101
|
Kloypan C, Saesong M, Sangsuemoon J, Chantharit P, Mongkhon P. CONVALESCENT plasma for COVID-19: A meta-analysis of clinical trials and real-world evidence. Eur J Clin Invest 2021; 51:e13663. [PMID: 34375445 PMCID: PMC8420367 DOI: 10.1111/eci.13663] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 08/05/2021] [Accepted: 08/07/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND There is still a lack of consensus on the efficacy of convalescent plasma (CP) treatment in COVID-19 patients. We performed a systematic review and meta-analysis to investigate the efficacy of CP vs standard treatment/non-CP on clinical outcomes in COVID-19 patients. METHODS Cochrane Library, PubMed, EMBASE and ClinicalTrials.gov were searched from December 2019 to 16 July 2021, for data from clinical trials and observational studies. The primary outcome was all-cause mortality. Risk estimates were pooled using a random-effect model. Risk of bias was assessed by Cochrane Risk of Bias tool for clinical trials and Newcastle-Ottawa Scale for observational studies. RESULTS In total, 18 peer-reviewed clinical trials, 3 preprints and 26 observational studies met the inclusion criteria. In the meta-analysis of 18 peer-reviewed trials, CP use had a 31% reduced risk of all-cause mortality compared with standard treatment use (pooled risk ratio [RR] = 0.69, 95% confidence interval [CI]: 0.56-0.86, P = .001, I2 = 50.1%). Based on severity and region, CP treatment significantly reduced risk of all-cause mortality in patients with severe and critical disease and studies conducted in Asia, pooled RR = 0.61, 95% CI: 0.47-0.81, P = .001, I2 = 0.0%; pooled RR = 0.67, 95% CI: 0.49-0.92, P = .013, I2 = 0.0%; and pooled RR = 0.62, 95% CI: 0.48-0.80, P < .001, I2 = 20.3%, respectively. The meta-analysis of observational studies showed the similar results to the clinical trials. CONCLUSIONS Convalescent plasma use was associated with reduced risk of all-cause mortality in severe or critical COVID-19 patients. However, the findings were limited with a moderate degree of heterogeneity. Further studies with well-designed and larger sample size are needed.
Collapse
Affiliation(s)
- Chiraphat Kloypan
- Division of Clinical Immunology and Transfusion ScienceDepartment of Medical TechnologySchool of Allied Health SciencesUniversity of PhayaoPhayaoThailand
- Unit of Excellence in Integrative Molecular BiomedicineSchool of Allied Health SciencesUniversity of PhayaoPhayaoThailand
- Institute of Transfusion MedicineCharité Universitätsmedizin BerlinBerlinGermany
| | - Matthanaporn Saesong
- Division of Clinical Immunology and Transfusion ScienceDepartment of Medical TechnologySchool of Allied Health SciencesUniversity of PhayaoPhayaoThailand
| | - Juthamat Sangsuemoon
- Division of Clinical Immunology and Transfusion ScienceDepartment of Medical TechnologySchool of Allied Health SciencesUniversity of PhayaoPhayaoThailand
| | - Prawat Chantharit
- Division of Infectious DiseasesDepartment of MedicineFaculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Pajaree Mongkhon
- Division of Pharmacy PracticeDepartment of Pharmaceutical CareUnit of Excellence on Research in Health Outcomes and Patient Safety in ElderlySchool of Pharmaceutical SciencesUniversity of PhayaoPhayaoThailand
- Pharmacoepidemiology and Statistics Research CenterFaculty of PharmacyChiang Mai UniversityChiang MaiThailand
| |
Collapse
|
102
|
Menichetti F, Popoli P, Puopolo M, Spila Alegiani S, Tiseo G, Bartoloni A, De Socio GV, Luchi S, Blanc P, Puoti M, Toschi E, Massari M, Palmisano L, Marano G, Chiamenti M, Martinelli L, Franchi S, Pallotto C, Suardi LR, Luciani Pasqua B, Merli M, Fabiani P, Bertolucci L, Borchi B, Modica S, Moneta S, Marchetti G, d’Arminio Monforte A, Stoppini L, Ferracchiato N, Piconi S, Fabbri C, Beccastrini E, Saccardi R, Giacometti A, Esperti S, Pierotti P, Bernini L, Bianco C, Benedetti S, Lanzi A, Bonfanti P, Massari M, Sani S, Saracino A, Castagna A, Trabace L, Lanza M, Focosi D, Mazzoni A, Pistello M, Falcone M. Effect of High-Titer Convalescent Plasma on Progression to Severe Respiratory Failure or Death in Hospitalized Patients With COVID-19 Pneumonia: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2136246. [PMID: 34842924 PMCID: PMC8630572 DOI: 10.1001/jamanetworkopen.2021.36246] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
IMPORTANCE Convalescent plasma (CP) has been generally unsuccessful in preventing worsening of respiratory failure or death in hospitalized patients with COVID-19 pneumonia. OBJECTIVE To evaluate the efficacy of CP plus standard therapy (ST) vs ST alone in preventing worsening respiratory failure or death in patients with COVID-19 pneumonia. DESIGN, SETTING, AND PARTICIPANTS This prospective, open-label, randomized clinical trial enrolled (1:1 ratio) hospitalized patients with COVID-19 pneumonia to receive CP plus ST or ST alone between July 15 and December 8, 2020, at 27 clinical sites in Italy. Hospitalized adults with COVID-19 pneumonia and a partial pressure of oxygen-to-fraction of inspired oxygen (Pao2/Fio2) ratio between 350 and 200 mm Hg were eligible. INTERVENTIONS Patients in the experimental group received intravenous high-titer CP (≥1:160, by microneutralization test) plus ST. The volume of infused CP was 200 mL given from 1 to a maximum of 3 infusions. Patients in the control group received ST, represented by remdesivir, glucocorticoids, and low-molecular weight heparin, according to the Agenzia Italiana del Farmaco recommendations. MAIN OUTCOMES AND MEASURES The primary outcome was a composite of worsening respiratory failure (Pao2/Fio2 ratio <150 mm Hg) or death within 30 days from randomization. RESULTS Of the 487 randomized patients (241 to CP plus ST; 246 to ST alone), 312 (64.1%) were men; the median (IQR) age was 64 (54.0-74.0) years. The modified intention-to-treat population included 473 patients. The primary end point occurred in 59 of 231 patients (25.5%) treated with CP and ST and in 67 of 239 patients (28.0%) who received ST (odds ratio, 0.88; 95% CI, 0.59-1.33; P = .54). Adverse events occurred more frequently in the CP group (12 of 241 [5.0%]) compared with the control group (4 of 246 [1.6%]; P = .04). CONCLUSIONS AND RELEVANCE In patients with moderate to severe COVID-19 pneumonia, high-titer anti-SARS-CoV-2 CP did not reduce the progression to severe respiratory failure or death within 30 days. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04716556.
Collapse
Affiliation(s)
- Francesco Menichetti
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Patrizia Popoli
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Maria Puopolo
- Department of Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | - Giusy Tiseo
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Alessandro Bartoloni
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Giuseppe Vittorio De Socio
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Sauro Luchi
- Infectious Disease Unit, Hospital of Lucca, Lucca, Italy
| | - Pierluigi Blanc
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Massimo Puoti
- University of Milano-Bicocca School of Medicine, Milan, Italy
- Azienda socio sanitaria territorial (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Elena Toschi
- Research Coordination and Support Service (CoRi), Istituto Superiore di Sanità, Rome, Italy
| | - Marco Massari
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Lucia Palmisano
- National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | | | | | - Laura Martinelli
- Internal Medicine, Unità Sanitaria Locale (USL)–Umbria 1, Ospedale Città di Castello, Città di Castello, Italy
| | - Silvia Franchi
- Internal Medicine, Unità Sanitaria Locale (USL)–Umbria 1, Ospedale Città di Castello, Città di Castello, Italy
| | - Carlo Pallotto
- Infectious Diseases Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, Empoli, Italy
| | - Lorenzo Roberto Suardi
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
- Infectious Diseases Unit, San Giuseppe Hospital, Azienda USL Toscana Centro, Empoli, Italy
| | - Barbara Luciani Pasqua
- Centro Regionale Sangue, Servizio Immunotrasfusionale, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Marco Merli
- Azienda socio sanitaria territorial (ASST) Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Plinio Fabiani
- Internal Medicine, Ospedale Unico della Versilia, Lido di Camaiore, Italy
| | - Luca Bertolucci
- Internal Medicine, Ospedale Unico della Versilia, Lido di Camaiore, Italy
| | - Beatrice Borchi
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Sara Modica
- Infectious and Tropical Diseases Unit, Florence Department of Medicine, Careggi University Hospital, Florence, Italy
| | - Sara Moneta
- Infectious Disease Unit, Hospital of Lucca, Lucca, Italy
| | - Giulia Marchetti
- Infectious Diseases Unit, Department of Health Sciences, ASST Santi Paolo e Carlo University Hospital, Milan, Italy
| | - Antonella d’Arminio Monforte
- Infectious Diseases Unit, Department of Health Sciences, ASST Santi Paolo e Carlo University Hospital, Milan, Italy
| | | | | | - Stefania Piconi
- Infectious Diseases, Azienda Ospedaliera di Lecco, Lecco, Italy
| | - Claudio Fabbri
- Infectious Diseases, Ospedale San Jacopo, Pistoia, Italy
| | - Enrico Beccastrini
- Cell Therapy and Transfusion Medicine, Careggi University Hospital, Florence, Italy
| | - Riccardo Saccardi
- Cell Therapy and Transfusion Medicine, Careggi University Hospital, Florence, Italy
| | - Andrea Giacometti
- Azienda Ospedaliera Universitaria, Ospedali Riuniti di Ancona, Ancona, Italy
| | - Sara Esperti
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Piera Pierotti
- Infectious Diseases, Ospedale S. Maria Annunziata, Firenze, Italy
| | - Laura Bernini
- Division of Infectious Diseases, Arezzo Hospital, Arezzo, Italy
| | - Claudia Bianco
- Division of Infectious Diseases, Arezzo Hospital, Arezzo, Italy
| | - Sara Benedetti
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Alessandra Lanzi
- Clinic of Infectious Diseases, “Santa Maria della Misericordia” Hospital, University of Perugia, Perugia, Italy
| | - Paolo Bonfanti
- Department of Infectious Diseases, ASST Monza, University of Milano-Bicocca, Milan, Italy
| | - Marco Massari
- Infectious Disease Unit, Azienda USL–Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) di Reggio Emilia, Reggio Emilia, Italy
| | - Spartaco Sani
- Infectious Diseases, Livorno Hospital, Livorno, Italy
| | - Annalisa Saracino
- Division of Infectious Diseases, Bari University Hospital, Bari, Italy
| | - Antonella Castagna
- Infectious Diseases, IRCCS Ospedale San Raffaele, Università Vita-Salute San Raffaele, Milan, Italy
| | - Luigia Trabace
- Department of Experimental and Clinical Medicine, University of Foggia, Foggia, Italy
| | - Maria Lanza
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Alessandro Mazzoni
- Division of Transfusion Medicine and Transplant Biology, Pisa University Hospital, Pisa, Italy
| | - Mauro Pistello
- Division of Virology, University Hospital of Pisa, Retrovirus Center, Department of Translational Research, University of Pisa, Pisa, Italy
| | - Marco Falcone
- Infectious Disease Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| |
Collapse
|
103
|
Prevention and Treatment of SARS-CoV2 Infection in People Living with HIV: The Need for Specific Data. Infect Dis Ther 2021; 11:1-13. [PMID: 34709579 PMCID: PMC8552207 DOI: 10.1007/s40121-021-00547-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022] Open
Abstract
The HIV pandemic has led to close to 40 million people living with HIV (PLWH) worldwide. To date, SARS-CoV2 has affected > 220 million people, and unprecedented global efforts have resulted in almost 6000 million doses of SARS-CoV2 vaccines being administered. Although several specific COVID-19 antiviral and anti-inflammatory treatments and SARS-CoV2 vaccines have been approved, the data available to support their use in specific populations such as PLWH remain limited. PLWH includes a range of individuals from practically unaffected immunity to severely immunocompromised individuals, and preventive and therapeutic interventions should be tailored for these subgroups . However, in most randomized clinical trials regarding antivirals, immunomodulators and vaccines for COVID-19, PLWH have been excluded or only enrolled in small numbers leading to a paucity of data. We briefly discuss the current evidence for prevention and treatment of COVID-19 in PLWH and identify key areas where more information is required.
