKGROUND Despite vaccination, patients receiving anti-CD20 monoclonal antibodies (mAbs) for multiple sclerosis (MS) or neuromyelitis optica spectrum disorders (NMOSD) have an increased risk of developing severe or protracted COVID-19. The aim of this study was to describe the effect of COVID-19 convalescent plasma (CCP) in patients with MS or NMOSD exposed to anti-CD20 and infected by SARS-CoV-2. METHODS This French national, retrospective cohort study was conducted between November 2020 and June 2023. Patients with MS or NMOSD, under anti-CD20 mAbs, with symptomatic COVID-19 and treated by CCP were screened. Protracted COVID-19 was defined by a duration of symptoms >21 days. The primary endpoint was the overall survival 30 days after CCP administration. RESULTS Ninety-two patients from 34 hospitals were included, 84 (91%) with MS and 8 (9%) with NMOSD. Overall, 30-day survival was 97% (IC95%: 91-99). SARS-CoV-2 viremia was positive in 47/75 (61%) patients before CCP versus 9/59 (15%) seven days post-CCP. In the 52 patients (57%) with protracted COVID-19, the duration of symptoms before CCP was 51 [28-69] days, including fever in 75% of cases, which disappeared in 100% of patients 7 days post-CCP. CONCLUSIONS CCP could be a therapeutic option in patients exposed to anti-CD20 mAbs for inflammatory demyelinating disease, particularly in those with protracted COVID-19.
Collapse
Affiliation(s)
- Tanguy Dequidt
- Department of Infectious Diseases, Guadeloupe University hospital, Pointe-à-Pitre, France.
| | - Quentin Richier
- Sorbonne University, Department of Infectious Diseases, Saint Antoine Hospital, APHP, Paris, France
| | - Céline Louapre
- Sorbonne University, Department of Neurology, Pitié Salpêtrière Hospital, APHP, CIC neurosciences, Paris Brain Institute, Paris, France
| | - Florence Ader
- Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
| | - Yanis Merad
- Department of Infectious Diseases, Hospices Civils de Lyon, Lyon, France
| | - Nicolas Lauwerier
- Department of Infectious Diseases, Lille University Hospital, Lille, France
| | - Christine Jacomet
- Department of Infectious Diseases, Clermont-Ferrand University Hospital, Clermont-Ferrand, France
| | - Michel Carles
- Department of Infectious Diseases, Nice University Hospital, Nice, France
| | - Charlotte Biron
- Department of Infectious Diseases, Nantes University Hospital, Nantes, France
| | - Vincent Gendrin
- Department of Infectious Diseases, Nord Franche-Comté Hospital, Belfort, France
| | - Clément Marlat
- Department of Infectious Diseases, Rouen University Hospital, Rouen, France
| | - François Danion
- Department of Infectious Diseases, Strasbourg University Hospital, Strasbourg, France
| | - Tristan M Lepage
- Department of Infectious Diseases, Montpellier University Hospital, Montpellier, France
| | - Albert Sotto
- Department of Infectious Diseases, Nimes University Hospital, Nimes, France
| | | | - Alexandre Mania
- Department of Internal Medicine, Henri Mondor Hospital, Aurillac, France
| | - Martin Martinot
- Department of Infectious Diseases, Colmar Hospital, Colmar, France
| | | | - Alexis Ferre
- Intensive Care Unit, Versailles Hospital, Le Chesnay, France
| | - Benoit Pilmis
- Antimicrobial Stewardship Team, Microbiology Unit, Groupe Hospitalier Paris Saint Joseph, Paris, France
| | - Guillaume Gondran
- Department of Internal Medicine, Limoges University Hospital, Limoges, France
| | - Pierre Simeone
- Department of Anesthesiology and Critical Care Medicine, University Hospital Timone, APHM, Marseille, France
| | - Matthieu Henry
- Medical-Surgical Intensive Care Unit, District Hospital Center, La Roche sur Yon, France
| | - Toufik Kamel
- Intensive Care Unit, Orléans University Hospital, Orléans, France
| | - Simon Ray
- Department of Infectious Diseases, Rodez Hospital, Rodez, France
| | - Sophie Ancellin
- Department of Infectious Diseases, Auch Hospital, Auch, France
| | - Nicolas Mélé
- Department of Neurology, GHU Paris Psychiatrie et Neurosciences, Sainte-Anne Hospital, Paris, France
| | - Fabrice Camou
- Intensive Care and Infectious Disease Unit, Groupe Saint-André, Bordeaux University Hospital, Bordeaux, France
| | - Marjolaine Destremau
- Intensive Care and Infectious Disease Unit, Groupe Saint-André, Bordeaux University Hospital, Bordeaux, France
| | - Jeremy Sellenet
- Department of Internal Medicine, Saint Jean Sud de France Clinic, Saint-Jean-de-Vedas, France
| | - Noémie Zucman
- Intensive Care Unit, CH Annecy Genevois, Epagny Metz-Tessy, France
| | - Marion Le Maréchal
- Univ. Grenoble Alpes, Infectious Diseases Department, CHU Grenoble Alpes, Grenoble, France
| | - Khawla Mellouki
- Department of Infectious Diseases, Claude Bernard University Lyon 1, Valence Hospital Center, Valence, France
| | - Marie-Elodie Langlois
- Department of Internal Medicine and Infectious Diseases, Saint Joseph Saint Luc Hospital, Lyon, France
| | - David Luque Paz
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, Rennes, France
| | - Maud Mousset
- Intensive Care Unit, Comminges Pyrénées Hospital, Saint-Gaudens, France
| | - Catherine Leclerc
- Department of Infectious Diseases, CHI Poissy - Saint-Germain-en-Laye, Saint-Germain-en-Laye, France
| | - Agnès Sommet
- Clinical Investigation Center, Toulouse University Hospital, Toulouse, France
| | - Karine Lacombe
- Sorbonne University, IPLESP Inserm, Infectious Diseases Department, Saint Antoine Hospital, APHP, Paris, France
| | - Guillaume Martin-Blondel
- Department of Infectious Diseases, Toulouse University Hospital, Toulouse, France; Toulouse Institute for Infectious and Inflammatory Diseases, INSERM UMR1291 - CNRS UMR5051 - Toulouse III University, Toulouse, France
| |
Collapse
|
6
|
Franchini M, Focosi D. Hyperimmune plasma against COVID-19: Does it work or not? Transfus Clin Biol 2025; 32:124-125. [PMID: 39395711 DOI: 10.1016/j.tracli.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/14/2024]
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantua, Italy.
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
7
|
Maison DP, Tasissa H, Deitchman A, Peluso MJ, Deng Y, Miller FD, Henrich TJ, Gerschenson M. COVID-19 clinical presentation, management, and epidemiology: a concise compendium. Front Public Health 2025; 13:1498445. [PMID: 39957982 PMCID: PMC11826932 DOI: 10.3389/fpubh.2025.1498445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/21/2025] [Indexed: 02/18/2025] Open
Abstract
Coronavirus Disease 2019, caused by severe acute respiratory coronavirus 2, has been an ever-evolving disease and pandemic, profoundly impacting clinical care, drug treatments, and understanding. In response to this global health crisis, there has been an unprecedented increase in research exploring new and repurposed drugs and advancing available clinical interventions and treatments. Given the widespread interest in this topic, this review aims to provide a current summary-for interested professionals not specializing in COVID-19-of the clinical characteristics, recommended treatments, vaccines, prevention strategies, and epidemiology of COVID-19. The review also offers a historical perspective on the pandemic to enhance understanding.
Collapse
Affiliation(s)
- David P. Maison
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Hawi Tasissa
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Amelia Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J. Peluso
- Division of HIV, Infectious Diseases, and Global Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - F. DeWolfe Miller
- Department of Tropical Medicine, Medical Microbiology and Pharmacology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| | - Timothy J. Henrich
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mariana Gerschenson
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI, United States
| |
Collapse
|
8
|
Parua P, Ghosh S, Jana K, Seth A, Debnath B, Rout SK, Sarangi MK, Dash R, Halder J, Rajwar TK, Pradhan D, Rai VK, Dash P, Das C, Kar B, Ghosh G, Rath G. Therapeutic Potential of Neutralizing Monoclonal Antibodies (nMAbs) against SARS-CoV-2 Omicron Variant. Curr Pharm Des 2025; 31:753-773. [PMID: 39543801 DOI: 10.2174/0113816128334441241108050528] [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: 07/06/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND The COVID-19 pandemic has spurred significant endeavors to devise treatments to combat SARS-CoV-2. A limited array of small-molecule antiviral drugs, specifically monoclonal antibodies and interferon therapy, have been sanctioned to treat COVID-19. These treatments typically necessitate administration within ten days of symptom onset. There have been reported reductions in the effectiveness of these medications due to mutations in non-structural protein genes, particularly against Omicron subvariants. This underscores the pressing requirement for healthcare systems to continually monitor pathogen variability and its impact on the efficacy of prevention and treatments. AIM This review aimed to comprehend the therapeutic benefits and recent progress of nMAbs for preventing and treating the Omicron variant of SARS-CoV-2. RESULTS AND DISCUSSION Neutralizing monoclonal antibodies (nMAbs) provide a treatment avenue for severely affected individuals, especially those at high risk for whom vaccination is not viable. With their specific epitope affinity, they pose no significant risk of severe adverse effects. The degree of reduction in neutralization varies significantly across different monoclonal antibodies and variant combinations. For instance, Sotrovimab maintained its neutralization effectiveness against Omicron BA.1, but exhibited diminished efficacy against BA.2, BA.4, BA.5, and BA.2.12.1. CONCLUSION Bebtelovimab has been observed to preserve its efficacy against all subtypes of the Omicron variant. Subsequently, WKS13, mAb-39, 19n01, F61-d2 cocktail, etc., have become effective. This review has highlighted the therapeutic implications of nMAbs in SARS-CoV-2 Omicron treatment and the progress of COVID-19 drug discovery.
Collapse
Affiliation(s)
- Pijus Parua
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal-711316, India
| | - Somnath Ghosh
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal-711316, India
| | - Koushik Jana
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal-711316, India
| | - Arnab Seth
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal-711316, India
| | - Biplab Debnath
- Department of Pharmaceutical Technology, Bharat Technology, Uluberia, Howrah, West Bengal-711316, India
| | - Saroj Kumar Rout
- LNK International, Inc., Hauppauge, New York-11788, United States
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Lucknow-226024, Uttar Pradesh, India
| | - Rasmita Dash
- Department of Pharmaceutics, School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Bhubaneswar-752050, Odisha, India
| | - Jitu Halder
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Tushar Kanti Rajwar
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Deepak Pradhan
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Vineet Kumar Rai
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Priyanka Dash
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Chandan Das
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Biswakanth Kar
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Goutam Ghosh
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| | - Goutam Rath
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar-751030, Odisha, India
| |
Collapse
|
9
|
Casadevall A, Focosi D, Pirofski LA, Shoham S. Single Monoclonal Antibodies Should Not Be Used for COVID-19 Therapy: A Call for Antiviral Stewardship. Clin Infect Dis 2024; 79:1404-1407. [PMID: 39115342 DOI: 10.1093/cid/ciae408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Indexed: 12/18/2024] Open
Abstract
The COVID-19 pandemic witnessed the greatest deployment of monoclonal antibody (mAb) therapies for an infectious disease, but all were defeated by SARS-CoV-2 evolution. As new mAbs are developed, the infectious disease community needs stewardship practices to reduce emergence of resistance.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, Maryland, USA
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Liise-Anne Pirofski
- Divison of Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Chaudhry A, Gallais F, Falcoz PE, De Verdiere SC, Villeneuve T, Horeau D, Chatron E, Blanchard E, Collange O, Renaud-Picard B. Efficacy of convalescent plasma for the treatment of COVID-19 in lung transplant recipients: A multicenter French study. Respir Med Res 2024; 87:101145. [PMID: 39689664 DOI: 10.1016/j.resmer.2024.101145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/26/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024]
Abstract
INTRODUCTION Lung transplant (LT) recipients are at greater risk of complications from COVID-19. Treatment options are limited partly due to interactions with immunosuppressive agents. Convalescent plasma (CP) is a potential treatment option, but it has not been extensively studied in LT patients. We aimed to assess the efficacy and safety of CP use in France for COVID-19 infected LT patients. MATERIAL AND METHODS We retrospectively recruited LT patients followed up in the 10 French LT centers, older than 18 years, infected with SARS-CoV-2 between the pandemic onset and July 1, 2023, and treated with high-titer CP. RESULTS We collected the data from 27 patients who received CP for a COVID-19 infection in six out of the 10 French LT centers. The average delay between symptom onset and CP administration was 19.5 days, and 51.8 % of patients received four units. In patients treated within the first 9 days of infection, the survival rate was 100 % at one and three months vs. 75 % (p = 0.28) for late administration patients. Average loss of forced expiratory volume in 1 second at three months was 10.5 % in the early group vs. 3.3 % in the late group (p = 0.58). The average length of hospital stay was 18 and 24 days respectively (p = 0.07). Early use of CP was also more frequent in 2023. DISCUSSION In this study highlighting the French experience for the use of CP in LT patients, we observed a limited, heterogenous but well-tolerated use of this therapy.
