1
|
Nierich A, Bihariesingh R, Bansie R. HemoClear: A Practical and Cost-Effective Alternative to Conventional Convalescent Plasma Retrieval Methods. Curr Top Microbiol Immunol 2024. [PMID: 39126485 DOI: 10.1007/82_2024_276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
Convalescent plasma has increasingly been used to treat various viral infections and confer post-exposure prophylactic protection during the last decade and has demonstrated favorable clinical outcomes in patients infected with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) during the recent COVID-19 pandemic. The pandemic has highlighted the need for cost-effective, accessible, and easy-to-use alternatives to conventional blood plasmapheresis techniques, allowing hospitals to become more self-sufficient in harvesting and transfusing donor plasma into recipients in a single setting. To this end, the use of a membrane-based bedside plasmapheresis device (HemoClear) was evaluated in an open-label, non-randomized prospective trial in Suriname in 2021, demonstrating its practicality and efficacy in a low-to middle-income country. This paper will review the use of this method and its potential to expedite the process of obtaining convalescent plasma, especially during pandemics and in resource-constrained settings.
Collapse
Affiliation(s)
- Arno Nierich
- Department of Anesthesiology, Academic Hospital Paramaribo, Paramaribo, Suriname.
- Chief Medical Officer Hemoclear, Zwolle, The Netherlands.
| | - Rosita Bihariesingh
- Department of Anesthesiology & Intensive Care, Academic Hospital Paramaribo, Paramaribo, Suriname
| | - Rakesh Bansie
- Department of Anesthesiology & Internal Medicine, Academic Hospital Paramaribo, Paramaribo, Suriname
| |
Collapse
|
2
|
Senefeld JW, Gorman EK, Johnson PW, Moir ME, Klassen SA, Carter RE, Paneth NS, Sullivan DJ, Morkeberg OH, Wright RS, Fairweather D, Bruno KA, Shoham S, Bloch EM, Focosi D, Henderson JP, Juskewitch JE, Pirofski LA, Grossman BJ, Tobian AA, Franchini M, Ganesh R, Hurt RT, Kay NE, Parikh SA, Baker SE, Buchholtz ZA, Buras MR, Clayburn AJ, Dennis JJ, Diaz Soto JC, Herasevich V, Klompas AM, Kunze KL, Larson KF, Mills JR, Regimbal RJ, Ripoll JG, Sexton MA, Shepherd JR, Stubbs JR, Theel ES, van Buskirk CM, van Helmond N, Vogt MN, Whelan ER, Wiggins CC, Winters JL, Casadevall A, Joyner MJ. Rates Among Hospitalized Patients With COVID-19 Treated With Convalescent Plasma: A Systematic Review and Meta-Analysis. Mayo Clin Proc Innov Qual Outcomes 2023; 7:499-513. [PMID: 37859995 PMCID: PMC10582279 DOI: 10.1016/j.mayocpiqo.2023.09.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023] Open
Abstract
Objective To examine the association of COVID-19 convalescent plasma transfusion with mortality and the differences between subgroups in hospitalized patients with COVID-19. Patients and Methods On October 26, 2022, a systematic search was performed for clinical studies of COVID-19 convalescent plasma in the literature from January 1, 2020, to October 26, 2022. Randomized clinical trials and matched cohort studies investigating COVID-19 convalescent plasma transfusion compared with standard of care treatment or placebo among hospitalized patients with confirmed COVID-19 were included. The electronic search yielded 3841 unique records, of which 744 were considered for full-text screening. The selection process was performed independently by a panel of 5 reviewers. The study followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Data were extracted by 5 independent reviewers in duplicate and pooled using an inverse-variance random effects model. The prespecified end point was all-cause mortality during hospitalization. Results Thirty-nine randomized clinical trials enrolling 21,529 participants and 70 matched cohort studies enrolling 50,160 participants were included in the systematic review. Separate meta-analyses reported that transfusion of COVID-19 convalescent plasma was associated with a decrease in mortality compared with the control cohort for both randomized clinical trials (odds ratio [OR], 0.87; 95% CI, 0.76-1.00) and matched cohort studies (OR, 0.76; 95% CI, 0.66-0.88). The meta-analysis of subgroups revealed 2 important findings. First, treatment with convalescent plasma containing high antibody levels was associated with a decrease in mortality compared with convalescent plasma containing low antibody levels (OR, 0.85; 95% CI, 0.73 to 0.99). Second, earlier treatment with COVID-19 convalescent plasma was associated with a decrease in mortality compared with the later treatment cohort (OR, 0.63; 95% CI, 0.48 to 0.82). Conclusion During COVID-19 convalescent plasma use was associated with a 13% reduced risk of mortality, implying a mortality benefit for hospitalized patients with COVID-19, particularly those treated with convalescent plasma containing high antibody levels treated earlier in the disease course.