Collapse
|
104
|
Díez JM, Romero C, Cruz M, Vandeberg P, Merritt WK, Pradenas E, Trinité B, Blanco J, Clotet B, Willis T, Gajardo R. Anti-SARS-CoV-2 hyperimmune globulin demonstrates potent neutralization and antibody-dependent cellular cytotoxicity and phagocytosis through N and S proteins. J Infect Dis 2021; 225:938-946. [PMID: 34693968 PMCID: PMC8574314 DOI: 10.1093/infdis/jiab540] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/22/2021] [Indexed: 11/13/2022] Open
Abstract
Background Although coronavirus disease 2019 (COVID-19) vaccinations have provided a significant reduction in infections, effective COVID-19 treatments remain an urgent need. Methods Functional characterization of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) hyperimmune immunoglobulin (hIG) from human convalescent plasma was performed by different virus neutralization methodologies (plaque reduction, virus-induced cytotoxicity, median tissue culture infectious dose [TCID50] reduction, and immunofluorimetry) at different laboratories using geographically different SARS-CoV-2 isolates (USA [1], Italy [1], and Spain [2]; 2 containing the D614G mutation). Neutralization capacity against the original Wuhan SARS-CoV-2 strain and variants (D614G mutant, B.1.1.7, P.1, and B.1.351) was evaluated using a pseudovirus expressing the corresponding spike (S) protein. Antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP) was also evaluated. Results All SARS-CoV-2 isolates were potently neutralized by hIG as shown by all 4 methodologies. Wild-type SARS-CoV-2 and variants were effectively neutralized using the pseudovirus. The hIG (IgG type) induced ADCC and ADCP against SARS-CoV-2 N and S proteins but not E protein. Very low concentrations (25–100 µg IgG/mL) were required. A potent effect was triggered by antibodies in hIG solutions against the SARS-CoV-2 S and N proteins. Conclusions Beyond neutralization, IgG Fc-dependent pathways may play a role in combatting SARS-CoV-2 infections using COVID-19 hIG. This could be especially relevant for the treatment of more neutralization-resistant SARS-CoV-2 variants.
Collapse
Affiliation(s)
- José María Díez
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - Carolina Romero
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - María Cruz
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - Peter Vandeberg
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - W Keither Merritt
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - Edwards Pradenas
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, 08916, Badalona, Catalonia, Spain
| | - Benjamin Trinité
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, 08916, Badalona, Catalonia, Spain
| | - Julià Blanco
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, 08916, Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain
| | - Bonaventura Clotet
- IrsiCaixa AIDS Research Institute, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, UAB, 08916, Badalona, Catalonia, Spain.,University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain
| | - Todd Willis
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| | - Rodrigo Gajardo
- Bioscience Research & Development, Scientific Innovation Office, Grifols, Barcelona, Spain
| |
Collapse
|
105
|
Fouka E, Kalomenidis I, Gianniou N, Gida S, Steiropoulos P. COVID-19 Advanced Care. J Pers Med 2021; 11:1082. [PMID: 34834434 PMCID: PMC8621119 DOI: 10.3390/jpm11111082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 10/10/2021] [Accepted: 10/22/2021] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, related to the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has caused a worldwide sudden and substantial burden in public health due to an enormous increase in hospitalizations for pneumonia with the multiorgan disease. Treatment for individuals with COVID-19 includes best practices for supportive management of acute hypoxic respiratory failure. Emerging data indicate that dexamethasone therapy reduces 28-day mortality in patients requiring supplemental oxygen compared with usual care, and ongoing trials are testing the efficacy of antiviral therapies, immune modulators and anticoagulants in the prevention of disease progression and complications, while monoclonal antibodies and hyperimmune globulin may provide additional preventive strategies. Consensus suggestions can standardize care, thereby improving outcomes and facilitating future research. This review discusses current evidence regarding the pharmacotherapy of COVID-19.
Collapse
Affiliation(s)
- Evangelia Fouka
- Pulmonary Department, Aristotle University of Thessaloniki, G. Papanikolaou Hospital, 57010 Thessaloniki, Greece
| | - Ioannis Kalomenidis
- 1st Department of Critical Care and Pulmonary Medicine, Evaggelismos Hospital, National and Kapodistrian University of Athens, 10679 Athens, Greece; (I.K.); (N.G.)
| | - Niki Gianniou
- 1st Department of Critical Care and Pulmonary Medicine, Evaggelismos Hospital, National and Kapodistrian University of Athens, 10679 Athens, Greece; (I.K.); (N.G.)
| | - Sofia Gida
- Intensive Care Unit, General Hospital of Trikala, 42100 Trikala, Greece;
| | - Paschalis Steiropoulos
- Department of Respiratory Medicine, Medical School, Democritus University of Thrace, 67100 Alexandroupolis, Greece;
| |
Collapse
|
106
|
Abstract
As the coronavirus disease (COVID-19) pandemic led to a global health crisis, there were limited treatment options and no prophylactic therapies for those exposed to Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). Convalescent plasma is quick to implement, potentially provides benefits, and has a good safety profile. The therapeutic potential of COVID-19 convalescent plasma (CCP) is likely mediated by antibodies through direct viral neutralization and Fc-dependent functions such as a phagocytosis, complement activation, and antibody-dependent cellular cytotoxicity. In the United States, CCP became one of the most common treatments with over half million units transfused despite limited efficacy data. More than a dozen randomized trials now demonstrate that CCP does not provide benefit for those with moderate to severe disease. However, similar to other passive antibody therapies, CCP is beneficial for early disease, when provided to elderly outpatients within 72 hours after symptom onset. Only high-titer CCP should be transfused. CCP should also be considered for immunosuppressed COVID-19 patients. CCP collected in proximity, by time and location, to the patient may be more beneficial due to SARS-CoV-2 variants. Additional randomized trial data are still accruing and should be incorporated with other trial data to optimize CCP indications.
Collapse
|
107
|
Zhang C, Jin H, Wen YF, Yin G. Efficacy of COVID-19 Treatments: A Bayesian Network Meta-Analysis of Randomized Controlled Trials. Front Public Health 2021; 9:729559. [PMID: 34650951 PMCID: PMC8506153 DOI: 10.3389/fpubh.2021.729559] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/01/2021] [Indexed: 01/09/2023] Open
Abstract
Background: We provided a comprehensive evaluation of efficacy of available treatments for coronavirus disease 2019 (COVID-19). Methods: We searched for candidate COVID-19 studies in WHO COVID-19 Global Research Database up to August 19, 2021. Randomized controlled trials for suspected or confirmed COVID-19 patients published on peer-reviewed journals were included, regardless of demographic characteristics. Outcome measures included mortality, mechanical ventilation, hospital discharge and viral clearance. Bayesian network meta-analysis with fixed effects was conducted to estimate the effect sizes using posterior means and 95% equal-tailed credible intervals (CrIs). Odds ratio (OR) was used as the summary measure for treatment effect. Bayesian hierarchical models were used to estimate effect sizes of treatments grouped by the treatment classifications. Results: We identified 222 eligible studies with a total of 102,950 patients. Compared with the standard of care, imatinib, intravenous immunoglobulin and tocilizumab led to lower risk of death; baricitinib plus remdesivir, colchicine, dexamethasone, recombinant human granulocyte colony stimulating factor and tocilizumab indicated lower occurrence of mechanical ventilation; tofacitinib, sarilumab, remdesivir, tocilizumab and baricitinib plus remdesivir increased the hospital discharge rate; convalescent plasma, ivermectin, ivermectin plus doxycycline, hydroxychloroquine, nitazoxanide and proxalutamide resulted in better viral clearance. From the treatment class level, we found that the use of antineoplastic agents was associated with fewer mortality cases, immunostimulants could reduce the risk of mechanical ventilation and immunosuppressants led to higher discharge rates. Conclusions: This network meta-analysis identified superiority of several COVID-19 treatments over the standard of care in terms of mortality, mechanical ventilation, hospital discharge and viral clearance. Tocilizumab showed its superiority compared with SOC on preventing severe outcomes such as death and mechanical ventilation as well as increasing the discharge rate, which might be an appropriate treatment for patients with severe or mild/moderate illness. We also found the clinical efficacy of antineoplastic agents, immunostimulants and immunosuppressants with respect to the endpoints of mortality, mechanical ventilation and discharge, which provides valuable information for the discovery of potential COVID-19 treatments.