Collapse
Affiliation(s)
- Abouzar Chaudhry
- Service de Pneumologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Floriane Gallais
- Laboratoire de Virologie, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Pierre-Emmanuel Falcoz
- Service de Chirurgie Thoracique, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | | | | | | - Eva Chatron
- Service de Pneumologie, Hospices Civils de Lyon, Lyon, France
| | - Elodie Blanchard
- Service de Pneumologie et Transplantation Pulmonaire, CHU de Bordeaux, Bordeaux, France
| | - Olivier Collange
- Service de réanimation chirurgicale polyvalente, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | | |
Collapse
|
11
|
Grasselli Kmet N, Mavrič M, Saletinger R. Epidemiological and Clinical Characteristics of the Course of COVID-19 Among Vaccinated and Unvaccinated Heart Transplant Recipients in Slovenia. Vaccines (Basel) 2024; 12:1366. [PMID: 39772027 PMCID: PMC11680375 DOI: 10.3390/vaccines12121366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Patients receiving heart transplantation require lifelong immunosuppression and compared to the general population, they have a more than five times higher chance of acquiring COVID-19, and their mortality rates are higher. The aim of the present study was to estimate the epidemiological and clinical characteristics of COVID-19 in heart transplant recipients (HTRs) in Slovenia to estimate the vaccination rate and evaluate possible vaccination-hesitant subgroups. Methods: All SARS-CoV-2-positive HTRs (N = 79) between 1 March 2020 and 31 December 2023 at the Infectious Diseases Department, University Medical Centre Ljubljana, Slovenia, were included retrospectively. Demographic, clinical and vaccination data were extracted from medical documentation and a statistical evaluation was performed. Results: The observed vaccination rate was 63.3%, but among patients who received transplants before the pandemic, it was statistically significantly higher (p = 0.027). Vaccinated HTRs were statistically significantly older (p = 0.004) and had a significantly higher Charlson Comorbidity Index (p = 0.018). Our results indicate no significant differences between vaccinated and unvaccinated HTRs regarding acute respiratory insufficiency (p = 0.135), length of hospital stay (p = 0.106), intensive care unit admission (0.414) and in-hospital mortality (p = 0.317), but we observed statistically more frequently an asymptomatic course in those vaccinated (p = 0.050), and a longer length of stay in vector vaccine recipients (p = 0.011) and in those not re-vaccinated (p = 0.030). There was a significantly higher re-vaccination rate in males (p = 0.005). Conclusions: An asymptomatic course of COVID-19 was more often observed in vaccinated HTRs. Our findings suggest statistically significant differences in COVID-19 vaccine acceptance rates; younger HTRs and those transplanted after the pandemic are more hesitant to vaccinate, while females accept booster doses less frequently.
Collapse
Affiliation(s)
- Nina Grasselli Kmet
- Infectious Diseases Department, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (M.M.); (R.S.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Matej Mavrič
- Infectious Diseases Department, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (M.M.); (R.S.)
| | - Rajko Saletinger
- Infectious Diseases Department, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia; (M.M.); (R.S.)
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
12
|
Franchini M, Cruciani M, Mengoli C, Casadevall A, Glingani C, Joyner MJ, Pirofski LA, Senefeld JW, Shoham S, Sullivan DJ, Zani M, Focosi D. Convalescent plasma and predictors of mortality among hospitalized patients with COVID-19: a systematic review and meta-analysis. Clin Microbiol Infect 2024; 30:1514-1522. [PMID: 39067517 DOI: 10.1016/j.cmi.2024.07.020] [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: 03/24/2024] [Revised: 07/08/2024] [Accepted: 07/21/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Plasma collected from recovered patients with COVID-19 (COVID-19 convalescent plasma [CCP]) was the first antibody-based therapy employed to fight the COVID-19 pandemic. While the therapeutic effect of early administration of CCP in COVID-19 outpatients has been recognized, conflicting data exist regarding the efficacy of CCP administration in hospitalized patients. OBJECTIVES To examine the effect of CCP compared to placebo or standard treatment, and to evaluate whether time from onset of symptoms to treatment initiation influenced the effect. DATA SOURCES Electronic databases were searched for studies published from January 2020 to January 2024. STUDY ELIGIBILITY CRITERIA Randomized clinical trials (RCTs) investigating the effect of CCP on COVID-19 mortality in hospitalized patients with COVID-19. PARTICIPANTS Hospitalized patients with COVID-19. INTERVENTIONS CCP versus no CCP. ASSESSMENT OF RISK OF BIAS Cochrane risk of bias tool for RCTs. METHODS OF DATA SYNTHESIS The random-effects model was used to calculate the pooled risk ratio (RR) with 95% CI for the pooled effect estimates of CCP treatment. The Grading of Recommendations Assessment, Development and Evaluation was used to evaluate the certainty of evidence. RESULTS Twenty-seven RCTs were included, representing 18,877 hospitalized patients with COVID-19. When transfused within 7 days from symptom onset, CCP significantly reduced the risk of death compared to standard therapy or placebo (RR, 0.76; 95% CI, 0.61-0.95), while later CCP administration was not associated with a mortality benefit (RR, 0.98; 95% CI, 0.90-1.06). The certainty of the evidence was graded as moderate. Meta-regression analysis demonstrated increasing mortality effects for longer interval to transfusion or worse initial clinical severity. CONCLUSIONS In-hospital transfusion of CCP within 7 days from symptom onset conferred a mortality benefit.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy.
| | - Mario Cruciani
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Carlo Mengoli
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Claudia Glingani
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - 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 Health and Kinesiology, University of Illinois at Urbana-Champaign, IL, USA
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David J Sullivan
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, MD, USA
| | - Matteo Zani
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
13
|
Di Germanio C, Deng X, Balasko BG, Simmons G, Martinelli R, Grebe E, Stone M, Spencer BR, Saa P, Yu EA, Lanteri MC, Green V, Wright D, Lartey I, Kleinman S, Jones J, Biggerstaff BJ, Contestable P, Busch MP. Spike and nucleocapsid antibody dynamics following SARS-CoV-2 infection and vaccination: Implications for sourcing COVID-19 convalescent plasma from routinely collected blood donations. Transfusion 2024; 64:2063-2074. [PMID: 39373096 DOI: 10.1111/trf.18017] [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: 08/12/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) remains a treatment option for immunocompromised patients; however, the current FDA qualification threshold of ≥200 BAU/mL of spike antibody appears to be relatively low. We evaluated the levels of binding (bAb) and neutralizing antibodies (nAb) on serial samples from repeat blood donors who were vaccinated and/or infected to inform criteria for qualifying CCP from routinely collected plasma components. METHODS Donors were categorized into four groups: (1) infected, then vaccinated, (2) vaccinated then infected during the delta, or (3) omicron waves, (4) vaccinated without infection. IgG Spike and total Nuclecapsid bAb were measured, along with S variants and nAb titers using reporter viral particle neutralization. RESULTS Mean S IgG bAb peaks after infection alone were lower than after primary and booster vaccinations, and higher after delta and omicron infection in previously vaccinated donors. Half-lives for S IgG ranged from 34 to 66 days after first infection/vaccination events and up to 108 days after second events. The levels of S IgG bAb and nAb were similar across different variants, except for omicron, which were lower. Better correlations of nAb with bAb were observed at higher levels (hybrid immunity) than at the current FDA CCP qualifying threshold. DISCUSSION Routine plasma donations from donors with hybrid immunity had high S bAb and potent neutralizing activity for 3-6 months after infection. In donations with high (>4000 BAU/mL) S IgG, >95% had high nAb titers (>500) against ancestral and variant S, regardless of COVID-19 symptoms. These findings provide the basis for test-based criteria for qualifying CCP from routine blood donations.
Collapse
Affiliation(s)
- Clara Di Germanio
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Xutao Deng
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | | | - Graham Simmons
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | | | - Eduard Grebe
- Vitalant Research Institute, San Francisco, California, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Bryan R Spencer
- American Red Cross, Scientific Affairs, Dedham, Massachusetts, USA
| | - Paula Saa
- American Red Cross, Scientific Affairs, Rockville, Maryland, USA
| | - Elaine A Yu
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Marion C Lanteri
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
- Creative Testing Solutions, Tempe, Arizona, USA
| | | | | | | | - Steven Kleinman
- University of British Columbia, Victoria, British Columbia, Canada
| | - Jefferson Jones
- Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | | | - Michael P Busch
- Vitalant Research Institute, San Francisco, California, USA
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Chandra H, Yadav A, Prasad R, Sagar K, Bhardwaj N, Kumar Gupta K, Singh Thakur G, Nigam M, Pezzani R, Paulo Martins de Lima J, Douglas Melo Coutinho H, Prakash Mishra A. COVID 19: Prevention and treatment through the Indian perspective. Cytokine 2024; 183:156756. [PMID: 39284260 DOI: 10.1016/j.cyto.2024.156756] [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: 07/26/2024] [Revised: 08/30/2024] [Accepted: 09/06/2024] [Indexed: 11/20/2024]
Abstract
The most destructive period the world has experienced seems to be behind us. Not a single nation was spared by this disease, and many continue to struggle today. Even after recovering from COVID, patient may continue to experience some post-COVID effects, such as heart irregularities or a decline in lung vitality. In the past three years (2019-2022), the world has witnessed the power of a small entity, a single peculiar virus. Science initially appeared to be helpless in this regard, but due to the emergence of disease, pharmaceutics (the development of anti-covid drugs), immunology (the rapid antigen test), microbiology (the isolation of viruses from infected people), biotechnology (the development of recombinant vaccines), biochemistry (the blood profile, the D-dimer test), and biochemistry (blood profile, D-dimer test), biophysics (PCR, RT-PCR, CT Scan, MRI) had worked together to fight the disease. The results of these efforts are the development of new diagnostic techniques, possible treatment and finally the availability of vaccines against COVID-19. However, it is not proven that the treatment through the traditional medical system is directly active on SARS-CoV-2 but is instead indirectly acting on SARS-CoV-2 effects by improving symptoms derived from the viral disease. In India, the traditional system of medicine and tradition knowledge together worked in the pandemic and proved effective strategies in prevention and treatment of SARS-CoV-2. The use of effective masks, PPE kits, plasma therapy, yoga, lockdowns and social seclusion, use of modern antiviral drugs, monoclonal antibodies, herbal remedies, homoeopathy, hygienic practice, as well as the willpower of people, are all contributing to the fight against COVID. Which methods or practices will be effective against COVID nobody is aware since medical professionals who wear PPE kits do not live longer, and some people in India who remained unprotected and roamed freely were not susceptible to infection. The focus of this review is on the mode of transmission, diagnosis, preventive measures, vaccines currently under development, modern medicine developed against SARS-CoV-2, ayurvedic medicine used during pandemic, homoeopathic medicine used during pandemic, and specific yoga poses that can be used to lessen COVID-related symptoms.