Collapse
Affiliation(s)
- Jonathon W. Senefeld
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL
| | - Ellen K. Gorman
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Patrick W. Johnson
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL
| | - M. Erin Moir
- Department of Kinesiology, University of Wisconsin-Madison, Madison
| | - Stephen A. Klassen
- Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada
| | - Rickey E. Carter
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL
| | - Nigel S. Paneth
- Department of Epidemiology and Biostatistics and Department of Pediatrics and Human Development, Michigan State University, East Lansing
| | - David J. Sullivan
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, ML
| | - Olaf H. Morkeberg
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - R. Scott Wright
- Human Research Protection Program, Mayo Clinic, Rochester, MN
| | | | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL
- Division of Cardiovascular Medicine, University of Florida, Gainesville
| | - Shmuel Shoham
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Evan M. Bloch
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, ML
| | - Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Italy
| | - Jeffrey P. Henderson
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in St. Louis, MO
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, MO
| | | | - Liise-Anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, NY
| | - Brenda J. Grossman
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, MO
| | - Aaron A.R. Tobian
- Department of Pathology Johns Hopkins University School of Medicine, Baltimore, ML
| | - Massimo Franchini
- Division of Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Ravindra Ganesh
- Department of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Ryan T. Hurt
- Department of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Neil E. Kay
- Division of Hematology, Mayo Clinic, Rochester, MN
- Department of Immunology, Mayo Clinic, Rochester, MN
| | | | - Sarah E. Baker
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Zachary A. Buchholtz
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Matthew R. Buras
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | - Andrew J. Clayburn
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Joshua J. Dennis
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Juan C. Diaz Soto
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Vitaly Herasevich
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Allan M. Klompas
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Katie L. Kunze
- Department of Quantitative Health Sciences, Mayo Clinic, Scottsdale, AZ
| | | | - John R. Mills
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Riley J. Regimbal
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Juan G. Ripoll
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Matthew A. Sexton
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - John R.A. Shepherd
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - James R. Stubbs
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Elitza S. Theel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | | | - Noud van Helmond
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Matthew N.P. Vogt
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL
| | - Chad C. Wiggins
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| | - Jeffrey L. Winters
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Arturo Casadevall
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, ML
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
3
|
Zeng Y, Guan W, Wang K, Jie Z, Zou X, Tan X, Li X, Chen X, Ren X, Jiang J, Zheng Z, Shi J, Zhong N. Effect of hydrogen/oxygen therapy for ordinary COVID-19 patients: a propensity-score matched case-control study. BMC Infect Dis 2023; 23:440. [PMID: 37386364 DOI: 10.1186/s12879-023-08424-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/24/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Hydrogen/oxygen therapy contribute to ameliorate dyspnea and disease progression in patients with respiratory diseases. Therefore, we hypothesized that hydrogen/oxygen therapy for ordinary coronavirus disease 2019 (COVID-19) patients might reduce the length of hospitalization and increase hospital discharge rates. METHODS This retrospective, propensity-score matched (PSM) case-control study included 180 patients hospitalized with COVID-19 from 3 centers. After assigned in 1:2 ratios by PSM, 33 patients received hydrogen/oxygen therapy and 55 patients received oxygen therapy included in this study. Primary endpoint was the length of hospitalization. Secondary endpoints were hospital discharge rates and oxygen saturation (SpO2). Vital signs and respiratory symptoms were also observed. RESULTS Findings confirmed a significantly lower median length of hospitalization (HR = 1.91; 95% CIs, 1.25-2.92; p < 0.05) in the hydrogen/oxygen group (12 days; 95% CI, 9-15) versus the oxygen group (13 days; 95% CI, 11-20). The higher hospital discharge rates were observed in the hydrogen/oxygen group at 21 days (93.9% vs. 74.5%; p < 0.05) and 28 days (97.0% vs. 85.5%; p < 0.05) compared with the oxygen group, except for 14 days (69.7% vs. 56.4%). After 5-day therapy, patients in hydrogen/oxygen group exhibited a higher level of SpO2 compared with that in the oxygen group (98.5%±0.56% vs. 97.8%±1.0%; p < 0.001). In subgroup analysis of patients received hydrogen/oxygen, patients aged < 55 years (p = 0.028) and without comorbidities (p = 0.002) exhibited a shorter hospitalization (median 10 days). CONCLUSION This study indicated that hydrogen/oxygen might be a useful therapeutic medical gas to enhance SpO2 and shorten length of hospitalization in patients with ordinary COVID-19. Younger patients or those without comorbidities are likely to benefit more from hydrogen/oxygen therapy.