Collapse
Affiliation(s)
- Chenyang Zhang
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong SAR, China
| | - Huaqing Jin
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong SAR, China
| | - Yi Feng Wen
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China
| | - Guosheng Yin
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong SAR, China.,Department of Biostatistics, MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
108
|
Körper S, Weiss M, Zickler D, Wiesmann T, Zacharowski K, Corman VM, Grüner B, Ernst L, Spieth P, Lepper PM, Bentz M, Zinn S, Paul G, Kalbhenn J, Dollinger MM, Rosenberger P, Kirschning T, Thiele T, Appl T, Mayer B, Schmidt M, Drosten C, Wulf H, Kruse JM, Jungwirth B, Seifried E, Schrezenmeier H. Results of the CAPSID randomized trial for high-dose convalescent plasma in patients with severe COVID-19. J Clin Invest 2021; 131:e152264. [PMID: 34464358 DOI: 10.1172/jci152264] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 08/26/2021] [Indexed: 02/01/2023] Open
Abstract
BACKGROUNDCOVID-19 convalescent plasma (CCP) has been considered a treatment option for COVID-19. This trial assessed the efficacy of a neutralizing antibody containing high-dose CCP in hospitalized adults with COVID-19 requiring respiratory support or intensive care treatment.METHODSPatients (n = 105) were randomized 1:1 to either receive standard treatment and 3 units of CCP or standard treatment alone. Control group patients with progress on day 14 could cross over to the CCP group. The primary outcome was a dichotomous composite outcome of survival and no longer fulfilling criteria for severe COVID-19 on day 21.ResultsThe primary outcome occurred in 43.4% of patients in the CCP group and 32.7% in the control group (P = 0.32). The median time to clinical improvement was 26 days in the CCP group and 66 days in the control group (P = 0.27). The median time to discharge from the hospital was 31 days in the CCP group and 51 days in the control group (P = 0.24). In the subgroup that received a higher cumulative amount of neutralizing antibodies, the primary outcome occurred in 56.0% of the patients (vs. 32.1%), with significantly shorter intervals to clinical improvement (20 vs. 66 days, P < 0.05) and to hospital discharge (21 vs. 51 days, P = 0.03) and better survival (day-60 probability of survival 91.6% vs. 68.1%, P = 0.02) in comparison with the control group.ConclusionCCP added to standard treatment was not associated with a significant improvement in the primary and secondary outcomes. A predefined subgroup analysis showed a significant benefit of CCP among patients who received a larger amount of neutralizing antibodies.Trial registrationClinicalTrials.gov NCT04433910.FundingBundesministerium für Gesundheit (German Federal Ministry of Health): ZMVI1-2520COR802.
Collapse
Affiliation(s)
- Sixten Körper
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Manfred Weiss
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Wiesmann
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Kai Zacharowski
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Beate Grüner
- Division of Infectious Diseases, University Hospital and Medical Center Ulm, Ulm, Germany
| | - Lucas Ernst
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Peter Spieth
- Department of Anesthesiology and Critical Care Medicine, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Philipp M Lepper
- Department of Internal Medicine V - Pneumology, Allergology, Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Martin Bentz
- Department of Internal Medicine III, Hospital of Karlsruhe, Karlsruhe, Germany
| | - Sebastian Zinn
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Gregor Paul
- Department of Gastroenterology, Hepatology, Pneumology and Infectious Diseases, Klinikum Stuttgart, Stuttgart, Germany
| | - Johannes Kalbhenn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Kirschning
- Department of Anaesthesiology and Surgical Intensive Care Medicine, University of Heidelberg, University Medical Centre Mannheim, Mannheim, Germany
| | - Thomas Thiele
- Institute of Immunology and Transfusion Medicine, University Hospital Greifswald, Greifswald, Germany
| | - Thomas Appl
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Hinnerk Wulf
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Jan Matthias Kruse
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bettina Jungwirth
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | | |
Collapse
|
109
|
Arnold Egloff SA, Junglen A, Restivo JS, Wongskhaluang M, Martin C, Doshi P, Schlauch D, Fromell G, Sears LE, Correll M, Burris HA, LeMaistre CF. Convalescent plasma associates with reduced mortality and improved clinical trajectory in patients hospitalized with COVID-19. J Clin Invest 2021; 131:e151788. [PMID: 34464352 DOI: 10.1172/jci151788] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUNDEvidence supporting convalescent plasma (CP), one of the first investigational treatments for coronavirus disease 2019 (COVID-19), has been inconclusive, leading to conflicting recommendations. The primary objective was to perform a comparative effectiveness study of CP for all-cause, in-hospital mortality in patients with COVID-19.METHODSThe multicenter, electronic health records-based, retrospective study included 44,770 patients hospitalized with COVID-19 in one of 176 HCA Healthcare-affiliated community hospitals. Coarsened exact matching (1:k) was employed, resulting in a sample of 3774 CP and 10,687 comparison patients.RESULTSExamination of mortality using a shared frailty model, controlling for concomitant medications, date of admission, and days from admission to transfusion, demonstrated a significant association of CP with lower mortality risk relative to the comparison group (adjusted hazard ratio [aHR] = 0.71; 95% CI, 0.59-0.86; P < 0.001). Examination of patient risk trajectories, represented by 400 clinico-demographic features from our real-time risk model (RTRM), indicated that patients who received CP recovered more quickly. The stratification of days to transfusion revealed that CP within 3 days after admission, but not within 4 to 7 days, was associated with a significantly lower mortality risk (aHR = 0.53; 95% CI, 0.47-0.60; P < 0.001). CP serology level was inversely associated with mortality when controlling for its interaction with days to transfusion (HR = 0.998; 95% CI, 0.997-0.999; P = 0.013), yet it did not reach univariable significance.CONCLUSIONSThis large, diverse, multicenter cohort study demonstrated that CP, compared with matched controls, is significantly associated with reduced risk of in-hospital mortality. These observations highlight the utility of real-world evidence and suggest the need for further evaluation prior to abandoning CP as a viable therapy for COVID-19.FUNDINGThis research was supported in whole by HCA Healthcare and/or an HCA Healthcare-affiliated entity, including Sarah Cannon and Genospace.
Collapse
Affiliation(s)
- Shanna A Arnold Egloff
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Angela Junglen
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Joseph Sa Restivo
- HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | | | - Casey Martin
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Pratik Doshi
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Daniel Schlauch
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Gregg Fromell
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Lindsay E Sears
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Mick Correll
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Howard A Burris
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Charles F LeMaistre
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| |
Collapse
|
110
|
Körper S, Weiss M, Zickler D, Wiesmann T, Zacharowski K, Corman VM, Grüner B, Ernst L, Spieth P, Lepper PM, Bentz M, Zinn S, Paul G, Kalbhenn J, Dollinger MM, Rosenberger P, Kirschning T, Thiele T, Appl T, Mayer B, Schmidt M, Drosten C, Wulf H, Kruse JM, Jungwirth B, Seifried E, Schrezenmeier H. Results of the CAPSID randomized trial for high-dose convalescent plasma in patients with severe COVID-19. J Clin Invest 2021. [PMID: 34464358 DOI: 10.1101/2021.05.10.21256192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023] Open
Abstract
BACKGROUNDCOVID-19 convalescent plasma (CCP) has been considered a treatment option for COVID-19. This trial assessed the efficacy of a neutralizing antibody containing high-dose CCP in hospitalized adults with COVID-19 requiring respiratory support or intensive care treatment.METHODSPatients (n = 105) were randomized 1:1 to either receive standard treatment and 3 units of CCP or standard treatment alone. Control group patients with progress on day 14 could cross over to the CCP group. The primary outcome was a dichotomous composite outcome of survival and no longer fulfilling criteria for severe COVID-19 on day 21.ResultsThe primary outcome occurred in 43.4% of patients in the CCP group and 32.7% in the control group (P = 0.32). The median time to clinical improvement was 26 days in the CCP group and 66 days in the control group (P = 0.27). The median time to discharge from the hospital was 31 days in the CCP group and 51 days in the control group (P = 0.24). In the subgroup that received a higher cumulative amount of neutralizing antibodies, the primary outcome occurred in 56.0% of the patients (vs. 32.1%), with significantly shorter intervals to clinical improvement (20 vs. 66 days, P < 0.05) and to hospital discharge (21 vs. 51 days, P = 0.03) and better survival (day-60 probability of survival 91.6% vs. 68.1%, P = 0.02) in comparison with the control group.ConclusionCCP added to standard treatment was not associated with a significant improvement in the primary and secondary outcomes. A predefined subgroup analysis showed a significant benefit of CCP among patients who received a larger amount of neutralizing antibodies.Trial registrationClinicalTrials.gov NCT04433910.FundingBundesministerium für Gesundheit (German Federal Ministry of Health): ZMVI1-2520COR802.