Collapse
Affiliation(s)
- Harish Chandra
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India; School of Agriculture, Uttaranchal University, Dehradun 248007, Uttarakhand, India.
| | - Archana Yadav
- Department of Microbiology, Institute of Biosciences and Biotechnology, C.S.J.M. University, Kanpur 208024, Uttar Pradesh, India.
| | - Rajendra Prasad
- School of Agriculture, Uttaranchal University, Dehradun 248007, Uttarakhand, India.
| | - Kalpana Sagar
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India
| | - Nitin Bhardwaj
- Department of Zoology and Environmental Sciences, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India.
| | - Kartikey Kumar Gupta
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar 249404, Uttarakhand, India.
| | - Ghanshyam Singh Thakur
- Department of Naturopathy & Yoga, H. N. B. Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India.
| | - Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India.
| | - Raffaele Pezzani
- Phytotherapy Lab (PhT-Lab), Endocrinology Unit, Department of Medicine (DIMED), University of Padova, via Ospedale 105, Padova 35128, Italy; AIROB, Associazione Italiana per la Ricerca Oncologica di Base, Padova, Italy.
| | | | | | - Abhay Prakash Mishra
- Department of Pharmacology, Faculty of Health Science, University of Free State, Bloemfontein 9300, South Africa.
| |
Collapse
|
15
|
Iasella CJ, Hannan SJ, Lyons EJ, Lieber SC, Das A, Dimitrov D, Li W, Saul M, Popescu I, Koshy R, Burke R, Lape B, Brown MJ, Chen X, Sembrat JC, Devonshire K, Kitsios GD, Konstantinidis I, Snyder ME, Chen BB, Merlo CA, Hager DN, Kiss JE, Yazer MH, Wells AH, Morris A, McVerry BJ, McMahon DK, Triulzi DJ, McDyer JF. Impact of variable titer COVID-19 convalescent plasma and recipient SARS-CoV2-specific humoral immunity on survival in hospitalized patients. PLoS One 2024; 19:e0309449. [PMID: 39446792 PMCID: PMC11500870 DOI: 10.1371/journal.pone.0309449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/12/2024] [Indexed: 10/26/2024] Open
Abstract
COVID-19 convalescent plasma (CCP) was one of the first therapies to receive emergency use authorization for management of COVID-19. We assessed the effectiveness of CCP in a propensity-matched analysis, and whether the presence of antibodies in the recipient at the time of treatment or the titer of antibodies in the administered CCP influenced clinical effectiveness. In an inpatient population within a single large health system, a total of 290 CCP patients were matched to 290 controls. While CCP increased titers of anti-SARS-CoV-2 RBD IgG titers post-CCP (p = <0.0001), no differences in 30-day survival were observed between CCP patients and controls in univariate and multivariate analyses. Survival at 30 days was numerically lower in recipients who were seronegative prior to CCP administration, compared to those with low titer and high titer anti-SARS-CoV-2 RBD IgG, respectively, but did not reach statistical significance (56% vs 82% vs 75%, p = 0.16). Patients who received 2 units of high-titer CCP had numerically better survival versus those who received fewer high-titer units, but this was not statistically significant (p = 0.08). CCP did not improve 30-day survival compared to propensity matched controls. Together these data support that CCP therapy provides limited benefit to hospitalized patients with SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Carlo J. Iasella
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Stefanie J. Hannan
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Emily J. Lyons
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Sophia C. Lieber
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Antu Das
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Dimiter Dimitrov
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Wei Li
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Melissa Saul
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Iulia Popescu
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ritchie Koshy
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Robin Burke
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Braidon Lape
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania, United States of America
| | - Mark J. Brown
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Xiaoping Chen
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John C. Sembrat
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kaitlyn Devonshire
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Georgios D. Kitsios
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ioannis Konstantinidis
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Mark E. Snyder
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Bill B. Chen
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Christian A. Merlo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - David N. Hager
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States of America
| | - Joseph E. Kiss
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Mark H. Yazer
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alan H. Wells
- Department of Pathology, Division of Laboratory Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Alison Morris
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Bryan J. McVerry
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Deborah K. McMahon
- Department of Medicine, Aging Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Darrell J. Triulzi
- Department of Pathology, Division of Transfusion Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - John F. McDyer
- Department of Medicine, Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
16
|
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
|
17
|
Bohoněk M, Máca J, Sagan J, Řezáč D, Fridrich V, Burantová A, Kutáč D, Vabroušek P, Kubů J, Chrdle A, Volfová K, Blahutová Š, Rychlík I, Vonášková K, Majerčin R, Králová R, Štěpánek P, Holub M. Convalescent anti-SARS-CoV-2 plasma for the treatment of patients with COVID-19: a retrospective study RESCOVID-19. Virol J 2024; 21:239. [PMID: 39350163 PMCID: PMC11443855 DOI: 10.1186/s12985-024-02475-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024] Open
Abstract
PURPOSE Convalescent plasma (CP) collected from people who recovered from COVID-19 became a rapidly available treatment modality in numerous countries, including the Czech Republic. The aims of our study were to evaluate the effectiveness and safety of CP in the treatment of COVID-19. METHODS This retrospective observational study involved six Czech hospitals. This study enrolled patients with and without CP treatment who were hospitalized between April 2020 and April 2021. Propensity score matching and logistic regression analysis were performed to evaluate the influence of CP administration and its timing on the in-hospital survival of COVID-19 patients. RESULTS A total of 1,498 patients were enrolled in the study; 406 (27%) were administered CP, and 1,092 (73%) were not treated with CP. The propensity score-matched control group consisted of 1,218 subjects. The survival of patients treated with CP was 79%, while that of patients in the matched control group was 62% (P<0.001). Moreover, the chance of survival was significantly greater when CP was administered within three days after the onset of COVID-19 symptoms than when CP was administered after four or more days (87% vs. 76%, P <0.001). In addition, adverse effects related to CP administration were recorded in only 2% of patients and were considered mild in all patients. CONCLUSIONS Our study demonstrated that the administration of CP was safe and possibly associated with positive effects that were more pronounced if CP was administered within the first three days after the onset of COVID-19 symptoms.
Collapse
Affiliation(s)
- Miloš Bohoněk
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University in Prague, Prague, Czech Republic
| | - Jan Máca
- Department of Anesthesiology and Intensive Care, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
- Institute of Physiology and Pathological Physiology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jiří Sagan
- Department of Infectious Diseases, Faculty of Medicine, University of Ostrava and University Hospital Ostrava, Ostrava, Czech Republic
- Department of Clinical Studies, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - David Řezáč
- The 7th Field Hospital of the Army of the Czech Republic, Hradec Králové, Czech Republic
| | - Viktor Fridrich
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic
| | - Anna Burantová
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic
| | - Dominik Kutáč
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
| | - Pavel Vabroušek
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
| | - Jan Kubů
- Department of Science and Research, University Hospital Bulovka Prague, Prague, Czech Republic
| | - Aleš Chrdle
- Department of Infectious Diseases, České Budejovice Hospital, České Budějovice, Czech Republic
- Faculty of Health and Social Sciences, University of South Bohemia, České Budějovice, Czech Republic
| | - Kateřina Volfová
- Department of Infectious Diseases, České Budejovice Hospital, České Budějovice, Czech Republic
| | - Šárka Blahutová
- Institute of Laboratory Hematology and Transfusion Medicine, Department of Biomedical Sciences, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Ivan Rychlík
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Kateřina Vonášková
- Department of Internal Medicine, Third Faculty of Medicine, Charles University and University Hospital Královské Vinohrady, Prague, Czech Republic
| | - Radek Majerčin
- Department of Anesthesiology and Resuscitation, Regional Hospital Jičín, Jičín, Czech Republic
| | - Radka Králová
- Department of Anesthesiology and Resuscitation, Regional Hospital Jičín, Jičín, Czech Republic
| | - Petr Štěpánek
- Department of Anesthesiology and Resuscitation, Regional Hospital Náchod, Náchod, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czech Republic.
- Ústřední vojenská nemocnice - Vojenská fakultní nemocnice Praha, U Vojenské nemocnice 1200, Praha 6, 169 06, Czech Republic.
| |
Collapse
|
18
|
Franchini M, Casadevall A, Dragotakes Q, Focosi D. Avoided and Avoidable Deaths with the Use of COVID-19 Convalescent Plasma in Italy during the First Two Years of Pandemic. Life (Basel) 2024; 14:1207. [PMID: 39337989 PMCID: PMC11433499 DOI: 10.3390/life14091207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Italy was the first western country to be hit by the COVID-19 pandemic and has suffered nearly 200,000 deaths so far during the four years of the pandemic. In March 2020, Italy first deployed COVID-19 convalescent plasma (CCP) to treat hospitalized patients. Despite this initial effort, the proportion of COVID-19 patients treated with CCP during the first two years of the pandemic (2020-2021) was very low (approximately 2% of individuals hospitalized for COVID-19). In this study, we estimated the number of actual inpatient lives saved by CCP treatment in Italy using national mortality data, and CCP mortality reduction data from meta-analyses of randomized controlled trials and real-world data. We also estimated the potential number of lives saved if CCP had been deployed to 100% of hospitalized patients or used in 15% to 75% of outpatients. According to these models, CCP usage in 2020-2021 saved between 385-1304 lives, but this number would have increased to 17,751-60,079 if 100% of inpatients had been transfused with CCP. Similarly, broader (15-75%) usage in outpatients could have prevented 21,187-190,689 hospitalizations (desaturating hospitals) and 6144-81,926 deaths. These data have important implications for convalescent plasma use in future infectious disease emergencies.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy;
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (A.C.); (Q.D.)
| | - Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (A.C.); (Q.D.)
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| |
Collapse
|
19
|
Lucaj T, Hay I, Samarbakhsh A, Bedi M, Iyer AK, Gavande NS. An overview of the development of pharmacotherapeutics targeting SARS-CoV-2. Drug Discov Today 2024; 29:104126. [PMID: 39097220 DOI: 10.1016/j.drudis.2024.104126] [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: 01/06/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Coronavirus disease 2019 (COVID-19) was declared a global pandemic in March 2020, which precipitated urgent public health responses. The causative agent, SARS-CoV-2, spreads primarily via respiratory droplets, necessitating precautions to mitigate transmission risks. Biopharmaceutical industries and academic institutions worldwide swiftly redirected their research endeavors towards developing therapeutic interventions, focusing on monoclonal antibodies, antiviral agents, and immunomodulatory therapies. The evolving body of evidence surrounding these treatments has prompted successive updates and revisions from the FDA, delineating the evolving landscape of COVID-19 therapeutics. This review comprehensively examines each treatment modality within the context of their developmental trajectories and regulatory approvals throughout the pandemic. Furthermore, it elucidates their mechanisms of action and presents clinical data underpinning their utility in combating the COVID-19 crisis.
Collapse
Affiliation(s)
- Tom Lucaj
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Ian Hay
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Amirreza Samarbakhsh
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Mel Bedi
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Arun K Iyer
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Navnath S Gavande
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
20
|
Kow CS, Ramachandram DS, Hasan SS, Thiruchelvam K. Exploring passive immunization therapies for COVID-19 management in rheumatic patients. Int J Rheum Dis 2024; 27:e15325. [PMID: 39221822 DOI: 10.1111/1756-185x.15325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 07/08/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Affiliation(s)
- Chia Siang Kow
- School of Pharmacy, IMU University, Kuala Lumpur, Malaysia
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | | | - Syed Shahzad Hasan
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | | |
Collapse
|
21
|
Janssen M, Leo A, Wolf C, Stenzinger M, Bartenschlager M, Brandt J, Sauer S, Schmitt M, Dreger P, Schlenk RF, Denkinger CM, Müller-Tidow C. Treatment of chronic COVID-19 with convalescent/postvaccination plasma in patients with hematologic malignancies. Int J Cancer 2024; 155:618-626. [PMID: 38721724 DOI: 10.1002/ijc.34988] [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: 11/11/2023] [Revised: 02/24/2024] [Accepted: 03/20/2024] [Indexed: 06/20/2024]
Abstract
Immunocompromised patients are at high risk to fail clearance of SARS-CoV-2. Prolonged COVID-19 constitutes a health risk and a management problem as cancer treatments often have to be disrupted. As SARS-CoV-2 evolves, new variants of concern have emerged that evade available monoclonal antibodies. Moreover, antiviral therapy promotes SARS-CoV-2 escape mutations, particularly in immunocompromised patients. These patients frequently suffer from prolonged infection. No successful treatment has been established for persistent COVID-19 infection. Here, we report on a series of 21 immunocompromised patients with COVID-19-most of them hematologic malignancies-treated with plasma obtained from recently convalescent or vaccinated donors or a combination thereof. Repeated dosing of SARS-CoV-2-antibody-containing plasma could clear SARS-CoV-2 infection in 16 out of 21 immunocompromised patients even if COVID-19-specific treatments failed to induce sustained viral clearance or to improve clinical course of SARS-CoV-2 infection. Ten patients were major responders defined as an increase delta(d)Ct of > = 5 after the first administration of convalescent and/or vaccinated plasma (C/VP). On average, SARS-CoV-2 PCR Ct values increased from a median value of 22.55 (IQR = 19.10-24.25) to a median value of 29.57 (IQR = 27.55-34.63; p = <.0001) in the major response subgroup. Furthermore, when treated a second time with C/VP, even 4 out of 5 of the initial nonresponders showed an increase in Ct-values from a median value of 23.13 (IQR = 17.75-28.05) to a median value of 32.79 (IQR = 31.75-33.75; p = .013). Our results suggest that C/VP could be a feasible treatment of COVID-19 infection in patients with hematologic malignancies who did not respond to antiviral treatment.