Collapse
Affiliation(s)
- Yingying Zeng
- Department of Respiratory and Critical Care Medicine, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Weijie Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510000, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Zhijun Jie
- Department of Respiratory and Critical Care Medicine, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Xu Zou
- Department of Critical Care Medicine, Guangdong Province Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xiaoping Tan
- Department of Respiratory and Critical Care Medicine, Jiangling County People's Hospital, Jingzhou, Hubei, China
| | - Xinyu Li
- Department of Infectious Diseases, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Jiaotong University Affiliated Sixth People Hospital, Shanghai, China
| | - Xiaoting Ren
- Department of Respiratory and Critical Care Medicine, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai, 200240, China
- Center of Community-Based Health Research, Fudan University, Shanghai, China
| | - Junhong Jiang
- Department of Respiratory and Critical Care Medicine, Dushu Lake Hospital Affiliated to Soochow University, Jiangsu, China
| | - Zeguang Zheng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510000, China
| | - Jindong Shi
- Department of Respiratory and Critical Care Medicine, Fifth People's Hospital of Shanghai, Fudan University, 801 Heqing Road, Shanghai, 200240, China.
- Center of Community-Based Health Research, Fudan University, Shanghai, China.
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Road, Guangzhou, 510000, China.
| |
Collapse
|
4
|
Wouters E, Verbrugghe C, Abdelnabi R, Devloo R, De Clippel D, Jochmans D, De Bleser D, Weynand B, Compernolle V, Neyts J, Feys HB. Intranasal administration of convalescent plasma protects against SARS-CoV-2 infection in hamsters. EBioMedicine 2023; 92:104597. [PMID: 37148586 PMCID: PMC10171892 DOI: 10.1016/j.ebiom.2023.104597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 05/08/2023] Open
Abstract
BACKGROUND Convalescent plasma (CP) transfusion is an early option for treating infections with pandemic potential, often preceding vaccine or antiviral drug rollout. Heterogenous findings from randomized clinical trials on transfusion of COVID-19 CP (CCP) have been reported. However, meta-analysis suggests that transfusion of high titer CCP is associated with a mortality benefit for COVID-19 outpatients or inpatients treated within 5 days after symptom onset, indicating the importance of early administration. METHODS We tested if CCP is an effective prophylactic against SARS-CoV-2 infection by the intranasal administration of 25 μL CCP/nostril (i.e. 0.01-0.06 mg anti-RBD antibodies/kg) in hamsters exposed to infected littermates. FINDINGS In this model, 40% of CCP treated hamsters were fully protected and 40% had significantly reduced viral loads, the remaining 20% was not protected. The effect seems dose-dependent because high-titer CCP from a vaccinated donor was more effective than low-titer CCP from a donation prior to vaccine rollout. Intranasal administration of human CCP resulted in a reactive (immune) response in hamster lungs, however this was not observed upon administration of hamster CCP. INTERPRETATION We conclude that CCP is an effective prophylactic when used directly at the site of primary infection. This option should be considered in future prepandemic preparedness plans. FUNDING Flanders Innovation & Entrepreneurship (VLAIO) and the Foundation for Scientific Research of the Belgian Red Cross Flanders.