Collapse
Affiliation(s)
- Sixten Körper
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Manfred Weiss
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Daniel Zickler
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Thomas Wiesmann
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Kai Zacharowski
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Beate Grüner
- Division of Infectious Diseases, University Hospital and Medical Center Ulm, Ulm, Germany
| | - Lucas Ernst
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Peter Spieth
- Department of Anesthesiology and Critical Care Medicine, Carl Gustav Carus University Hospital, Technische Universität Dresden, Dresden, Germany
| | - Philipp M Lepper
- Department of Internal Medicine V - Pneumology, Allergology, Intensive Care Medicine, Saarland University Hospital, Homburg, Germany
| | - Martin Bentz
- Department of Internal Medicine III, Hospital of Karlsruhe, Karlsruhe, Germany
| | - Sebastian Zinn
- Clinic of Anaesthesiology, Intensive Care Medicine and Pain Therapy, University Hospital Frankfurt, Frankfurt, Germany
| | - Gregor Paul
- Department of Gastroenterology, Hepatology, Pneumology and Infectious Diseases, Klinikum Stuttgart, Stuttgart, Germany
| | - Johannes Kalbhenn
- Department of Anesthesiology and Critical Care, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Peter Rosenberger
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Kirschning
- Department of Anaesthesiology and Surgical Intensive Care Medicine, University of Heidelberg, University Medical Centre Mannheim, Mannheim, Germany
| | - Thomas Thiele
- Institute of Immunology and Transfusion Medicine, University Hospital Greifswald, Greifswald, Germany
| | - Thomas Appl
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| | - Benjamin Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | - Michael Schmidt
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Christian Drosten
- Institute of Virology, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health and German Centre for Infection Research, Berlin, Germany
| | - Hinnerk Wulf
- Department of Anaesthesiology and Intensive Care Medicine, Philipps University Marburg, Marburg, Germany
| | - Jan Matthias Kruse
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, corporate member of Free University Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bettina Jungwirth
- Department of Anaesthesiology and Intensive Care Medicine, University Hospital Ulm, Ulm University, Ulm, Germany
| | - Erhard Seifried
- Institute of Transfusion Medicine and Immunohematology, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen, Frankfurt, Germany
| | - Hubert Schrezenmeier
- Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service Baden-Württemberg-Hessen and University Hospital Ulm, and Institute of Transfusion Medicine, and
| |
Collapse
|
111
|
Avendaño-Solá C, Ramos-Martínez A, Muñez-Rubio E, Ruiz-Antorán B, Malo de Molina R, Torres F, Fernández-Cruz A, Calderón-Parra J, Payares-Herrera C, Díaz de Santiago A, Romera-Martínez I, Pintos I, Lora-Tamayo J, Mancheño-Losa M, Paciello ML, Martínez-González AL, Vidán-Estévez J, Nuñez-Orantos MJ, Saez-Serrano MI, Porras-Leal ML, Jarilla-Fernández MC, Villares P, de Oteyza JP, Ramos-Garrido A, Blanco L, Madrigal-Sánchez ME, Rubio-Batllés M, Velasco-Iglesias A, Paño-Pardo JR, Moreno-Chulilla JA, Muñiz-Díaz E, Casas-Flecha I, Pérez-Olmeda M, García-Pérez J, Alcamí J, Bueno JL, Duarte RF. A multicenter randomized open-label clinical trial for convalescent plasma in patients hospitalized with COVID-19 pneumonia. J Clin Invest 2021; 131:e152740. [PMID: 34473652 DOI: 10.1172/jci152740] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/31/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUNDPassive immunotherapy with convalescent plasma (CP) is a potential treatment for COVID-19. Evidence from controlled clinical trials is inconclusive.METHODSWe conducted a randomized, open-label, controlled clinical trial at 27 hospitals in Spain. Patients had to be admitted for COVID-19 pneumonia within 7 days from symptom onset and not on mechanical ventilation or high-flow oxygen devices. Patients were randomized 1:1 to treatment with CP in addition to standard of care (SOC) or to the control arm receiving only SOC. The primary endpoint was the proportion of patients in categories 5 (noninvasive ventilation or high-flow oxygen), 6 (invasive mechanical ventilation or extracorporeal membrane oxygenation [ECMO]), or 7 (death) at 14 days. Primary analysis was performed in the intention-to-treat population.RESULTSBetween April 4, 2020, and February 5, 2021, 350 patients were randomly assigned to either CP (n = 179) or SOC (n = 171). At 14 days, proportion of patients in categories 5, 6, or 7 was 11.7% in the CP group versus 16.4% in the control group (P = 0.205). The difference was greater at 28 days, with 8.4% of patients in categories 5-7 in the CP group versus 17.0% in the control group (P = 0.021). The difference in overall survival did not reach statistical significance (HR 0.46, 95% CI 0.19-1.14, log-rank P = 0.087).CONCLUSIONCP showed a significant benefit in preventing progression to noninvasive ventilation or high-flow oxygen, invasive mechanical ventilation or ECMO, or death at 28 days. The effect on the predefined primary endpoint at 14 days and the effect on overall survival were not statistically significant.TRIAL REGISTRATIONClinicaltrials.gov, NCT04345523.FUNDINGGovernment of Spain, Instituto de Salud Carlos III.
Collapse
Affiliation(s)
- Cristina Avendaño-Solá
- Clinical Pharmacology Department, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Sanitaria Hospital Puerta de Hierro-Segovia de Arana, SCReN Clinical Trials Platform, Madrid, Spain
| | | | | | - Belen Ruiz-Antorán
- Clinical Pharmacology Department, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Sanitaria Hospital Puerta de Hierro-Segovia de Arana, SCReN Clinical Trials Platform, Madrid, Spain
| | - Rosa Malo de Molina
- Respiratory Medicine Service, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Ferran Torres
- Clinical Pharmacology Department, Hospital Clinic Barcelona, Barcelona, Spain.,Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | - Concepcion Payares-Herrera
- Clinical Pharmacology Department, Hospital Universitario Puerta de Hierro Majadahonda, Instituto de Investigación Sanitaria Hospital Puerta de Hierro-Segovia de Arana, SCReN Clinical Trials Platform, Madrid, Spain
| | | | - Irene Romera-Martínez
- Department of Hematology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Ilduara Pintos
- Infectious Diseases Unit, Department of Internal Medicine and
| | - Jaime Lora-Tamayo
- Department of Internal Medicine, Instituto de Investigación "i+12" and
| | | | - Maria L Paciello
- Department of Hematology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - Julia Vidán-Estévez
- Department of Hematology, Complejo Asistencial Universitario de León, León, Spain
| | | | | | | | - Maria C Jarilla-Fernández
- Hematology and Hemotherapy Service, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Paula Villares
- Department of Internal Medicine, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Jaime Pérez de Oteyza
- Department of Hematology, Hospital Universitario HM Sanchinarro, Universidad CEU San Pablo Madrid, Spain
| | | | - Lydia Blanco
- Centro de Hemoterapia y Hemodonación de Castilla y León, Valladolid, Spain
| | - Maria E Madrigal-Sánchez
- Centro de Transfusión de Ciudad Real, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | | | - Ana Velasco-Iglesias
- Spanish Clinical Research Network (ISCIII), IIS -Hospital Puerta de Hierro-Segovia de Arana, Madrid, Spain
| | | | - J A Moreno-Chulilla
- Division of Hematology, Hospital Clínico Universitario Zaragoza-IIS Aragón, Zaragoza, Spain
| | | | | | - Mayte Pérez-Olmeda
- Laboratorio de Serología, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, Spain
| | | | - Jose Alcamí
- AIDS Immunopathogenesis Unit, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose L Bueno
- Department of Hematology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Rafael F Duarte
- Department of Hematology, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | | |
Collapse
|
112
|
Glatt TN, Hilton C, Nyoni C, Swarts A, Swanevelder R, Cowley J, Mmenu C, Moyo-Gwete T, Moore PL, Kutama M, Jaza J, Phayane I, Brits T, Koekemoer J, Jentsch U, Nelson D, van den Berg K, Vermeulen M. Rapid and Successful Implementation of a COVID-19 Convalescent Plasma Programme-The South African Experience. Viruses 2021; 13:2050. [PMID: 34696480 PMCID: PMC8539971 DOI: 10.3390/v13102050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 10/04/2021] [Accepted: 10/04/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND COVID-19 convalescent plasma (CCP) has been considered internationally as a treatment option for COVID-19. CCP refers to plasma collected from donors who have recovered from and made antibodies to SARS-CoV-2. To date, convalescent plasma has not been collected in South Africa. As other investigational therapies and vaccination were not widely accessible, there was an urgent need to implement a CCP manufacture programme to service South Africans. METHODS The South African National Blood Service and the Western Cape Blood Service implemented a CCP programme that included CCP collection, processing, testing and storage. CCP units were tested for SARS-CoV-2 Spike ELISA and neutralising antibodies and routine blood transfusion parameters. CCP units from previously pregnant females were tested for anti-HLA and anti-HNA antibodies. RESULTS A total of 987 CCP units were collected from 243 donors, with a median of three donations per donor. Half of the CCP units had neutralising antibody titres of >1:160. One CCP unit was positive on the TPHA serology. All CCP units tested for anti-HLA antibodies were positive. CONCLUSION Within three months of the first COVID-19 diagnosis in South Africa, a fully operational CCP programme was set up across South Africa. The infrastructure and skills implemented will likely benefit South Africans in this and future pandemics.
Collapse
Affiliation(s)
- Tanya Nadia Glatt
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - Caroline Hilton
- Medical Division, Western Cape Blood Service, Cape Town 7405, South Africa;
| | - Cynthia Nyoni
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - Avril Swarts
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - Ronel Swanevelder
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - James Cowley
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
| | - Cordelia Mmenu
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
| | - Thandeka Moyo-Gwete
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2192, South Africa; (T.M.-G.); (P.L.M.)
- MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Penny L. Moore
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg 2192, South Africa; (T.M.-G.); (P.L.M.)
- MRC Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2000, South Africa
| | - Munzhedzi Kutama
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
| | - Jabulisile Jaza
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
| | - Itumeleng Phayane
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
| | - Tinus Brits
- Information Technology Division, South African National Blood Service, Roodepoort 1709, South Africa; (T.B.); (J.K.)
| | - Johan Koekemoer
- Information Technology Division, South African National Blood Service, Roodepoort 1709, South Africa; (T.B.); (J.K.)
| | - Ute Jentsch
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - Derrick Nelson
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
| | - Karin van den Berg
- Medical Division, South African National Blood Service, Roodepoort 1709, South Africa; (C.N.); (A.S.); (R.S.); (U.J.); (D.N.); (K.v.d.B.)
- Division of Clinical Haematology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town 7935, South Africa
- Division of Clinical Haematology, University of the Free State, Bloemfontein 9301, South Africa
| | - Marion Vermeulen
- Operations Division, South African National Blood Service, Roodepoort 1709, South Africa; (J.C.); (C.M.); (M.K.); (J.J.); (I.P.); (M.V.)
- Division of Clinical Haematology, University of the Free State, Bloemfontein 9301, South Africa
| |
Collapse
|
113
|
Abstract
PURPOSE OF REVIEW Over the course of the coronavirus disease 2019 (COVID-19) pandemic, it has become clear that the clinical features, epidemiology, and outcomes of COVID-19 are distinct in children relative to adults. In this review, we will present recent pediatric studies informing our current understanding of COVID-19 in children, and review pediatric considerations surrounding disease transmission, currently available therapies, and vaccination. RECENT FINDINGS Recent studies have shed light on the clinical epidemiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in children, identifying a high prevalence of asymptomatic and mild infections, with severe COVID-19 infrequently reported. Several adult clinical trials have informed the use of remdesivir, anti-SARS-CoV-2 monoclonal antibodies, dexamethasone, and tocilizumab in the management of COVID-19. Associations between underlying comorbid medical conditions and severe outcomes, as well as transmission dynamics of SARS-CoV-2 in children, are complex and warrant further study. Finally, highly efficacious vaccines are available for adults and adolescents, with pediatric trials ongoing. SUMMARY Children generally fare well with acute COVID-19 infection, though critical illness is possible. Future research should focus on clarifying the role of children in SARS-CoV-2 transmission and optimal prevention strategies, particularly in the school setting, as well as evaluating pediatric vaccine candidates.
Collapse
Affiliation(s)
- Emily R Levy
- Division of Pediatric Infectious Diseases
- Division of Pediatric Critical Care Medicine, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Jennifer Blumenthal
- Division of Infectious Diseases, Department of Medicine
- Division of Critical Care Medicine, Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Kathleen Chiotos
- Division of Infectious Diseases, Department of Pediatrics
- Division of Critical Care Medicine, Department of Anesthesia and Critical Care, Children's Hospital of Philadelphia, Pennsylvania, USA
| |
Collapse
|
114
|
Cao H, Ming L, Chen L, Zhu X, Shi Y. The Effectiveness of Convalescent Plasma for the Treatment of Novel Corona Virus Disease 2019: A Systematic Review and Meta-Analysis. Front Med (Lausanne) 2021; 8:641429. [PMID: 34646833 PMCID: PMC8502818 DOI: 10.3389/fmed.2021.641429] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/24/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Coronavirus disease 2019 (COVID-19), sweeping across the world, has created a worldwide pandemic. Effective treatments of COVID-19 are extremely urgent. Objective: To analyze the efficacy and safety of convalescent plasma (CCP) on patients with COVID-19. Methods: All the relevant studies were searched from PubMed, EMBASE,Cochrane library, Scopus, Web of Science, CBM, CNKI, Wan fang, VIP, Medrxiv, Biorxiv, and SSRN on July 19, 2021. PICOS criteria were as follows: (P) the study interests were human subjects with the infection of COVID-19; (I) the intervention of interest was CCP; (C) comparator treatments contained placebo, sham therapy, and standard treatment; (O) the primary outcome was mortality rates by the novel coronavirus. The secondary outcomes included the incidence of serious adverse events, the rate of ICU admission and mechanical ventilation (MV); the length of hospital stay; the duration of MV and ICU stay; the antibody levels, inflammatory factor levels, and viral loads. (S) Only randomized controlled trials (RCTs) of CCP were included. Subanalysis, quality assessment, sensitive analysis, and publication bias were conducted by two reviewers independently. Results: Sixteen RCTs were included and enrolled a total of 16,296 participants in this meta-analysis. The pooled data showed that no significant difference was observed in reducing the rate of overall mortality between CCP treatment group and placebo group (OR 0.96; 95% CI 0.90 to 1.03; p = 0.30; I 2 = 6%). According to the results of subgroup analysis, severe or critical patients with CCP showed significant difference in reducing the 28-day mortality of compared with placebo (OR 0.58, 95% CI 0.36 to 0.93, p = 0.02, I 2 = 0%). CCP groups have a significantly shorter duration of MV compared with the control group (weighted MD -1.00, 95% CI -1.86 to -0.14 d p = 0.02, I 2 = 0%). No significant difference was observed in the length of hospital stay, the duration of ICU, and the rate of ICU and MV. There is no conclusive evidence about the safety of CCP. Conclusion: Convalescent plasma can significantly reduce the 28-day mortality of severe or critical COVID-19 patients and the duration of MV. However, more evidence was needed to prove the safety of convalescent plasma.
Collapse
Affiliation(s)
- Huiling Cao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Ming
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Department of Cardiology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Long Chen
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xingwang Zhu
- Department of Pediatrics, Jiulongpo People's Hospital, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Chongqing, China
- National Clinical Research Center for Child Health and Disorders, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
115
|
Siemieniuk RA, Bartoszko JJ, Díaz Martinez JP, Kum E, Qasim A, Zeraatkar D, Izcovich A, Mangala S, Ge L, Han MA, Agoritsas T, Arnold D, Ávila C, Chu DK, Couban R, Cusano E, Darzi AJ, Devji T, Foroutan F, Ghadimi M, Khamis A, Lamontagne F, Loeb M, Miroshnychenko A, Motaghi S, Murthy S, Mustafa RA, Rada G, Rochwerg B, Switzer C, Vandvik PO, Vernooij RW, Wang Y, Yao L, Guyatt GH, Brignardello-Petersen R. Antibody and cellular therapies for treatment of covid-19: a living systematic review and network meta-analysis. BMJ 2021; 374:n2231. [PMID: 34556486 PMCID: PMC8459162 DOI: 10.1136/bmj.n2231] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To evaluate the efficacy and safety of antiviral antibody therapies and blood products for the treatment of novel coronavirus disease 2019 (covid-19). DESIGN Living systematic review and network meta-analysis, with pairwise meta-analysis for outcomes with insufficient data. DATA SOURCES WHO covid-19 database, a comprehensive multilingual source of global covid-19 literature, and six Chinese databases (up to 21 July 2021). STUDY SELECTION Trials randomising people with suspected, probable, or confirmed covid-19 to antiviral antibody therapies, blood products, or standard care or placebo. Paired reviewers determined eligibility of trials independently and in duplicate. METHODS After duplicate data abstraction, we performed random effects bayesian meta-analysis, including network meta-analysis for outcomes with sufficient data. We assessed risk of bias using a modification of the Cochrane risk of bias 2.0 tool. The certainty of the evidence was assessed using the grading of recommendations assessment, development, and evaluation (GRADE) approach. We meta-analysed interventions with ≥100 patients randomised or ≥20 events per treatment arm. RESULTS As of 21 July 2021, we identified 47 trials evaluating convalescent plasma (21 trials), intravenous immunoglobulin (IVIg) (5 trials), umbilical cord mesenchymal stem cells (5 trials), bamlanivimab (4 trials), casirivimab-imdevimab (4 trials), bamlanivimab-etesevimab (2 trials), control plasma (2 trials), peripheral blood non-haematopoietic enriched stem cells (2 trials), sotrovimab (1 trial), anti-SARS-CoV-2 IVIg (1 trial), therapeutic plasma exchange (1 trial), XAV-19 polyclonal antibody (1 trial), CT-P59 monoclonal antibody (1 trial) and INM005 polyclonal antibody (1 trial) for the treatment of covid-19. Patients with non-severe disease randomised to antiviral monoclonal antibodies had lower risk of hospitalisation than those who received placebo: casirivimab-imdevimab (odds ratio (OR) 0.29 (95% CI 0.17 to 0.47); risk difference (RD) -4.2%; moderate certainty), bamlanivimab (OR 0.24 (0.06 to 0.86); RD -4.1%; low certainty), bamlanivimab-etesevimab (OR 0.31 (0.11 to 0.81); RD -3.8%; low certainty), and sotrovimab (OR 0.17 (0.04 to 0.57); RD -4.8%; low certainty). They did not have an important impact on any other outcome. There was no notable difference between monoclonal antibodies. No other intervention had any meaningful effect on any outcome in patients with non-severe covid-19. No intervention, including antiviral antibodies, had an important impact on any outcome in patients with severe or critical covid-19, except casirivimab-imdevimab, which may reduce mortality in patients who are seronegative. CONCLUSION In patients with non-severe covid-19, casirivimab-imdevimab probably reduces hospitalisation; bamlanivimab-etesevimab, bamlanivimab, and sotrovimab may reduce hospitalisation. Convalescent plasma, IVIg, and other antibody and cellular interventions may not confer any meaningful benefit. SYSTEMATIC REVIEW REGISTRATION This review was not registered. The protocol established a priori is included as a data supplement. FUNDING This study was supported by the Canadian Institutes of Health Research (grant CIHR- IRSC:0579001321). READERS' NOTE This article is a living systematic review that will be updated to reflect emerging evidence. Interim updates and additional study data will be posted on our website (www.covid19lnma.com).