Collapse
Affiliation(s)
- Maike Janssen
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Albrecht Leo
- Institute for Clinical Transfusion Medicine and Cell Therapy Heidelberg, Heidelberg, Germany
| | - Cornelia Wolf
- Institute for Clinical Transfusion Medicine and Cell Therapy Heidelberg, Heidelberg, Germany
| | - Miriam Stenzinger
- Institute for Clinical Transfusion Medicine and Cell Therapy Heidelberg, Heidelberg, Germany
| | - Marie Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Juliane Brandt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Sandra Sauer
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard F Schlenk
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- NCT-Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Claudia M Denkinger
- Division of Tropical Medicine, Department of Infectious Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
22
|
Yoon H, Pirofski LA. Generating the Evidence Base for Convalescent Plasma Use for a New Infectious Disease. Curr Top Microbiol Immunol 2024. [PMID: 39117847 DOI: 10.1007/82_2024_275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) swept across the world in the waning months of 2019 and emerged as the cause of the coronavirus disease 19 (COVID-19) pandemic in early 2020. The use of convalescent plasma (CP) for prior respiratory pandemics provided a strong biological rationale for the rapid deployment of COVID-19 convalescent plasma (CCP) in early 2020 when no validated treatments or prior immunity existed. CCP is an antiviral agent, with its activity against SARS-CoV-2 stemming from specific antibodies elicited by the virus. Early efforts to investigate the efficacy of CCP in randomized clinical trials (RCTs) that targeted hospitalized patients with COVID-19 did not demonstrate the overall efficacy of CCP despite signals of benefit in certain subgroups, such as those treated earlier in disease. In contrast, studies adhering to the principles of antibody therapy in their study design, choice of patient population, and product qualification, i.e., those that administered high levels of specific antibody during the viral phase of disease in immunocompromised or very early in immunocompetent individuals, demonstrated benefits. In this chapter, we leverage the knowledge gained from clinical studies of CCP for COVID-19 to propose a framework for future studies of CP for a new infectious disease. This framework includes obtaining high-quality CP and designing clinical studies that adhere to the principles of antibody therapy to generate a robust evidence base for using CP.
Collapse
Affiliation(s)
- Hyunah Yoon
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
- Department of Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA.
| |
Collapse
|
23
|
Shoham S. Convalescent Plasma for Immunocompromised Patients. Curr Top Microbiol Immunol 2024. [PMID: 39117848 DOI: 10.1007/82_2024_272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
COVID-19 convalescent plasma (CCP) is an important therapeutic option for immunocompromised patients with COVID-19. Such patients are at increased risk for serious complications of infection and may also develop a unique syndrome of persistent infection. This article reviews the rationale for CCP utilization in immunocompromised patients and the evidence for its value in immunosuppressed patients with both acute and persistent COVID-19. Both historical precedence and understanding of the mechanisms of action of antibody treatment support this use, as do several lines of evidence derived from case series, comparative studies, randomized trials, and systematic reviews of the literature. A summary of recommendations from multiple practice guidelines is also provided.
Collapse
Affiliation(s)
- Shmuel Shoham
- Department of Medicine, Johns Hopkins School of Medicine, 1830 East Monument St., Room 447, Baltimore, MD, 21205, USA.
| |
Collapse
|
24
|
Wang Y, Xu Z, Xu X, Yang S, Li Y, Zhang H, Zhang Y, Wang FS, Wang Y, Bi J. The effect of convalescent plasma therapy on the rate of nucleic acid negative conversion in patients with persistent COVID-19 test positivity. Front Pharmacol 2024; 15:1421516. [PMID: 39148549 PMCID: PMC11324536 DOI: 10.3389/fphar.2024.1421516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
Objective This study investigates the association between convalescent plasma therapy and the negative conversion rate in patients with persistent COVID-19 test positivity. Method A retrospective analysis was conducted on patients with severe or mild to moderate COVID-19 whose viral nucleic acid tests remained positive for over 30 days. Patients were categorized into two groups: those who administered convalescent plasma therapy and those who were not. Data collected included information on therapy strategies used (convalescent plasma, corticosteroids, interferons, etc.), patients' demographic characteristics, comorbidities, therapeutic medications, and nucleic acid testing results. Patients in the convalescent plasma therapy group were matched 1:2 ratio with those in the non-convalescent plasma therapy group. Cumulative negative conversion rates on the fifth, tenth, and fifteenth days post-therapy initiation were analyzed as dependent variables. Independent variables included therapy strategies, demographic characteristics, comorbidities, and therapeutic medication usage. Univariate analysis was conducted, and factors with a p-value (P) less than 0.2 were included in a paired Cox proportional hazards model. Results There was no statistically significant difference in the cumulative negative conversion rate between the convalescent plasma therapy group and the non-convalescent plasma therapy group on the fifth, tenth, and fifteenth days. Specifically, on day the fifth, the negative conversion rate was 41.46% in the convalescent plasma therapy group compared to 34.15% in the non-convalescent plasma therapy group (HR: 1.72, 95% CI: 0.82-3.61, P = 0.15). On the tenth day, it was 63.41% in the convalescent plasma therapy group and 63.41% in the non-convalescent plasma therapy group (HR: 1.25, 95% CI: 0.69∼2.26, P = 0.46). On the fifteenth day, the negative conversion rate was 85.37% in the convalescent plasma therapy group and 75.61% in the non-convalescent plasma therapy group (HR: 1.19, 95% CI: 0.71-1.97, P = 0.51). Conclusion Our finding does not support the hypothesis that convalescent plasma therapy could accelerate the time to negative conversion in patients who consistently test positive for COVID-19.
Collapse
Affiliation(s)
- Yixuan Wang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| | - Zhe Xu
- Treatment and Research Center for Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Xue Xu
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| | - Shuwen Yang
- School of Pharmaceutical Sciences, Shandong University of Traditional Chinese Medicine, Jinan, China
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| | - Yuanyuan Li
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| | - Hanwen Zhang
- Department of Gastroenterology of Chinese PLA General Hospital, Beijing, China
| | - Yufeng Zhang
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| | - Fu-Sheng Wang
- Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ying Wang
- Respiratory Department No. 960 Hospital, The PLA, Jinan, China
| | - Jingfeng Bi
- Phase I Clinical Trial Ward, The Fifth Medical Center of Chinese the PLA General Hospital, Beijing, China
| |
Collapse
|
25
|
Meijer SE, Paran Y, Belkin A, Ben-Ami R, Maor Y, Nesher L, Hussein K, Rahav G, Brosh-Nissimov T. Persistent COVID-19 in immunocompromised patients-Israeli society of infectious diseases consensus statement on diagnosis and management. Clin Microbiol Infect 2024; 30:1012-1017. [PMID: 38642895 DOI: 10.1016/j.cmi.2024.04.009] [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: 01/05/2024] [Revised: 04/05/2024] [Accepted: 04/15/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Immunocompromised patients with impaired humoral immunity are at risk for persistent COVID-19 (pCOVID), a protracted symptomatic disease with active viral replication. OBJECTIVES To establish a national consensus statement on the diagnosis, treatment, management, isolation, and prevention of pCOVID in adults. SOURCES We base our suggestions on the available literature, our own experience, and clinical reasoning. CONTENT Literature on the treatment of pCOVID is scarce and consists of few case reports and case series. The available studies provide low-quality evidence for monoclonal antibodies, convalescent plasma, antiviral drugs, and immunomodulators. Different combination therapies are described. Continuous viral replication and antiviral treatment may lead to the development of mutations that confer resistance to therapy. IMPLICATIONS To reduce the risk of resistance and improve outcomes, we suggest treating pCOVID with a combination of antibody-based therapy and two antiviral drugs for duration of 5-10 days. Immunomodulatory therapy can be added in patients with an inflammatory clinical picture. In cases of treatment failure or relapse, prolonged antiviral treatment can be considered. For the prevention of pCOVID, we suggest active and passive vaccination and early initiation of treatment for acute COVID-19. Additional research on pCOVID treatment is urgently needed.
Collapse
Affiliation(s)
- Suzy E Meijer
- Infectious Disease Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Yael Paran
- Infectious Disease Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ana Belkin
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Sheba Medical Center, Tel Hashomer, Israel
| | - Ronen Ben-Ami
- Infectious Disease Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Yasmin Maor
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Infectious Disease Unit, Edith Wolfson Medical Center, Holon, Israel
| | - Lior Nesher
- Infectious Disease Institute, Soroka University Medical Center, Beer Sheba, Israel; Faculty of Health Sciences, Ben Gurion University in the Negev, Beer Sheba, Israel
| | | | - Galia Rahav
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Sheba Medical Center, Tel Hashomer, Israel
| | - Tal Brosh-Nissimov
- Infectious Diseases Unit, Samson Assuta Ashdod University Hospital, Ashdod, Israel; Faculty of Health Sciences, Ben Gurion University in the Negev, Beer Sheba, Israel
| |
Collapse
|
26
|
Gröning R, Walde J, Ahlm C, Forsell MNE, Normark J, Rasmuson J. Intravenous immunoglobulin therapy for COVID-19 in immunocompromised patients: A retrospective cohort study. Int J Infect Dis 2024; 144:107046. [PMID: 38615825 DOI: 10.1016/j.ijid.2024.107046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024] Open
Abstract
OBJECTIVES To investigate the effectiveness of intravenous immunoglobulin (IVIG) as treatment for COVID-19 in immunocompromised patients. METHODS This retrospective study investigated outcomes for immunocompromised, vaccine non-responsive, patients that between September 2022 and April 2023 received IVIG as treatment for COVID-19 in the region of Västerbotten, Sweden. We analyzed clinical data, viral load, and anti-SARS-CoV-2 IgG binding and neutralization levels of patient serum samples and IVIG production batches. Primary and secondary outcomes were clinical cure and viral clearance, respectively. RESULTS Sixteen patients were analyzed. After a median COVID-19 duration of 4 weeks, a median 60 g IVIG infusion increased SARS-CoV-2 binding and neutralizing antibody levels, with broad in vitro activity against tested variants. The treatment resulted in abrogation of viremia in all patients and general improvement in 15 survivors that all met the primary endpoint. Thirteen patients met the secondary endpoint at follow-up after a median of four months. Two subjects with persistent SARS-CoV-2 carriage relapsed but were successfully retreated with IVIG. CONCLUSIONS Antibodies in IVIG efficiently neutralized several SARS-CoV-2 variants. Treatment with IVIG was associated with clinical cure and viral clearance in immunocompromised patients. Our data suggests that IVIG could be a novel treatment alternative for COVID-19 for this patient category.
Collapse
Affiliation(s)
- Remigius Gröning
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Jonatan Walde
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Clas Ahlm
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | | | - Johan Normark
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Johan Rasmuson
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden.
| |
Collapse
|
27
|
Van Coillie S, Prévot J, Sánchez-Ramón S, Lowe DM, Borg M, Autran B, Segundo G, Pecoraro A, Garcelon N, Boersma C, Silva SL, Drabwell J, Quinti I, Meyts I, Ali A, Burns SO, van Hagen M, Pergent M, Mahlaoui N. Charting a course for global progress in PIDs by 2030 - proceedings from the IPOPI global multi-stakeholders' summit (September 2023). Front Immunol 2024; 15:1430678. [PMID: 39055704 PMCID: PMC11270239 DOI: 10.3389/fimmu.2024.1430678] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
The International Patient Organisation for Primary Immunodeficiencies (IPOPI) held its second Global Multi-Stakeholders' Summit, an annual stimulating and forward-thinking meeting uniting experts to anticipate pivotal upcoming challenges and opportunities in the field of primary immunodeficiency (PID). The 2023 summit focused on three key identified discussion points: (i) How can immunoglobulin (Ig) therapy meet future personalized patient needs? (ii) Pandemic preparedness: what's next for public health and potential challenges for the PID community? (iii) Diagnosing PIDs in 2030: what needs to happen to diagnose better and to diagnose more? Clinician-Scientists, patient representatives and other stakeholders explored avenues to improve Ig therapy through mechanistic insights and tailored Ig preparations/products according to patient-specific needs and local exposure to infectious agents, amongst others. Urgency for pandemic preparedness was discussed, as was the threat of shortage of antibiotics and increasing antimicrobial resistance, emphasizing the need for representation of PID patients and other vulnerable populations throughout crisis and care management. Discussion also covered the complexities of PID diagnosis, addressing issues such as global diagnostic disparities, the integration of patient-reported outcome measures, and the potential of artificial intelligence to increase PID diagnosis rates and to enhance diagnostic precision. These proceedings outline the outcomes and recommendations arising from the 2023 IPOPI Global Multi-Stakeholders' Summit, offering valuable insights to inform future strategies in PID management and care. Integral to this initiative is its role in fostering collaborative efforts among stakeholders to prepare for the multiple challenges facing the global PID community.