Collapse
Affiliation(s)
- Elise Wouters
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Caro Verbrugghe
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Rana Abdelnabi
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Rosalie Devloo
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | | | - Dirk Jochmans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | | | - Birgit Weynand
- KU Leuven Department of Imaging and Pathology, Translational Cell and Tissue Research, Division of Translational Cell and Tissue Research, B-3000, Leuven, Belgium
| | - Veerle Compernolle
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Blood Services of the Belgian Red Cross-Flanders, Mechelen, Belgium; Transfusion Innovation Center, Belgian Red Cross-Flanders, Ghent, Belgium
| | - Johan Neyts
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory of Virology and Chemotherapy, B-3000, Leuven, Belgium
| | - Hendrik B Feys
- Transfusion Research Center, Belgian Red Cross-Flanders, Ghent, Belgium; Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; Blood Services of the Belgian Red Cross-Flanders, Mechelen, Belgium.
| |
Collapse
|
5
|
Tworek A, Jaroń K, Cicha M, Rydzewski A, Wierzba W, Zaczyński A, Król Z, Rydzewska G. The persistence of SARS-CoV-2 neutralizing antibodies after COVID-19: A one-year observation. Is a SARS-CoV-2 vaccination booster dose necessary? Cent Eur J Immunol 2023; 48:92-96. [PMID: 37692027 PMCID: PMC10485689 DOI: 10.5114/ceji.2023.126206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 02/27/2023] [Indexed: 09/12/2023] Open
Abstract
Introduction The aim of this study was to investigate the persistence of SARS-CoV-2 neutralizing antibodies (NAbs) one year after contracting COVID-19. Material and methods The study included 38 patients - 34 men and 4 women - suffering from COVID-19 between March 15 and May 26, 2020. The median age in the group was 31 years, ranging from 22 to 67 years. The levels of neutralizing antibodies were measured at three time-points - baseline, 6 months, and 12 months. The primary endpoint was a post-infection positive result for NAbs (> 15 AU/ml; Liaison SARS-CoV-2 S1/S2 IgG quantitative test) 12 months after infection. Results The median level of NAbs after 12 months was 26.5 AU/ml. At the end of observation (12 months), 21 of the 38 patients had a NAb level of >15 AU/ml (positive). The median antibody half-life was 5.8 months. Conclusions A high percentage of the patients maintained positive levels of antibodies 6 and 12 months after COVID-19 infection. The dynamics of the antibody level decline suggests the need for booster vaccination at least once a year.
Collapse
Affiliation(s)
- Adam Tworek
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Krzysztof Jaroń
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Małgorzata Cicha
- Diagnostic Laboratory of Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Andrzej Rydzewski
- Department of Internal Medicine, Nephrology and Transplantation Medicine, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
- Department of Internal Medicine, Nephrology and Transplantation Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Waldemar Wierzba
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
- University of Humanities and Economics in Łódz, Satellite Campus in Warsaw, Warsaw, Poland
| | - Artur Zaczyński
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Zbigniew Król
- Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
| | - Grażyna Rydzewska
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Interior and Administration, Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
6
|
Reduced Mortality among COVID-19 ICU Patients after Treatment with HemoClear Convalescent Plasma in Suriname. mBio 2023; 14:e0337922. [PMID: 36815780 PMCID: PMC10127603 DOI: 10.1128/mbio.03379-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023] Open
Abstract