Collapse
Affiliation(s)
- Reed Ac Siemieniuk
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Joint first authors
| | - Jessica J Bartoszko
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Joint first authors
| | - Juan Pablo Díaz Martinez
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Joint first authors
| | - Elena Kum
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Joint first authors
| | - Anila Qasim
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Joint first authors
| | - Dena Zeraatkar
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Joint first authors
| | - Ariel Izcovich
- Servicio de Clinica Médica del Hospital Alemán, Buenos Aires, Argentina
| | - Sophia Mangala
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Long Ge
- Evidence Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, Gansu, China
| | - Mi Ah Han
- Department of Preventive Medicine, College of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Thomas Agoritsas
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Division of General Internal Medicine & Division of Clinical Epidemiology, University Hospitals of Geneva, Geneva, Switzerland
| | - Donald Arnold
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | | - Derek K Chu
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Rachel Couban
- Department of Anesthesia, McMaster University, Hamilton, ON, Canada
| | - Ellen Cusano
- Department of Medicine, University of Calgary, Calgary, AB, Canada
| | - Andrea J Darzi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Tahira Devji
- Medical school, University of Toronto, Toronto, ON, Canada
| | - Farid Foroutan
- Ted Rogers Center for Heart Research, University Health Network, Toronto, ON, Canada
| | - Maryam Ghadimi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Assem Khamis
- Wolfson Palliative Care Research Centre, Hull York Medical School, Hull, UK
| | - Francois Lamontagne
- Department of Medicine and Centre de recherche du CHU de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Mark Loeb
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Anna Miroshnychenko
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Sharhzad Motaghi
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Srinivas Murthy
- Department of Pediatrics, Faculty of Medicine, University of British Columbia, Vancouver
| | - Reem A Mustafa
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, University of Kansas Medical Center, Kansas City, MO, USA
| | | | - Bram Rochwerg
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Charlotte Switzer
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Per O Vandvik
- Institute of Health and Society, University of Oslo, Oslo, Norway
| | - Robin Wm Vernooij
- Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Ying Wang
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Liang Yao
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gordon H Guyatt
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Medicine, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
116
|
Devos T, Van Thillo Q, Compernolle V, Najdovski T, Romano M, Dauby N, Jadot L, Leys M, Maillart E, Loof S, Seyler L, Moonen M, Moutschen M, Van Regenmortel N, Ariën KK, Barbezange C, Betrains A, Garigliany M, Engelen MM, Gyselinck I, Maes P, Schauwvlieghe A, Liesenborghs L, Belmans A, Verhamme P, Meyfroidt G. Early high antibody-titre convalescent plasma for hospitalised COVID-19 patients: DAWn-plasma. Eur Respir J 2021; 59:13993003.01724-2021. [PMID: 34446469 PMCID: PMC8576805 DOI: 10.1183/13993003.01724-2021] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/24/2021] [Indexed: 12/15/2022]
Abstract
Background Several randomised clinical trials have studied convalescent plasma for coronavirus disease 2019 (COVID-19) using different protocols, with different severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) neutralising antibody titres, at different time-points and severities of illness. Methods In the prospective multicentre DAWn-plasma trial, adult patients hospitalised with COVID-19 were randomised to 4 units of open-label convalescent plasma combined with standard of care (intervention group) or standard of care alone (control group). Plasma from donors with neutralising antibody titres (50% neutralisation titre (NT50)) ≥1/320 was the product of choice for the study. Results Between 2 May 2020 and 26 January 2021, 320 patients were randomised to convalescent plasma and 163 patients to the control group according to a 2:1 allocation scheme. A median (interquartile range) volume of 884 (806–906) mL) convalescent plasma was administered and 80.68% of the units came from donors with neutralising antibody titres (NT50) ≥1/320. Median time from onset of symptoms to randomisation was 7 days. The proportion of patients alive and free of mechanical ventilation on day 15 was not different between both groups (convalescent plasma 83.74% (n=267) versus control 84.05% (n=137)) (OR 0.99, 95% CI 0.59–1.66; p=0.9772). The intervention did not change the natural course of antibody titres. The number of serious or severe adverse events was similar in both study arms and transfusion-related side-effects were reported in 19 out of 320 patients in the intervention group (5.94%). Conclusions Transfusion of 4 units of convalescent plasma with high neutralising antibody titres early in hospitalised COVID-19 patients did not result in a significant improvement of clinical status or reduced mortality. Early transfusion of 4 units of high neutralising antibody titre convalescent plasma in hospitalised COVID-19 patients does not reduce mortality or the need for mechanical ventilationhttps://bit.ly/3fiRY2I
Collapse
Affiliation(s)
- Timothy Devos
- Department of Hematology, University Hospitals Leuven and Department of Microbiology and Immunology, Laboratory of Molecular Immunology (Rega Institute), KU Leuven, Leuven, Belgium
| | - Quentin Van Thillo
- Center for Cancer Biology, Vlaams Instituut voor Biotechnologie (VIB), Leuven and Center for Human Genetics, KU Leuven, Leuven, Belgium
| | - Veerle Compernolle
- Belgian Red Cross, Blood Services, Mechelen, Belgium. Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | | | - Marta Romano
- Immune Response Service; Infectious Diseases in Humans Scientific Directorate, Sciensano, Brussels, Belgium
| | - Nicolas Dauby
- Department of Infectious Diseases, CHU Saint-Pierre, Universite Libre de Bruxelles (ULB), School of Public Health, Universite Libre de Bruxelles (ULB), Institute for Medical Immunology, Universite Libre de Bruxelles (ULB), Belgium
| | - Laurent Jadot
- Department of Anesthesiology and Intensive Care Medicine, and Department of Infectious diseases, CHC Mont Legia, Liege, Belgium
| | - Mathias Leys
- Department of Pulmonary Medicine, AZ Groeninge, Kortrijk, Belgium
| | - Evelyne Maillart
- Department of Infectious Diseases, Brugmann University Hospital, Brussels, Belgium
| | - Sarah Loof
- Department of Respiratory Medicine, AZ Maria Middelares Gent, Ghent, Belgium. Department of Respiratory Medicine, AZ Sint-Vincentius Deinze, Deinze, Belgium
| | - Lucie Seyler
- Department of Infectious Diseases and Internal Medicine, UZ Brussel Hospital, Brussels, Belgium
| | - Martial Moonen
- Department of Internal Medicine and Infectious Diseases, Centre Hospitalier Regional (CHR), Liege, Belgium
| | - Michel Moutschen
- Infectious Diseases and General Internal Medicine, CHU de Liege, ULiege, Belgium
| | - Niels Van Regenmortel
- Department of Intensive Care Medicine, Ziekenhuis Netwerk Antwerpen Campus Stuivenberg, Antwerp, Belgium
| | - Kevin K Ariën
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp and Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Albrecht Betrains
- Department of General Internal Medicine, University Hospitals Leuven, Leuven and Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium
| | - Mutien Garigliany
- University of Liege, Faculty of Veterinary Medicine, Animal Pathology, Liege, Belgium
| | | | - Iwein Gyselinck
- Laboratory of Respiratory Diseases and Thoracic Surgery (BREATHE), Department CHROMETA, KU Leuven, Respiratory Diseases UZ Leuven, Leuven, Belgium
| | - Piet Maes
- KU Leuven, Rega Institute for Medical Research, Clinical and Epidemiological Virology, Leuven, Belgium
| | | | - Laurens Liesenborghs
- Laboratory of Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ann Belmans
- I-BioStat, KU Leuven, Leuven, Belgium and University Hasselt, Hasselt, Belgium
| | - Peter Verhamme
- Department of Cardiovascular Sciences, UZ and KU Leuven, Belgium
| | - Geert Meyfroidt
- Department of Intensive Care Medicine, University Hospitals Leuven, and Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, KU Leuven, Leuven, Belgium
| | | |
Collapse
|
117
|
Natarajan H, Xu S, Crowley AR, Butler SE, Weiner JA, Bloch EM, Littlefield K, Benner SE, Shrestha R, Ajayi O, Wieland-alter W, Sullivan D, Shoham S, Quinn TC, Casadevall A, Pekosz A, Redd AD, Tobian AA, Connor RI, Wright PF, Ackerman ME. Antibody Attributes that Predict the Neutralization and Effector Function of Polyclonal Responses to SARS-CoV-2.. [PMID: 34401890 PMCID: PMC8366811 DOI: 10.1101/2021.08.06.21261710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
While antibodies provide significant protection from SARS-CoV-2 infection and disease sequelae, the specific attributes of the humoral response that contribute to immunity are incompletely defined. In this study, we employ machine learning to relate characteristics of the polyclonal antibody response raised by natural infection to diverse antibody effector functions and neutralization potency with the goal of generating both accurate predictions of each activity based on antibody response profiles as well as insights into antibody mechanisms of action. To this end, antibody-mediated phagocytosis, cytotoxicity, complement deposition, and neutralization were accurately predicted from biophysical antibody profiles in both discovery and validation cohorts. These predictive models identified SARS-CoV-2-specific IgM as a key predictor of neutralization activity whose mechanistic relevance was supported experimentally by depletion. Validated models of how different aspects of the humoral response relate to antiviral antibody activities suggest desirable attributes to recapitulate by vaccination or other antibody-based interventions.