Collapse
Affiliation(s)
- Samya Van Coillie
- International Patient Organisation for Primary Immunodeficiencies (IPOPI), Brussels, Belgium
| | - Johan Prévot
- International Patient Organisation for Primary Immunodeficiencies (IPOPI), Brussels, Belgium
| | - Silvia Sánchez-Ramón
- Department of Clinical Immunology, Health Research Institute of the Hospital Clínico San Carlos/Fundación para la Investigación Biomédica del Hospital Clínico San Carlos (IML and IdISSC), Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - David M. Lowe
- Department of Immunology, Royal Free London National Heath System (NHS) Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Michael Borg
- Department of Infection Control & Sterile Services, Mater Dei Hospital, Msida, Malta
| | - Brigitte Autran
- Sorbonne-Université, Cimi-Paris, Institut national de la santé et de la recherche médicale (INSERM) U1135, centre national de la recherche scientifique (CNRS) ERL8255, Université Pierre et Marie Curie Centre de Recherche n°7 (UPMC CR7), Paris, France
| | - Gesmar Segundo
- Departamento de Pediatra, Universidade Federal de Uberlândia, Uberlandia, MG, Brazil
| | - Antonio Pecoraro
- Transfusion Medicine Unit, Azienda Sanitaria Territoriale, Ascoli Piceno, Italy
| | - Nicolas Garcelon
- Université de Paris, Imagine Institute, Data Science Platform, Institut national de la santé et de la recherche médicale Unité Mixte de Recherche (INSERM UMR) 1163, Paris, France
| | - Cornelis Boersma
- Health-Ecore B.V., Zeist, Netherlands
- Unit of Global Health, Department of Health Sciences, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
- Department of Management Sciences, Open University, Heerlen, Netherlands
| | - Susana L. Silva
- Serviço de Imunoalergologia, Unidade Local de Saúde de Santa Maria, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Jose Drabwell
- International Patient Organisation for Primary Immunodeficiencies (IPOPI), Brussels, Belgium
| | - Isabella Quinti
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Department of Microbiology, Immunology and Transplantation, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Adli Ali
- Department of Paediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Hospital Tunku Ampuan Besar Tuanku Aishah Rohani, Universiti Kebangsaan Malaysia (UKM) Specialist Children’s Hospital, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Siobhan O. Burns
- Department of Immunology, Royal Free London National Heath System (NHS) Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Martin van Hagen
- Department of Internal Medicine, Division of Allergy & Clinical Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Immunology, Erasmus University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Martine Pergent
- International Patient Organisation for Primary Immunodeficiencies (IPOPI), Brussels, Belgium
| | - Nizar Mahlaoui
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants malades University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- French National Reference Center for Primary Immune Deficiencies (CEREDIH), Necker-Enfants malades University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| |
Collapse
|
28
|
Park H, Yu C, Pirofski LA, Yoon H, Wu D, Li Y, Tarpey T, Petkova E, Antman EM, Troxel AB. Association between COVID-19 convalescent plasma antibody levels and COVID-19 outcomes stratified by clinical status at presentation. BMC Infect Dis 2024; 24:639. [PMID: 38926676 PMCID: PMC11201301 DOI: 10.1186/s12879-024-09529-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND There is a need to understand the relationship between COVID-19 Convalescent Plasma (CCP) anti-SARS-CoV-2 IgG levels and clinical outcomes to optimize CCP use. This study aims to evaluate the relationship between recipient baseline clinical status, clinical outcomes, and CCP antibody levels. METHODS The study analyzed data from the COMPILE study, a meta-analysis of pooled individual patient data from 8 randomized clinical trials (RCTs) assessing the efficacy of CCP vs. control, in adults hospitalized for COVID-19 who were not receiving mechanical ventilation at randomization. SARS-CoV-2 IgG levels, referred to as 'dose' of CCP treatment, were retrospectively measured in donor sera or the administered CCP, semi-quantitatively using the VITROS Anti-SARS-CoV-2 IgG chemiluminescent immunoassay (Ortho-Clinical Diagnostics) with a signal-to-cutoff ratio (S/Co). The association between CCP dose and outcomes was investigated, treating dose as either continuous or categorized (higher vs. lower vs. control), stratified by recipient oxygen supplementation status at presentation. RESULTS A total of 1714 participants were included in the study, 1138 control- and 576 CCP-treated patients for whom donor CCP anti-SARS-CoV2 antibody levels were available from the COMPILE study. For participants not receiving oxygen supplementation at baseline, higher-dose CCP (/control) was associated with a reduced risk of ventilation or death at day 14 (OR = 0.19, 95% CrI: [0.02, 1.70], posterior probability Pr(OR < 1) = 0.93) and day 28 mortality (OR = 0.27 [0.02, 2.53], Pr(OR < 1) = 0.87), compared to lower-dose CCP (/control) (ventilation or death at day 14 OR = 0.79 [0.07, 6.87], Pr(OR < 1) = 0.58; and day 28 mortality OR = 1.11 [0.10, 10.49], Pr(OR < 1) = 0.46), exhibiting a consistently positive CCP dose effect on clinical outcomes. For participants receiving oxygen at baseline, the dose-outcome relationship was less clear, although a potential benefit for day 28 mortality was observed with higher-dose CCP (/control) (OR = 0.66 [0.36, 1.13], Pr(OR < 1) = 0.93) compared to lower-dose CCP (/control) (OR = 1.14 [0.73, 1.78], Pr(OR < 1) = 0.28). CONCLUSION Higher-dose CCP is associated with its effectiveness in patients not initially receiving oxygen supplementation, however, further research is needed to understand the interplay between CCP anti-SARS-CoV-2 IgG levels and clinical outcome in COVID-19 patients initially receiving oxygen supplementation.
Collapse
Affiliation(s)
- Hyung Park
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Chang Yu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Liise-Anne Pirofski
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Hyunah Yoon
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY, USA
| | - Danni Wu
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Yi Li
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Thaddeus Tarpey
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
| | - Eva Petkova
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA
- Department of Child and Adolescent Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Elliott M Antman
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrea B Troxel
- Department of Population Health, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Franchini M, Mengoli C, Casadevall A, Focosi D. Exploring Study Design Foibles in Randomized Controlled Trials on Convalescent Plasma in Hospitalized COVID-19 Patients. Life (Basel) 2024; 14:792. [PMID: 39063547 PMCID: PMC11278192 DOI: 10.3390/life14070792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Background: Sample size estimation is an essential step in the design of randomized controlled trials (RCTs) evaluating a treatment effect. Sample size is a critical variable in determining statistical significance and, thus, it significantly influences RCTs' success or failure. During the COVID-19 pandemic, many RCTs tested the efficacy of COVID-19 convalescent plasma (CCP) in hospitalized patients but reported different efficacies, which could be attributed to, in addition to timing and dose, inadequate sample size estimates. Methods: To assess the sample size estimation in RCTs evaluating the effect of treatment with CCP in hospitalized COVID-19 patients, we searched the medical literature between January 2020 and March 2024 through PubMed and other electronic databases, extracting information on expected size effect, statistical power, significance level, and measured efficacy. Results: A total of 32 RCTs were identified. While power and significance level were highly consistent, heterogeneity in the expected size effect was relevant. Approximately one third of the RCTs did not reach the planned sample size for various reasons, with the most important one being slow patient recruitment during the pandemic's peaks. RCTs with a primary outcome in favor of CCP treatment had a significant lower median absolute difference in the expected size effect than unfavorable RCTs (20.0% versus 33.9%, P = 0.04). Conclusions: The analyses of sample sizes in RCTs of CCP treatment in hospitalized COVID-19 patients reveal that many underestimated the number of participants needed because of excessively high expectations on efficacy, and thus, these studies had low statistical power. This, in combination with a lower-than-planned recruitment of cases and controls, could have further negatively influenced the primary outcomes of the RCTs.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, 46100 Mantua, Italy
| | - Carlo Mengoli
- Department of Hematology and Transfusion Medicine, Carlo Poma Hospital, 46100 Mantua, Italy
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, MD 21205, USA;
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy
| |
Collapse
|
30
|
Joyner MJ. Convalescent Plasma and the US Expanded Access Program: A Personal Narrative. Curr Top Microbiol Immunol 2024. [PMID: 38877204 DOI: 10.1007/82_2024_269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2024]
Abstract
Between early April 2020 and late August 2020, nearly 100,000 patients hospitalized with SARS-CoV2 infections were treated with COVID-19 convalescent plasma (CCP) in the US under the auspices of an FDA-authorized Expanded Access Program (EAP) housed at the Mayo Clinic. Clinicians wishing to provide CCP to their patients during that 5-month period early in the COVID pandemic had to register their patients and provide clinical information to the EAP program. This program was utilized by some 2,200 US hospitals located in every state ranging from academic medical centers to small rural hospitals and facilitated the treatment of an ethnically and socio-economically diverse cross section of patients. Within 6 weeks of program initiation, the first signals of safety were found in 5,000 recipients of CCP, supported by a later analysis of 20,000 recipients (Joyner et al. in J Clin Invest 130:4791-4797, 2020a; Joyner et al. in Mayo Clin Proc 95:1888-1897, 2020b). By mid-summer of 2020, strong evidence was produced showing that high-titer CCP given early in the course of hospitalization could lower mortality by as much as a third (Joyner et al. in N Engl J Med 384:1015-1027, 2021; Senefeld et al. in PLoS Med 18, 2021a). These data were used by the FDA in its August decision to grant Emergency Use Authorization for CCP use in hospitals. This chapter provides a personal narrative by the principal investigator of the EAP that describes the events leading up to the program, some of its key outcomes, and some lessons learned that may be applicable to the next pandemic. This vast effort was a complete team response to a crisis and included an exceptional level of collaboration both inside and outside of the Mayo Clinic. Writing just 4 years after the initiation of the EAP, this intense professional effort, comprising many moving parts, remains hard to completely understand or fully explain in this brief narrative. As Nelson Mandela said of the perception of time during his decades in prison, "the days seemed like years, and the years seemed like days."
Collapse
Affiliation(s)
- Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
31
|
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
|
32
|
Huygens S, GeurtsvanKessel C, Gharbharan A, Bogers S, Worp N, Boter M, Bax HI, Kampschreur LM, Hassing RJ, Fiets RB, Levenga H, Afonso PM, Koopmans M, Rijnders BJA, Oude Munnink BB. Clinical and Virological Outcome of Monoclonal Antibody Therapies Across SARS-CoV-2 Variants in 245 Immunocompromised Patients: A Multicenter Prospective Cohort Study. Clin Infect Dis 2024; 78:1514-1521. [PMID: 38445721 PMCID: PMC11175671 DOI: 10.1093/cid/ciae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Immunocompromised patients (ICPs) have an increased risk for a severe and prolonged COVID-19. SARS-CoV-2 monoclonal antibodies (mAbs) were extensively used in these patients, but data from randomized trials that focus on ICPs are lacking. We evaluated the clinical and virological outcome of COVID-19 in ICPs treated with mAbs across SARS-CoV-2 variants. METHODS In this multicenter prospective cohort study, we enrolled B-cell- and/or T-cell-deficient patients treated with casirivimab/imdevimab, sotrovimab, or tixagevimab/cilgavimab. SARS-CoV-2 RNA was quantified and sequenced weekly, and time to viral clearance, viral genome mutations, hospitalization, and death rates were registered. RESULTS Two hundred and forty five patients infected with the Delta (50%) or Omicron BA.1, 2, or 5 (50%) variant were enrolled. Sixty-seven percent were vaccinated; 78 treated as outpatients, of whom 2 required hospital admission, but both survived. Of the 159 patients hospitalized at time of treatment, 43 (27%) required mechanical ventilation or died. The median time to viral clearance was 14 days (interquartile range, 7-22); however, it took >30 days in 15%. Resistance-associated spike mutations emerged in 9 patients in whom the median time to viral clearance was 63 days (95% confidence interval, 57-69; P < .001). Spike mutations were observed in 1 of 42 (2.4%) patients after treatment with 2 active mAbs, in 5 of 34 (14.7%) treated with actual monotherapy (sotrovimab), and 3 of 20 (12%) treated with functional monotherapy (ie, tixagevimab/cilgavimab against tixagevimab-resistant variant). CONCLUSIONS Despite treatment with mAbs, morbidity and mortality of COVID-19 in ICPs remained substantial. Combination antiviral therapy should be further explored and may be preferred in severely ICPs.
Collapse
Affiliation(s)
- Sammy Huygens
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Corine GeurtsvanKessel
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Arvind Gharbharan
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Susanne Bogers
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Nathalie Worp
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marjan Boter
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Hannelore I Bax
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Linda M Kampschreur
- Department of Internal Medicine, Medical Center Leeuwarden, Leeuwarden, The Netherlands
| | - Robert-Jan Hassing
- Department of Internal Medicine, Rijnstate Hospital, Arnhem, The Netherlands
| | - Roel B Fiets
- Department of Internal Medicine, Amphia Hospital, Breda, The Netherlands
| | - Henriette Levenga
- Department of Internal Medicine, Groene Hart Gouda, Gouda, The Netherlands
| | - Pedro Miranda Afonso
- Department of Biostatistics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Epidemiology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Marion Koopmans
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Bas B Oude Munnink
- Department of Viroscience, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
33
|
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
|
34
|
Franchini M, Casadevall A, Senefeld JW, Joyner MJ, Sullivan DJ, Focosi D. Recommendations on the use of COVID-19 Convalescent Plasma to Treat Immunocompromised Patients. Semin Thromb Hemost 2024; 50:648-653. [PMID: 37984358 DOI: 10.1055/s-0043-1776876] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Jonathon W Senefeld
- Department of Kinesiology and Community Healthy, University of Illinois at Urbana-Champaign, Illinois
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
| | - David J Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
35
|
Ripoll JG, Tulledge-Scheitel SM, Stephenson AA, Ford S, Pike ML, Gorman EK, Hanson SN, Juskewitch JE, Miller AJ, Zaremba S, Ovrom EA, Razonable RR, Ganesh R, Hurt RT, Fischer EN, Derr AN, Eberle MR, Larsen JJ, Carney CM, Theel ES, Parikh SA, Kay NE, Joyner MJ, Senefeld JW. Outpatient treatment with concomitant vaccine-boosted convalescent plasma for patients with immunosuppression and COVID-19. mBio 2024; 15:e0040024. [PMID: 38602414 PMCID: PMC11078006 DOI: 10.1128/mbio.00400-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 03/21/2024] [Indexed: 04/12/2024] Open
Abstract
Although severe coronavirus disease 2019 (COVID-19) and hospitalization associated with COVID-19 are generally preventable among healthy vaccine recipients, patients with immunosuppression have poor immunogenic responses to COVID-19 vaccines and remain at high risk of infection with SARS-CoV-2 and hospitalization. In addition, monoclonal antibody therapy is limited by the emergence of novel SARS-CoV-2 variants that have serially escaped neutralization. In this context, there is interest in understanding the clinical benefit associated with COVID-19 convalescent plasma collected from persons who have been both naturally infected with SARS-CoV-2 and vaccinated against SARS-CoV-2 ("vax-plasma"). Thus, we report the clinical outcome of 386 immunocompromised outpatients who were diagnosed with COVID-19 and who received contemporary COVID-19-specific therapeutics (standard-of-care group) and a subgroup who also received concomitant treatment with very high titer COVID-19 convalescent plasma (vax-plasma group) with a specific focus on hospitalization rates. The overall hospitalization rate was 2.2% (5 of 225 patients) in the vax-plasma group and 6.2% (10 of 161 patients) in the standard-of-care group, which corresponded to a relative risk reduction of 65% (P = 0.046). Evidence of efficacy in nonvaccinated patients cannot be inferred from these data because 94% (361 of 386 patients) of patients were vaccinated. In vaccinated patients with immunosuppression and COVID-19, the addition of vax-plasma or very high titer COVID-19 convalescent plasma to COVID-19-specific therapies reduced the risk of disease progression leading to hospitalization.IMPORTANCEAs SARS-CoV-2 evolves, new variants of concern (VOCs) have emerged that evade available anti-spike monoclonal antibodies, particularly among immunosuppressed patients. However, high-titer COVID-19 convalescent plasma continues to be effective against VOCs because of its broad-spectrum immunomodulatory properties. Thus, we report clinical outcomes of 386 immunocompromised outpatients who were treated with COVID-19-specific therapeutics and a subgroup also treated with vaccine-boosted convalescent plasma. We found that the administration of vaccine-boosted convalescent plasma was associated with a significantly decreased incidence of hospitalization among immunocompromised COVID-19 outpatients. Our data add to the contemporary data providing evidence to support the clinical utility of high-titer convalescent plasma as antibody replacement therapy in immunocompromised patients.
Collapse
Affiliation(s)
- Juan G. Ripoll
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Anthony A. Stephenson
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Shane Ford
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Marsha L. Pike
- Department of Nursing, Mayo Clinic, Rochester, Rochester, Minnesota, USA
| | - Ellen K. Gorman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Sara N. Hanson
- Department of Family Medicine, Mayo Clinic Health Care System, Mankato, Minnesota, USA
| | - Justin E. Juskewitch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Alex J. Miller
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Solomiia Zaremba
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Erik A. Ovrom
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Raymund R. Razonable
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ravindra Ganesh
- Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Ryan T. Hurt
- Division of General Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Erin N. Fischer
- Department of Nursing, Mayo Clinic, Rochester, Rochester, Minnesota, USA
| | - Amber N. Derr
- Division of Hematology and Infusion Therapy, Rochester, Minnesota, USA
| | - Michele R. Eberle
- Mayo Clinic Health System Northwest Wisconsin, Eau Claire, Wisconsin, USA
| | | | | | - Elitza S. Theel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, USA
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, USA
- Department of Health and Kinesiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
36
|
Trepl J, Pasin C, Schneidawind D, Mueller NJ, Manz MG, Bankova AK, Abela IA. Evaluating tixagevimab/cilgavimab prophylaxis in allogeneic haematopoietic cell transplantation recipients for COVID-19 prevention. Br J Haematol 2024; 204:1908-1912. [PMID: 38327109 DOI: 10.1111/bjh.19321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
Allogeneic haematopoietic cell transplantation (allo-HCT) recipients exhibit an increased risk of COVID-19, particularly in the early post-transplant phase, due to insufficient vaccine responses. This retrospective study investigated the incidence of SARS-CoV-2 infection in allo-HCT recipients who received tixagevimab/cilgavimab pre-exposure prophylaxis (T/C PrEP) compared to those who did not. Logistic regression, adjusted for sex, age, SARS-CoV-2 vaccination status and immunosuppressive treatment, revealed a significant reduction in the likelihood of SARS-CoV-2 infection risk with T/C PrEP (adjusted odds ratio aOR = 0.26 [0.07, 0.91]). These findings suggest the potential efficacy of monoclonal antibody PrEP in protecting this vulnerable patient population from COVID-19.
Collapse
Affiliation(s)
- Julia Trepl
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Chloé Pasin
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Dominik Schneidawind
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Nicolas J Mueller
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Andriyana K Bankova
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
- National Specialized Hospital for Hematological Diseases, Sofia, Bulgaria
| | - Irene A Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
37
|
Billi B, Cholley P, Grobost V, Clément M, Rieu V, Le Guenno G, Lobbes H. Intravenous immunoglobulins for the treatment of prolonged COVID-19 in immunocompromised patients: a brief report. Front Immunol 2024; 15:1399180. [PMID: 38707896 PMCID: PMC11069322 DOI: 10.3389/fimmu.2024.1399180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Primary humoral deficiency and secondary B-cell depletion may lead to prolonged Sars-Cov-2 infection due to a decreased viral clearance. Prolonged infection is mainly driven by the lack of anti-Sars-Cov-2 immunoglobulin (IVIg) especially in patients with no vaccine response. Anti-spike immunoglobulin can be provided by infusion of convalescent patients' plasma: recent studies highlighted that commercial immunoglobulin show high titers of neutralizing IgG. We conducted a single center retrospective cohort. We included 9 patients (6 males, median age 74 years old): one patient with X-linked agammaglobulinemia and 8 patients treated with rituximab (2 granulomatosis with polyangiitis, 1 neuromyelitis optica, 4 low grade B-cell lymphoma and 1 EBV post-transplant lymphoproliferative disorder). Mean serum globulin was 4 ± 1.6 g/L. 7/8 had received at least 3 doses of mRNA anti-Sars-Cov-2 vaccine (median 4) with no response (anti-Spike IgG 0 for 6 patients). In this specific population requiring oxygen therapy but no intensive care support, the administration of IVIg was well tolerated and provided a swift improvement of clinical status, a significant decrease of inflammation associated to the an improvement of radiological patterns. Our results suggest that immunoglobulin could be used as a salvage therapy as an alternative to convalescent plasma but highly stringent patient selection is required due to the worldwide shortage of IVIg.
Collapse
Affiliation(s)
- Bénédicte Billi
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Paul Cholley
- Service de Radiologie, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Vincent Grobost
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Mélissa Clément
- Service de Médecine Interne, Hôpital Henri Mondor, Aurillac, France
| | - Virginie Rieu
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Guillaume Le Guenno
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Hervé Lobbes
- Service de Médecine Interne, Hôpital Estaing, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Institut Pascal, Unité Mixte de Recherche (UMR) 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
38
|
Yalan Q, Lingfang H, Xisong L, Run L, Junjing Z, An Z. Treatment for Covid-19 with SARS-CoV-2 neutralizing antibody BRII-196(Ambavirumab) plus BRII-198(Lomisivir): a retrospective cohort study. BMC Pharmacol Toxicol 2024; 25:29. [PMID: 38641625 PMCID: PMC11027409 DOI: 10.1186/s40360-024-00753-7] [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: 07/15/2023] [Accepted: 04/10/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Monoclonal antibody therapy for Covid-19 springs up all over the world and get some efficiency. This research aims to explore the treating effect of BRII-196(Ambavirumab) plus BRII-198(Lomisivir) on Covid-19. METHODS In this retrospective cohort research, patients received standard care or plus BRII-196 /BRII-198 monoclonal antibodies. General comparison of clinical indexes and prognosis between Antibody Group and Control Group was made. Further, according to the antibody using time and patients' condition, subgroups included Early antibody group, Late antibody group, Mild Antibody Group, Mild Control Group, Severe Antibody Group and Severe Control Group. RESULTS Length of stay(LOS) and interval of Covid-19 nucleic acid from positive to negative of Antibody Group were 12.0(IQR 9.0-15.0) and 14.0(IQR 10.0-16.0) days, less than those(13.0 (IQR 11.0-18.0) and 15.0 (IQR 12.8-17.0) days) of Control Group(p = 0.004, p = 0.004). LOS(median 10days) of Early Antibody Group was the shortest, significantly shorter than that of Control Group (median 13days)(p < 0.001). Interval(median 12days) of Covid-19 nucleic acid from positive to negative of Early Antibody Group also was significantly shorter than that of Control Group(median 15days) and Late Antibody Group(median 14days)(p = 0.001, p = 0.042). LOS(median 12days) and interval(median 13days) of Covid-19 nucleic acid from positive to negative of Mild Antibody Group was shorter than that of Mild Control Group(median 13days; median 14.5days)(p = 0.018, p = 0.033). CONCLUSION The neutralizing antibody therapy, BRII-196 plus BRII-198 could shorten LOS and interval of Covid-19 nucleic acid from positive to negative. However, it didn't show efficacy for improving clinical outcomes among severe or critical cases.
Collapse
Affiliation(s)
- Qin Yalan
- Department of Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76# Linjiang Road, Yuzhong District, 400016, Chongqing, China
| | - Hao Lingfang
- Department of Oncology, The Hohhot First Hospital, 010030, Hohhot, China
| | - Liu Xisong
- Department of Critical Care Medicine, Chongqing Public Health Treatment Center, 400030, Chongqing, China
| | - Liang Run
- Department of Oncology, The Hohhot First Hospital, 010030, Hohhot, China
| | - Zhang Junjing
- Department of Hepatobiliary Surgery, The Hohhot First Hospital, 150# South Second Ring Road, Yuquan District, Inner Mongolia Autonomous Region, 010030, Hohhot, China.
| | - Zhang' An
- Department of Critical Care Medicine, The Second Affiliated Hospital of Chongqing Medical University, 76# Linjiang Road, Yuzhong District, 400016, Chongqing, China.
| |
Collapse
|
39
|
Destremau M, Chaussade H, Hemar V, Beguet M, Bellecave P, Blanchard E, Barret A, Laboure G, Vasco-Moynet C, Lacassin F, Morisse E, Aguilar C, Lafarge X, Lafon ME, Bonnet F, Issa N, Camou F. Convalescent plasma transfusion for immunocompromised viremic patients with COVID-19: A retrospective multicenter study. J Med Virol 2024; 96:e29603. [PMID: 38619025 DOI: 10.1002/jmv.29603] [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: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/03/2024] [Indexed: 04/16/2024]
Abstract
This study aims to assess the safety, virological, and clinical outcomes of convalescent plasma transfusion (CPT) in immunocompromised patients hospitalized for coronavirus disease 2019 (COVID-19). We conducted a retrospective multicenter cohort study that included all immunosuppressed patients with COVID-19 and RNAemia from May 2020 to March 2023 treated with CPT. We included 81 patients with hematological malignancies (HM), transplants, or autoimmune diseases (69% treated with anti-CD20). Sixty patients (74%) were vaccinated, and 14 had pre-CPT serology >264 BAU/mL. The median delay between symptom onset and CPT was 23 days [13-31]. At D7 post-CPT, plasma PCR was negative in 43/64 patients (67.2%), and serology became positive in 25/30 patients (82%). Post-CPT positive serology was associated with RNAemia negativity (p < 0.001). The overall mortality rate at D28 was 26%, being higher in patients with non-B-cell HM (62%) than with B-cell HM (25%) or with no HM (11%) (p = 0.02). Patients receiving anti-CD20 without chemotherapy had the lowest mortality rate (8%). Positive RNAemia at D7 was associated with mortality at D28 in univariate analysis (HR: 3.05 [1.14-8.19]). Eight patients had adverse events, two of which were severe but transient. Our findings suggest that CPT can abolish RNAemia and ameliorate the clinical course in immunocompromised patients with COVID-19.