Collapse
|
118
|
Lyman GH, Desai A, Leyfman Y, Kuderer NM. Opportunities and Challenges of Observational Studies and Randomized Controlled Trials for Evaluating the Therapeutic Efficacy of COVID-19 Convalescent Plasma. Cancer Invest 2021; 39:449-456. [PMID: 34134587 DOI: 10.1080/07357907.2021.1942127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Large randomized controlled trials (RCTs) remain the gold standard for evaluating treatment efficacy. However, observational studies, including non-randomized cohort studies, as well as small RCTs have gained increasing attention especially during the SARS-CoV-2 pandemic where critical evaluation of limited therapeutic options are sought to improve patient care while awaiting results for subsequent RCTs. As the authors have previously discussed, RCTs and observational studies are complementary approaches which often appear synergistic with one another. While not all real-world studies are the same, the results of observational studies are notoriously subject to both known and unknown confounding factors. The utilization of COVID-19 Convalescent Plasma is a timely illustration of evaluating the efficacy and safety of a COVID-19 therapy given the dangerous and often lethal effects of the virus and the limited approved therapeutic options for the disease. While awaiting the results of large RCTS of convalescent plasma, serval observational cohorts and small RCTs have attempted to assess the efficacy and safety of this approach with very mixed results. Among the likely reasons for this failure to provide a definitive answer concerning the value of convalescent plasma are the many limitations inherent to addressing treatment efficacy in non-randomized studies. While such studies are often able to capture information on large numbers of individuals rapidly, it is important to understand that although larger numbers may enhance the precision of estimates provided, larger numbers, in and of themselves, do not increase the accuracy of estimates due to patient selection and other biases. At the same time, both observational studies and small RCTS are at risk for publication bias due to investigator, reviewer and editorial bias toward positive studies. In this commentary we discuss the advantages and limitations of these methodologic approaches when addressing urgently needed evidence on the effectiveness and safety of therapies in a crisis such as the COVID-19 pandemic.
Collapse
Affiliation(s)
- Gary H Lyman
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Aakash Desai
- Mayo Clinic School of Medicine, Rochester, MN, USA
| | - Yan Leyfman
- Penn State College of Medicine, Hershey, PA, USA
| | | |
Collapse
|
119
|
Klassen SA, Senefeld JW, Senese KA, Johnson PW, Wiggins CC, Baker SE, van Helmond N, Bruno KA, Pirofski LA, Shoham S, Grossman BJ, Henderson JP, Wright RS, Fairweather D, Paneth NS, Carter RE, Casadevall A, Joyner MJ. Convalescent Plasma Therapy for COVID-19: A Graphical Mosaic of the Worldwide Evidence. Front Med (Lausanne) 2021; 8:684151. [PMID: 34164419 PMCID: PMC8215127 DOI: 10.3389/fmed.2021.684151] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/04/2021] [Indexed: 12/15/2022] Open
Abstract
Convalescent plasma has been used worldwide to treat patients hospitalized with coronavirus disease 2019 (COVID-19) and prevent disease progression. Despite global usage, uncertainty remains regarding plasma efficacy, as randomized controlled trials (RCTs) have provided divergent evidence regarding the survival benefit of convalescent plasma. Here, we argue that during a global health emergency, the mosaic of evidence originating from multiple levels of the epistemic hierarchy should inform contemporary policy and healthcare decisions. Indeed, worldwide matched-control studies have generally found convalescent plasma to improve COVID-19 patient survival, and RCTs have demonstrated a survival benefit when transfused early in the disease course but limited or no benefit later in the disease course when patients required greater supportive therapies. RCTs have also revealed that convalescent plasma transfusion contributes to improved symptomatology and viral clearance. To further investigate the effect of convalescent plasma on patient mortality, we performed a meta-analytical approach to pool daily survival data from all controlled studies that reported Kaplan-Meier survival plots. Qualitative inspection of all available Kaplan-Meier survival data and an aggregate Kaplan-Meier survival plot revealed a directionally consistent pattern among studies arising from multiple levels of the epistemic hierarchy, whereby convalescent plasma transfusion was generally associated with greater patient survival. Given that convalescent plasma has a similar safety profile as standard plasma, convalescent plasma should be implemented within weeks of the onset of future infectious disease outbreaks.
Collapse
Affiliation(s)
- Stephen A. Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Katherine A. Senese
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Patrick W. Johnson
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | - Chad C. Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Sarah E. Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| | - Noud van Helmond
- Department of Anesthesiology, Cooper Medical School of Rowan University, Cooper University Health Care, Camden, NJ, United States
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, New York, NY, United States
| | - Shmuel Shoham
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - Jeffrey P. Henderson
- Division of Infectious Diseases, Department of Medicine, Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, St. Louis, MO, United States
| | - R. Scott Wright
- Department of Cardiovascular Medicine, Human Research Protection Program, Mayo Clinic, Rochester, MN, United States
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, United States
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics, Department of Pediatrics and Human Development, College of Human Medicine, Michigan State University, East Lansing, MI, United States
| | - Rickey E. Carter
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, United States
| | - Arturo Casadevall
- Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
120
|
Casadevall A, Dragotakes Q, Johnson PW, Senefeld JW, Klassen SA, Wright RS, Joyner MJ, Paneth N, Carter RE. Convalescent plasma use in the USA was inversely correlated with COVID-19 mortality. eLife 2021; 10:e69866. [PMID: 34085928 PMCID: PMC8205484 DOI: 10.7554/elife.69866] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Background The US Food and Drug Administration authorized COVID-19 convalescent plasma (CCP) therapy for hospitalized COVID-19 patients via the Expanded Access Program (EAP) and the Emergency Use Authorization (EUA), leading to use in about 500,000 patients during the first year of the pandemic for the USA. Methods We tracked the number of CCP units dispensed to hospitals by blood banking organizations and correlated that usage with hospital admission and mortality data. Results CCP usage per admission peaked in Fall 2020, with more than 40% of inpatients estimated to have received CCP between late September and early November 2020. However, after randomized controlled trials failed to show a reduction in mortality, CCP usage per admission declined steadily to a nadir of less than 10% in March 2021. We found a strong inverse correlation (r = -0.52, p=0.002) between CCP usage per hospital admission and deaths occurring 2 weeks after admission, and this finding was robust to examination of deaths taking place 1, 2, or 3 weeks after admission. Changes in the number of hospital admissions, SARS-CoV-2 variants, and age of patients could not explain these findings. The retreat from CCP usage might have resulted in as many as 29,000 excess deaths from mid-November 2020 to February 2021. Conclusions A strong inverse correlation between CCP use and mortality per admission in the USA provides population-level evidence consistent with the notion that CCP reduces mortality in COVID-19 and suggests that the recent decline in usage could have resulted in excess deaths. Funding There was no specific funding for this study. AC was supported in part by RO1 HL059842 and R01 AI1520789; MJJ was supported in part by 5R35HL139854. This project has been funded in whole or in part with Federal funds from the Department of Health and Human Services; Office of the Assistant Secretary for Preparedness and Response; Biomedical Advanced Research and Development Authority under Contract No. 75A50120C00096.
Collapse
Affiliation(s)
- Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public HealthBaltimoreUnited States
| | - Quigly Dragotakes
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public HealthBaltimoreUnited States
| | - Patrick W Johnson
- Department of Quantitative Health Sciences, Mayo ClinicJacksonvilleUnited States
| | - Jonathon W Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo ClinicRochesterUnited States
| | - Stephen A Klassen
- Department of Anesthesiology and Perioperative Medicine, Mayo ClinicRochesterUnited States
| | - R Scott Wright
- Department of Cardiology, Mayo ClinicRochesterUnited States
| | - Michael J Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo ClinicRochesterUnited States
| | - Nigel Paneth
- Department of Epidemiology and Biostatistics and Department of Pediatrics and Human Development, College of Human Medicine, Michigan State UniversityEast LansingUnited States
| | - Rickey E Carter
- Department of Quantitative Health Sciences, Mayo ClinicJacksonvilleUnited States
| |
Collapse
|
121
|
Piechotta V, Iannizzi C, Chai KL, Valk SJ, Kimber C, Dorando E, Monsef I, Wood EM, Lamikanra AA, Roberts DJ, McQuilten Z, So-Osman C, Estcourt LJ, Skoetz N. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 5:CD013600. [PMID: 34013969 PMCID: PMC8135693 DOI: 10.1002/14651858.cd013600.pub4] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Convalescent plasma and hyperimmune immunoglobulin may reduce mortality in patients with viral respiratory diseases, and are being investigated as potential therapies for coronavirus disease 2019 (COVID-19). A thorough understanding of the current body of evidence regarding benefits and risks of these interventions is required. OBJECTIVES: Using a living systematic review approach, to assess whether convalescent plasma or hyperimmune immunoglobulin transfusion is effective and safe in the treatment of people with COVID-19; and to maintain the currency of the evidence. SEARCH METHODS To identify completed and ongoing studies, we searched the World Health Organization (WHO) COVID-19 Global literature on coronavirus disease Research Database, MEDLINE, Embase, the Cochrane COVID-19 Study Register, the Epistemonikos COVID-19 L*OVE Platform, and trial registries. Searches were done on 17 March 2021. SELECTION CRITERIA We included randomised controlled trials (RCTs) evaluating convalescent plasma or hyperimmune immunoglobulin for COVID-19, irrespective of disease severity, age, gender or ethnicity. For safety assessments, we also included non-controlled non-randomised studies of interventions (NRSIs) if 500 or more participants were included. We excluded studies that included populations with other coronavirus diseases (severe acute respiratory syndrome (SARS) or Middle East respiratory syndrome (MERS)), as well as studies evaluating standard immunoglobulin. DATA COLLECTION AND ANALYSIS We followed standard Cochrane methodology. To assess bias in included studies, we used the Cochrane 'Risk of Bias 2' tool for RCTs, and for NRSIs, the assessment criteria for observational studies, provided by Cochrane Childhood Cancer. We rated the certainty of evidence, using the GRADE approach, for the following outcomes: all-cause mortality, improvement and worsening of clinical status (for individuals with moderate to severe disease), development of severe clinical COVID-19 symptoms (for individuals with asymptomatic or mild disease), quality of life (including fatigue and functional independence), grade 3 or 4 adverse events, and serious adverse events. MAIN RESULTS We included 13 studies (12 RCTs, 1 NRSI) with 48,509 participants, of whom 41,880 received convalescent plasma. We did not identify any completed studies evaluating hyperimmune immunoglobulin. We identified a further 100 ongoing studies evaluating convalescent plasma or hyperimmune immunoglobulin, and 33 studies reporting as being completed or terminated. Individuals with a confirmed diagnosis of COVID-19 and moderate to severe disease Eleven RCTs and one NRSI investigated the use of convalescent plasma for 48,349 participants with moderate to severe disease. Nine RCTs compared convalescent plasma to placebo treatment or standard care alone, and two compared convalescent plasma to standard plasma (results not included in abstract). Effectiveness of convalescent plasma We included data on nine RCTs (12,875 participants) to assess the effectiveness of convalescent plasma compared to placebo or standard care alone. Convalescent plasma does not reduce all-cause mortality at up to day 28 (risk ratio (RR) 0.98, 95% confidence interval (CI) 0.92 to 1.05; 7 RCTs, 12,646 participants; high-certainty evidence). It has little to no impact on clinical improvement for all participants when assessed by liberation from respiratory support (RR not estimable; 8 RCTs, 12,682 participants; high-certainty evidence). It has little to no impact on the chance of being weaned or liberated from invasive mechanical ventilation for the subgroup of participants requiring invasive mechanical ventilation at baseline (RR 1.04, 95% CI 0.57 to 1.93; 2 RCTs, 630 participants; low-certainty evidence). It does not reduce the need for invasive mechanical ventilation (RR 0.98, 95% CI 0.89 to 1.08; 4 RCTs, 11,765 participants; high-certainty evidence). We did not identify any subgroup differences. We did not identify any studies reporting quality of life, and therefore, do not know whether convalescent plasma has any impact on quality of life. One RCT assessed resolution of fatigue on day 7, but we are very uncertain about the effect (RR 1.21, 95% CI 1.02 to 1.42; 309 participants; very low-certainty evidence). Safety of convalescent plasma We included results from eight RCTs, and one NRSI, to assess the safety of convalescent plasma. Some of the RCTs reported on safety data only for the convalescent plasma group. We are uncertain whether convalescent plasma increases or reduces the risk of grade 3 and 4 adverse events (RR 0.90, 95% CI 0.58 to 1.41; 4 RCTs, 905 participants; low-certainty evidence), and serious adverse events (RR 1.24, 95% CI 0.81 to 1.90; 2 RCTs, 414 participants; low-certainty evidence). A summary of reported events of the NRSI (reporting safety data for 20,000 of 35,322 transfused participants), and four RCTs reporting safety data only for transfused participants (6125 participants) are included in the full text. Individuals with a confirmed diagnosis of SARS-CoV-2 infection and asymptomatic or mild disease We identified one RCT reporting on 160 participants, comparing convalescent plasma to placebo treatment (saline). Effectiveness of convalescent plasma We are very uncertain about the effect of convalescent plasma on all-cause mortality (RR 0.50, 95% CI 0.09 to 2.65; very low-certainty evidence). We are uncertain about the effect of convalescent plasma on developing severe clinical COVID-19 symptoms (RR not estimable; low-certainty evidence). We identified no study reporting quality of life. Safety of convalescent plasma We do not know whether convalescent plasma is associated with a higher risk of grade 3 or 4 adverse events (very low-certainty evidence), or serious adverse events (very low-certainty evidence). This is a living systematic review. We search weekly for new evidence and update the review when we identify relevant new evidence. Please refer to the Cochrane Database of Systematic Reviews for the current status of this review. AUTHORS' CONCLUSIONS We have high certainty in the evidence that convalescent plasma for the treatment of individuals with moderate to severe disease does not reduce mortality and has little to no impact on measures of clinical improvement. We are uncertain about the adverse effects of convalescent plasma. While major efforts to conduct research on COVID-19 are being made, heterogeneous reporting of outcomes is still problematic. There are 100 ongoing studies and 33 studies reporting in a study registry as being completed or terminated. Publication of ongoing studies might resolve some of the uncertainties around hyperimmune immunoglobulin therapy for people with any disease severity, and convalescent plasma therapy for people with asymptomatic or mild disease.
Collapse
Affiliation(s)
- Vanessa Piechotta
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Claire Iannizzi
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Khai Li Chai
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Sarah J Valk
- Jon J van Rood Center for Clinical Transfusion Research, Sanquin/Leiden University Medical Center, Leiden, Netherlands
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Catherine Kimber
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Elena Dorando
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ina Monsef
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Erica M Wood
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | | | - David J Roberts
- Systematic Review Initiative, NHS Blood and Transplant, Oxford, UK
| | - Zoe McQuilten
- Transfusion Research Unit, School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Cynthia So-Osman
- Sanquin Blood Bank, Amsterdam, Netherlands
- Erasmus Medical Centre, Rotterdam, Netherlands
| | - Lise J Estcourt
- Haematology/Transfusion Medicine, NHS Blood and Transplant, Oxford, UK
| | - Nicole Skoetz
- Cochrane Cancer, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
122
|
Chalmers JD, Crichton ML, Goeminne PC, Cao B, Humbert M, Shteinberg M, Antoniou KM, Ulrik CS, Parks H, Wang C, Vandendriessche T, Qu J, Stolz D, Brightling C, Welte T, Aliberti S, Simonds AK, Tonia T, Roche N. Management of hospitalised adults with coronavirus disease 2019 (COVID-19): a European Respiratory Society living guideline. Eur Respir J 2021; 57:2100048. [PMID: 33692120 PMCID: PMC7947358 DOI: 10.1183/13993003.00048-2021] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Hospitalised patients with coronavirus disease 2019 (COVID-19) as a result of SARS-CoV-2 infection have a high mortality rate and frequently require noninvasive respiratory support or invasive ventilation. Optimising and standardising management through evidence-based guidelines may improve quality of care and therefore patient outcomes. METHODS A task force from the European Respiratory Society and endorsed by the Chinese Thoracic Society identified priority interventions (pharmacological and non-pharmacological) for the initial version of this "living guideline" using the PICO (population, intervention, comparator, outcome) format. The GRADE approach was used for assessing the quality of evidence and strength of recommendations. Systematic literature reviews were performed, and data pooled by meta-analysis where possible. Evidence tables were presented and evidence to decision frameworks were used to formulate recommendations. RESULTS Based on the available evidence at the time of guideline development (20 February, 2021), the panel makes a strong recommendation in favour of the use of systemic corticosteroids in patients requiring supplementary oxygen or ventilatory support, and for the use of anticoagulation in hospitalised patients. The panel makes a conditional recommendation for interleukin (IL)-6 receptor antagonist monoclonal antibody treatment and high-flow nasal oxygen or continuous positive airway pressure in patients with hypoxaemic respiratory failure. The panel make strong recommendations against the use of hydroxychloroquine and lopinavir-ritonavir. Conditional recommendations are made against the use of azithromycin, hydroxychloroquine combined with azithromycin, colchicine, and remdesivir, in the latter case specifically in patients requiring invasive mechanical ventilation. No recommendation was made for remdesivir in patients requiring supplemental oxygen. Further recommendations for research are made. CONCLUSION The evidence base for management of COVID-19 now supports strong recommendations in favour and against specific interventions. These guidelines will be regularly updated as further evidence becomes available.
Collapse
Affiliation(s)
- James D Chalmers
- School of Medicine, University of Dundee, Dundee, UK
- J.D. Chalmers and N. Roche are task force co-chairs
| | | | - Pieter C Goeminne
- Department of Respiratory Medicine, AZ Nikolaas, Sint-Niklaas, Belgium
| | - Bin Cao
- Department of Respiratory and Critical Care Medicine, Clinical Microbiology and Infectious Disease Lab, China-Japan Friendship Hospital, National Center for Respiratory Medicine, Institute of Respiratory Medicine, Chinese Academy of Medical Science, National Clinical Research Center of Respiratory Diseases, Beijing, China
| | - Marc Humbert
- Service de Pneumologie et Soins Intensifs, Hôpital Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP); Université Paris-Saclay; Inserm UMR_S 999, Le Kremlin Bicêtre, France
| | - Michal Shteinberg
- Pulmonology institute and CF Center, Carmel Medical Center and the Technion-Israel Institute of Technology, Haifa, Israel
| | - Katerina M Antoniou
- Laboratory of Molecular and Cellular Pneumonology, Department of Respiratory Medicine, School of Medicine, University of Crete, Heraklion, Greece
| | - Charlotte Suppli Ulrik
- Department of Respiratory Medicine, Copenhagen University Hospital-Hvidovre Hospital, Hvidovre, Denmark
| | | | - Chen Wang
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center of Respiratory Diseases, Beijing, China
| | | | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Ruijin Hospital, Shanghai, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Daiana Stolz
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, Basel, Switzerland
- Clinic of Respiratory Medicine, Medical Center - University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Tobias Welte
- Medizinische Hochschule Hannover, Direktor der Abteilung Pneumologie, Hannover, Germany
| | - Stefano Aliberti
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- IRCCS Humanitas Research Hospital, Respiratory Unit, Rozzano, Italy
| | - Anita K Simonds
- Sleep and Ventilation Unit, Royal Brompton and Harefield Hospital, Guys and St Thomas NHS Foundation Trust, London, UK
| | - Thomy Tonia
- Institute of Social and Preventive Medicine, University Bern, Bern, Switzerland
| | - Nicolas Roche
- Respiratory Medicine, Cochin Hospital, APHP Centre-University of Paris, Cochin Institute (INSERM UMR1016), Paris, France
- J.D. Chalmers and N. Roche are task force co-chairs
| |
Collapse
|