Collapse
Affiliation(s)
- Marjolaine Destremau
- CHU Bordeaux, Service de médecine interne et maladies infectieuses, Bordeaux, France
| | - Hélène Chaussade
- CHU Bordeaux, Service de médecine interne et maladies infectieuses, Bordeaux, France
| | - Victor Hemar
- CHU Bordeaux, Service de médecine interne et maladies infectieuses, Bordeaux, France
| | - Mathilde Beguet
- Etablissement français du sang Nouvelle Aquitaine, Bordeaux, France
| | | | | | - Amaury Barret
- CH Arcachon, Service de médecine interne, La Teste-de-Buch, France
| | | | | | - Flore Lacassin
- CH Mont-de-Marsan, Service de médecine interne, Mont-de-Marsan, France
| | | | - Claire Aguilar
- CH Périgueux, Service de maladies infectieuses, Périgueux, France
| | - Xavier Lafarge
- Etablissement français du sang Nouvelle Aquitaine, Bordeaux, France
- Université de Bordeaux, INSERM U1211 "Maladies Rares: Génétique et Métabolisme", Talence, France
| | | | - Fabrice Bonnet
- CHU Bordeaux, Service de médecine interne et maladies infectieuses, Bordeaux, France
- Université de Bordeaux, Bordeaux Population Health, INSERM U1219, Bordeaux, France
| | - Nahéma Issa
- CHU Bordeaux, Service de réanimation médicale, Bordeaux, France
| | - Fabrice Camou
- CHU Bordeaux, Service de réanimation médicale, Bordeaux, France
| |
Collapse
|
40
|
Huygens S, Preijers T, Swaneveld FH, Kleine Budde I, GeurtsvanKessel CH, Koch BCP, Rijnders BJA. Dosing of Convalescent Plasma and Hyperimmune Anti-SARS-CoV-2 Immunoglobulins: A Phase I/II Dose-Finding Study. Clin Pharmacokinet 2024; 63:497-509. [PMID: 38427270 PMCID: PMC11052786 DOI: 10.1007/s40262-024-01351-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/28/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND AND OBJECTIVE During the COVID-19 pandemic, trials on convalescent plasma (ConvP) were performed without preceding dose-finding studies. This study aimed to assess potential protective dosing regimens by constructing a population pharmacokinetic (popPK) model describing anti-SARS-CoV-2 antibody titers following the administration of ConvP or hyperimmune globulins (COVIg). METHODS Immunocompromised patients, testing negative for anti-SARS-CoV-2 spike antibodies despite vaccination, received a range of anti-SARS-CoV-2 antibodies in the form of COVIg or ConvP infusion. The popPK analysis was performed using NONMEM v7.4. Monte Carlo simulations were performed to assess potential COVIg and ConvP dosing regimens for prevention of COVID-19. RESULTS Forty-four patients were enrolled, and data from 42 were used for constructing the popPK model. A two-compartment elimination model with mixed residual error best described the Nab-titers after administration. Inter-individual variation was associated to CL (44.3%), V1 (27.3%), and V2 (29.2%). Lean body weight and type of treatment (ConvP/COVIg) were associated with V1 and V2, respectively. Median elimination half-life was 20 days (interquartile range: 17-25 days). Simulations demonstrated that even monthly infusions of 600 mL of the ConvP or COVIg used in this trial would not achieve potentially protective serum antibody titers for > 90% of the time. However, as a result of hybrid immunity and/or repeated vaccination, plasma donors with extremely high antibody titers are now readily available, and a > 90% target attainment should be possible. CONCLUSION The results of this study may inform future intervention studies on the prophylactic and therapeutic use of antiviral antibodies in the form of ConvP or COVIg. CLINICAL TRIAL REGISTRATION NUMBER NL9379 (The Netherlands Trial Register).
Collapse
Affiliation(s)
- Sammy Huygens
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Tim Preijers
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Francis H Swaneveld
- Unit of Transfusion Medicine, Sanquin Blood Supply Foundation, 1066 CX, Amsterdam, The Netherlands
| | - Ilona Kleine Budde
- Clinical Operations, Prothya Biosolutions, 1066 CX, Amsterdam, The Netherlands
| | - Corine H GeurtsvanKessel
- Department of Viroscience, Erasmus University Medical Center Rotterdam, WHO Collaborating Centre for Arbovirus and Viral Hemorrhagic Fever Reference and Research, Rotterdam, The Netherlands
| | - Birgit C P Koch
- Department of Hospital Pharmacy, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
- Rotterdam Clinical Pharmacometrics group, Rotterdam, The Netherlands
| | - Bart J A Rijnders
- Department of Internal Medicine, Section of Infectious Diseases and Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Willis ZI, Oliveira CR, Abzug MJ, Anosike BI, Ardura MI, Bio LL, Boguniewicz J, Chiotos K, Downes K, Grapentine SP, Hersh AL, Heston SM, Hijano DR, Huskins WC, James SH, Jones S, Lockowitz CR, Lloyd EC, MacBrayne C, Maron GM, Hayes McDonough M, Miller CM, Morton TH, Olivero RM, Orscheln RC, Schwenk HT, Singh P, Soma VL, Sue PK, Vora SB, Nakamura MM, Wolf J. Guidance for prevention and management of COVID-19 in children and adolescents: A consensus statement from the Pediatric Infectious Diseases Society Pediatric COVID-19 Therapies Taskforce. J Pediatric Infect Dis Soc 2024; 13:159-185. [PMID: 38339996 PMCID: PMC11494238 DOI: 10.1093/jpids/piad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 12/27/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Since November 2019, the SARS-CoV-2 pandemic has created challenges for preventing and managing COVID-19 in children and adolescents. Most research to develop new therapeutic interventions or to repurpose existing ones has been undertaken in adults, and although most cases of infection in pediatric populations are mild, there have been many cases of critical and fatal infection. Understanding the risk factors for severe illness and the evidence for safety, efficacy, and effectiveness of therapies for COVID-19 in children is necessary to optimize therapy. METHODS A panel of experts in pediatric infectious diseases, pediatric infectious diseases pharmacology, and pediatric intensive care medicine from 21 geographically diverse North American institutions was re-convened. Through a series of teleconferences and web-based surveys and a systematic review with meta-analysis of data for risk factors, a guidance statement comprising a series of recommendations for risk stratification, treatment, and prevention of COVID-19 was developed and refined based on expert consensus. RESULTS There are identifiable clinical characteristics that enable risk stratification for patients at risk for severe COVID-19. These risk factors can be used to guide the treatment of hospitalized and non-hospitalized children and adolescents with COVID-19 and to guide preventative therapy where options remain available.
Collapse
Affiliation(s)
- Zachary I Willis
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Carlos R Oliveira
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Mark J Abzug
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Brenda I Anosike
- Department of Pediatrics, The Children’s Hospital at Montefiore and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Monica I Ardura
- Department of Pediatrics, ID Host Defense Program, Nationwide Children’s Hospital & The Ohio State University, Columbus, OH, USA
| | - Laura L Bio
- Department of Pharmacy, Lucile Packard Children’s Hospital, Stanford, CA, USA
| | - Juri Boguniewicz
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, USA
| | - Kathleen Chiotos
- Departments of Anesthesiology, Critical Care Medicine, and Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Divisions of Critical Care Medicine and Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kevin Downes
- Department of Pediatrics, Perelman School of Medicine of the University of Pennsylvania, Division of Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Steven P Grapentine
- Department of Pharmacy, University of California San Francisco Benioff Children’s Hospital, San Francisco, CA, USA
| | - Adam L Hersh
- Department of Pediatrics, Division of Infectious Diseases, University of Utah, Salt Lake City, UT, USA
| | - Sarah M Heston
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Diego R Hijano
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - W Charles Huskins
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, USA
| | - Scott H James
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sarah Jones
- Department of Pharmacy, Boston Children’s Hospital, Boston, MA, USA
| | | | - Elizabeth C Lloyd
- Department of Pediatrics, University of Michigan, Ann Arbor, MI, USA
| | | | - Gabriela M Maron
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Molly Hayes McDonough
- Center for Healthcare Quality & Analytics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Christine M Miller
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Theodore H Morton
- Department of Pharmacy, St Jude’s Children’s Research Hospital, Memphis, Tennessee, USA
| | - Rosemary M Olivero
- Department of Pediatrics and Human Development, Michigan State College of Human Medicine and Helen DeVos Children’s Hospital of Corewell Health, Grand Rapids, MI, USA
| | | | - Hayden T Schwenk
- Department of Pediatrics, Stanford School of Medicine, Stanford, CA, USA
| | - Prachi Singh
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Vijaya L Soma
- Department of Pediatrics, NYU Grossman School of Medicine, New York, NY, USA
| | - Paul K Sue
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Surabhi B Vora
- Department of Pediatrics, University of Washington School of Medicine, and Division of Infectious Diseases, Seattle Children’s Hospital, Seattle, WA, USA
| | - Mari M Nakamura
- Antimicrobial Stewardship Program and Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
| | - Joshua Wolf
- Department of Infectious Diseases, St. Jude Children’s Research Hospital and Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
42
|
Fihn SD, Rivara FP. JAMA Network Open-The Year in Review, 2023. JAMA Netw Open 2024; 7:e246541. [PMID: 38502132 DOI: 10.1001/jamanetworkopen.2024.6541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2024] Open
Affiliation(s)
- Stephan D Fihn
- Deputy Editor, JAMA Network Open
- Department of Medicine, University of Washington, Seattle
| | - Frederick P Rivara
- Editor in Chief, JAMA Network Open
- Department of Pediatrics, University of Washington, Seattle
| |
Collapse
|
43
|
Calderón-Parra J, Gutiérrez-Villanueva A, Ronda-Roca G, Jimenez MLM, de la Torre H, Ródenas-Baquero M, Paniura-Pinedo M, Lozano-Llano C, Pintos-Pascual I, Fernández-Cruz A, Ramos-Martínez A, Muñez-Rubio E. Efficacy and safety of antiviral plus anti-spike monoclonal antibody combination therapy vs. monotherapy for high-risk immunocompromised patients with mild-to-moderate SARS-CoV2 infection during the Omicron era: A prospective cohort study. Int J Antimicrob Agents 2024; 63:107095. [PMID: 38244814 DOI: 10.1016/j.ijantimicag.2024.107095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
INTRODUCTION Antivirals and monoclonal antibodies lower the risk of progression in immunocompromised patients. However, combination therapy with both types of agents has not been studied. PATIENTS AND METHODS This was a single-centre, prospective, cohort study. All immunocompromised patients who received treatment for mild-to-moderate COVID-19 from 1 January 2022 to 30 October 2022 were enrolled. The primary endpoint was COVID-19 progression at 90 days, defined as hospital admission or death due to COVID-19 and/or seronegative persistent COVID-19. RESULTS A total of 304 patients were included: 43 patients (14.1%) received sotrovimab plus a direct-acting antiviral, and 261 (85.9%) received monotherapy. Primary outcome occurred more frequently after monotherapy (4.6% vs. 0%, P=0.154). Among patients with anti-spike immunoglobulin G (anti-S IgG) titre <750 BAU/mL, COVID-19 progression was more common after monotherapy (23.9% vs. 0%, P=0.001), including more frequent COVID-related admission (15.2% vs. 0%, P=0.014) and seronegative persistent COVID-19 (10.9% vs. 0%, P=0.044). Combination therapy was associated with lower risk of progression (odds ratio [OR] 0.08, 95% confidence interval [95% CI] 0.01-0.64). Anti-S IgG titre <750 BAU/mL and previous anti-CD20 were associated with higher risk of progression (OR 13.70, 95% CI 2.77-67.68; and OR 3.05, 95% CI 1.20-10.94, respectively). CONCLUSIONS In immunocompromised patients, combination therapy with sotrovimab plus an antiviral may be more effective than monotherapy for SARS-CoV2.
Collapse
Affiliation(s)
- Jorge Calderón-Parra
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain
| | - Andrea Gutiérrez-Villanueva
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain.
| | - Gerard Ronda-Roca
- Pharmacology department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | | | - Helena de la Torre
- Emergency department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - María Ródenas-Baquero
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - María Paniura-Pinedo
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Carla Lozano-Llano
- Pharmacy department, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Ilduara Pintos-Pascual
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain
| | - Ana Fernández-Cruz
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain; Autónoma University of Madrid, Spain
| | - Antonio Ramos-Martínez
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain; Autónoma University of Madrid, Spain
| | - Elena Muñez-Rubio
- Infectious Diseases Unit, Department of Internal Medicine, University Hospital Puerta de Hierro, Majadahonda, Spain; Research Institute Puerta de Hierro-Segovia de Arana (IDIPHSA), Majadahonda, Spain
| |
Collapse
|
44
|
Inbar T, Dann EJ, Kerner O, Stern A. Charting a path forward: Promising outcomes of convalescent plasma therapy in the care of severely B-cell depleted patients with persistent COVID-19. Transfusion 2024; 64:443-448. [PMID: 38327238 DOI: 10.1111/trf.17737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/24/2023] [Accepted: 12/24/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Patients with severe B-cell depletion related to hematological malignancies or B-cell targeted therapy suffer from impaired antibody responses to SARS-CoV-2 and are at risk for prolonged COVID-19. In this population, COVID-19 convalescent plasma (CCP) may provide passive immunity, enhance immune response, and promote virus neutralization. This study evaluated outcomes of B-cell depleted patients with persistent COVID-19 treated with CCP. STUDY DESIGN AND METHODS This analysis included all consecutive severely B-cell depleted patients with persistent COVID-19, receiving CCP at Rambam between 01.2022-02.2023. Persistent COVID-19 was defined as the presence of symptoms for ≥14 days in patients with negative SARS-CoV-2 nucleocapsid antibody test results. RESULTS Twenty patients met inclusion criteria, 17 of whom had hematological malignancies, two suffered from rheumatoid arthritis and one had both. Twelve patients received anti-CD-20 treatment, one - CAR-T cells and three underwent stem cell transplantation. The median duration of COVID-19 symptoms was 27.5 days (range 14-97); 12 patients had mild-to-moderate COVID-19 and 8 had severe infection. Sixteen patients required hospitalization. The majority of patients received other COVID-19 therapies before CCP. Within a median of two days (range 1-16) post-infusion, 19/20 patients clinically improved. No CCP-associated adverse events were documented. COVID-19 symptoms recurred in 3 of the improved patients. Two patients died from COVID-19 on days 1 and 90 following the first CCP infusion. DISCUSSION In severely B-cell depleted patients with persistent COVID-19, CCP is safe and associated with rapid clinical improvement. This subset of immunocompromised patients could particularly benefit from CCP administration.
Collapse
Affiliation(s)
- Tsofia Inbar
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- Blood Bank and Apheresis Unit, Rambam Health Care Campus, Haifa, Israel
| | - Eldad J Dann
- Department of Hematology and Bone Marrow Transplantation, Rambam Health Care Campus, Haifa, Israel
- Blood Bank and Apheresis Unit, Rambam Health Care Campus, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Omer Kerner
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
| | - Anat Stern
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion, Israel Institute of Technology, Haifa, Israel
- Division of Infectious Diseases, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
45
|
Franchini M, Focosi D, Cruciani M, Joyner MJ, Pirofski LA, Senefeld JW, Shoham S, Sullivan DJ, Casadevall A. Safety and Efficacy of Convalescent Plasma Combined with Other Pharmaceutical Agents for Treatment of COVID-19 in Hospitalized Patients: A Systematic Review and Meta-Analysis. Diseases 2024; 12:41. [PMID: 38534965 PMCID: PMC10969279 DOI: 10.3390/diseases12030041] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 02/12/2024] [Accepted: 02/20/2024] [Indexed: 01/06/2025] Open
Abstract
Plasma collected from people recovered from COVID-19 (COVID-19 convalescent plasma, CCP) was the first antibody-based therapy employed to fight the pandemic. CCP was, however, often employed in combination with other drugs, such as the antiviral remdesivir and glucocorticoids. The possible effect of such interaction has never been investigated systematically. To assess the safety and efficacy of CCP combined with other agents for treatment of patients hospitalized for COVID-19, a systematic literature search using appropriate Medical Subject Heading (MeSH) terms was performed through PubMed, EMBASE, Cochrane central, medRxiv and bioRxiv. The main outcomes considered were mortality and safety of CCP combined with other treatments versus CCP alone. This review was carried out in accordance with Cochrane methodology including risk of bias assessment and grading of the quality of evidence. Measure of treatment effect was the risk ratio (RR) together with 95% confidence intervals (CIs). A total of 11 studies (8 randomized controlled trials [RCTs] and 3 observational) were included in the systematic review, 4 studies with CCP combined with remdesivir and 6 studies with CCP combined with corticosteroids, all involving hospitalized patients. One RCT reported information on both remdesivir and steroids use with CCP. The use of CCP combined with remdesivir was associated with a significantly reduced risk of death (RR 0.74; 95% CI 0.56-0.97; p = 0.03; moderate certainty of evidence), while the use of steroids with CCP did not modify the mortality risk (RR 0.72; 95% CI 0.34-1.51; p = 0.38; very low certainty of evidence). Not enough safety data were retrieved form the systematic literature analysis. The current evidence from the literature suggests a potential beneficial effect on mortality of combined CCP plus remdesivir compared to CCP alone in hospitalized COVID-19 patients. No significant clinical interaction was found between CCP and steroids.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy;
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy;
| | - Mario Cruciani
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy;
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY 10467, USA;
| | - Jonathon W. Senefeld
- Department of Kinesiology and Community Healthy, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA;
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MA 21205, USA;
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (D.J.S.); (A.C.)
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (D.J.S.); (A.C.)
| |
Collapse
|
46
|
Vita S, D’Abramo A, Coppola A, Farroni C, Iori AP, Faraglia F, Sette A, Grifoni A, Lindestam Arlehamn C, Bibas M, Goletti D, Nicastri E. Combined antiviral therapy as effective and feasible option in allogenic hematopoietic stem cell transplantation during SARS-COV-2 infection: a case report. Front Oncol 2024; 14:1290614. [PMID: 38414746 PMCID: PMC10896944 DOI: 10.3389/fonc.2024.1290614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 01/29/2024] [Indexed: 02/29/2024] Open
Abstract
Here we describe the case of a 51 years old Italian woman with acute lymphoblastic leukemia who underwent to hematopoietic stem cell transplantation (HSCT) during SARS-COV-2 infection. She presented a prolonged COVID-19 successfully treated with dual anti SARS-COV-2 antiviral plus monoclonal antibody therapy.
Collapse
Affiliation(s)
- Serena Vita
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alessandra D’Abramo
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Coppola
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Chiara Farroni
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Paola Iori
- Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Francesca Faraglia
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Cecilia Lindestam Arlehamn
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Michele Bibas
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Delia Goletti
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Emanuele Nicastri
- Clinical Department, National Institute for Infectious Diseases ‘Lazzaro Spallanzani’ Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
47
|
Gentile S, Sullivan LR, Brooks H, Simeunovic G. A Descriptive, Retrospective Analysis of COVID-19 Passive Antibody Therapy and Its Effects on Morbidity and Mortality in Patients Receiving B-Cell-Depleting Therapies. Diseases 2024; 12:33. [PMID: 38391780 PMCID: PMC10887790 DOI: 10.3390/diseases12020033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/30/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
Patients receiving B-cell-depleting therapies (BCDT) are at an increased risk for severe COVID-19. Passive antibody therapy (PAT), including COVID-19 convalescent plasma (CCP) and monoclonal antibodies (mAb), may be an effective treatment in this population. Real-world data on PAT effectiveness are limited. To evaluate response to PAT measured through 90-day all-cause morbidity and mortality, we performed a retrospective review of patients who contracted COVID-19 within a year from the last BCDT. From 64 included patients, the majority were Caucasians (95%), female (56%), vaccinated (67%), treated outpatients (64%), with multiple comorbidities. Examined BCDT were rituximab (55%), obinutuzumab (33%), ocrelizumab (11%) and ofatumumab (1%), used for underlying hematological malignancy (HEM) (40%), multiple sclerosis (34%), and rheumatoid arthritis (16%). Of seven deceased patients, three died from COVID-19. All three were elderly males with multiple comorbidities, treated inpatient for severe COVID-19. Four of 41 patients treated as outpatients were hospitalized for non-COVID-19-related reasons. All deceased and hospitalized patients had an underlying HEM. All but one were on rituximab. PAT may be an effective treatment for patients receiving BCDT, especially if given early for non-severe disease. Patients with underlying HEM may be at increased risk for severe disease compared with others receiving the same BCDT.
Collapse
Affiliation(s)
- Sonia Gentile
- Department of Internal Medicine and Pediatrics, Corewell Health, Grand Rapids, MI 49503, USA
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
| | - Liam R Sullivan
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Infectious Disease, Corewell Health, Grand Rapids, MI 49503, USA
| | - Heather Brooks
- Office of Research and Education, Corewell Health, Grand Rapids, MI 49503, USA
| | - Gordana Simeunovic
- College of Human Medicine, Michigan State University, Grand Rapids, MI 49503, USA
- Department of Infectious Disease, Corewell Health, Grand Rapids, MI 49503, USA
| |
Collapse
|
48
|
Franchini M, Cruciani M, Casadevall A, Joyner MJ, Senefeld JW, Sullivan DJ, Zani M, Focosi D. Safety of COVID-19 convalescent plasma: A definitive systematic review and meta-analysis of randomized controlled trials. Transfusion 2024; 64:388-399. [PMID: 38156374 DOI: 10.1111/trf.17701] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 12/17/2023] [Accepted: 12/18/2023] [Indexed: 12/30/2023]
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Mario Cruciani
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Arturo Casadevall
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, Maryland, USA
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Jonathon W Senefeld
- Department of Kinesiology and Community Healthy, University of Illinois at Urbana-Champaign, Champaign, Illinois, USA
| | - David J Sullivan
- Johns Hopkins Bloomberg School of Public Health, Department of Molecular Microbiology and Immunology, Baltimore, Maryland, USA
| | - Matteo Zani
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, Mantova, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| |
Collapse
|
49
|
Franchini M, Focosi D. Hyperimmune Plasma and Immunoglobulins against COVID-19: A Narrative Review. Life (Basel) 2024; 14:214. [PMID: 38398723 PMCID: PMC10890293 DOI: 10.3390/life14020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Since late 2019, the new SARS-CoV-2 virus belonging to the Coronaviridae family has been responsible for COVID-19 pandemic, a severe acute respiratory syndrome. Several antiviral therapies, mostly derived from previous epidemics, were initially repurposed to fight this not rarely life-threatening respiratory illness. Among them, however, the only specific antibody-based therapy available against SARS-CoV-2 infection during the first year of the pandemic was represented by COVID-19 convalescent plasma (CCP). CCP, collected from recovered individuals, contains high levels of polyclonal antibodies of different subclasses able to neutralize SARS-CoV-2 infection. Tens of randomized controlled trials have been conducted during the last three years of the pandemic to evaluate the safety and the clinical efficacy of CCP in both hospitalized and ambulatory COVID-19 patients, whose main results will be summarized in this narrative review. In addition, we will present the current knowledge on the development of anti-SARS-CoV-2 hyperimmune polyclonal immunoglobulins.
Collapse
Affiliation(s)
- Massimo Franchini
- Department of Transfusion Medicine and Hematology, Carlo Poma Hospital, 46100 Mantua, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, 56124 Pisa, Italy;
| |
Collapse
|
50
|
Librizzi G, Modi V, Lier AJ. SARS-CoV-2 Persistence in Immunocompromised Patients Requiring Treatment With Convalescent Plasma: A Case Report. Cureus 2024; 16:e54564. [PMID: 38516449 PMCID: PMC10957151 DOI: 10.7759/cureus.54564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2024] [Indexed: 03/23/2024] Open
Abstract
Severe acute respiratory syndrome-2 (SARS-CoV-2) infection in immunocompromised patients presents a challenge, as patients with such conditions may have severe courses. Identifying modalities to shorten the course or lessen the severity of infection could be potentially beneficial. A 76-year-old male with follicular lymphoma on rituximab and lenalidomide presented with COVID-19 pneumonia requiring intensive care unit (ICU) level care for persistent hypoxemia. He was treated with an extended course of remdesivir, as recommended by the Infectious Diseases service, but he maintained a persistently high viral load, necessitating a delay of his cancer treatment until he had recovered from his infection. On hospital day 31, he was given one dose of convalescent plasma with improvement in his SARS-CoV-2 viral load. He was able to be discharged and resumed cancer treatment soon thereafter. Convalescent plasma is a potential therapeutic option for immunocompromised patients with SARS-CoV-2 infection and should be considered early in the hospital course. Additionally, cycle threshold monitoring may be beneficial in certain scenarios: for instance to guide consideration of alternative therapies in patients with severe COVID-19 who have persistent symptoms and viremia while on guideline-directed therapy.
Collapse
Affiliation(s)
- Gabrielle Librizzi
- Internal Medicine, Stony Brook University Hospital, Stony Brook, USA
- Internal Medicine, Northport Veterans Affairs Medical Center, Northport, USA
| | - Viraj Modi
- Internal Medicine, Northport Veterans Affairs Medical Center, Northport, USA
| | - Audun J Lier
- Infectious Diseases, Northport Veterans Affairs Medical Center, Northport, USA
- Infectious Diseases, Stony Brook University Hospital, Stony Brook, USA
| |
Collapse
|