Convalescent plasma is a promising therapy for coronavirus disease 2019 (COVID-19), but its efficacy in intensive care unit (ICU) patients in low- and middle-income country settings such as Suriname is unknown. Bedside plasma separation using the HemoClear device made convalescent plasma therapy accessible as a treatment option in Suriname. Two hundred patients with severe SARS-CoV-2 infection requiring intensive care were recruited. Fifty eight patients (29%) received COVID-19 convalescent plasma (CCP) treatment in addition to standard of care (SOC). The CCP treatment and SOC groups were matched by age, sex, and disease severity scores. Mortality in the CCP treatment group was significantly lower than that in the SOC group (21% versus 39%; Fisher's exact test P = 0.0133). Multivariate analysis using ICU days showed that CCP treatment reduced mortality (hazard ratio [HR], 0.35; 95% confidence interval [CI], 0.18 to 0.66; P = 0.001), while complication of acute renal failure (creatinine levels, >110 mol/L; HR, 4.45; 95% CI, 2.54 to 7.80; P < 0.0001) was independently associated with death. Decrease in chest X-ray score in the CCP treatment group (median -3 points, interquartile range [IQR] -4 to -1) was significantly greater than that in the SOC group (median -1 point, IQR -3 to 1, Mann-Whitney test P = 0.0004). Improvement in the PaO2/FiO2 ratio was also significantly greater in the CCP treatment group (median 83, IQR 8 to 140) than in the SOC group (median 35, IQR -3 to 92, Mann-Whitney P = 0.0234). Further research is needed for HemoClear-produced CCP as a therapy for SARS-CoV-2 infection together with adequately powered, randomized controlled trials. IMPORTANCE This study compares mortality and other endpoints between intensive care unit COVID-19 patients treated with convalescent plasma plus standard of care (CCP), and a control group of patients hospitalized in the same medical ICU facility treated with standard of care alone (SOC) in a low- and middle-income country (LMIC) setting using bedside donor whole blood separation by gravity (HemoClear) to produce the CCP. It demonstrates a significant 65% survival improvement in HemoClear-produced CCP recipients (HR, 0.35; 95% CI, 0.19 to 0.66; P = 0.001). Although this is an exploratory study, it clearly shows the benefit of using the HemoClear-produced CCP in ICU patients in the Suriname LMIC setting. Additional studies could further substantiate our findings and their applicability for both LMICs and high-income countries and the use of CCP as a prepared readiness method to combat new viral pandemics.
Collapse
|
7
|
Impact of Convalescent Plasma Therapy in Hospitalized Patients With Severe COVID-19. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2022. [DOI: 10.1097/ipc.0000000000001161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
8
|
Amin A, Moon R, Agiro A, Rosenthal N, Brown H, Legg R, Pottorf W. In-hospital mortality, length of stay, and hospitalization cost of COVID-19 patients with and without hyperkalemia. Am J Med Sci 2022; 364:444-453. [PMID: 35490703 PMCID: PMC9050185 DOI: 10.1016/j.amjms.2022.04.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/11/2022] [Accepted: 04/22/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND Hyperkalemia (HK) may be associated with poor clinical outcomes among COVID-19 patients. This study aimed to describe the prevalence of HK and evaluate the associations between HK and in-hospital mortality, intensive care unit (ICU) admission, length of hospital stay (LOS), and hospitalization cost among COVID-19 inpatients. METHODS A retrospective cohort study was conducted using a large hospital discharge database (PINC AI Healthcare Database) for COVID-19 inpatients discharged between April 1 and August 31, 2020. HK was defined with discharge diagnosis and potassium binder use. RESULTS Of 192,182 COVID-19 inpatients, 12% (n = 22,702) had HK. HK patients were more likely to be older (median age 67 vs 63 years), male (63% vs 50%), black (30% vs 22%), and have a history of chronic kidney disease (45% vs 16%) or diabetes mellitus (55% vs 35%) than non-HK patients (all p<.001). A significantly higher proportion of patients with HK had in-hospital mortality (42% vs 11%, p<.001) than those without HK; this was persistent after adjusting for confounders (adjusted odds ratio [aOR] 1.69, 95% confidence interval [CI]1.62-1.77). Patients with HK were also more likely to be admitted to ICU (aOR 1.05, 95% CI 1.01-1.09), incur higher cost of care (adjusted mean difference $5,389) and have longer LOS (adjusted mean difference 1.3 days) than non-HK patients. CONCLUSIONS Presence of HK was independently associated with higher in-hospital mortality, LOS, and cost of care among COVID-19 inpatients. Detecting and closely monitoring HK are recommended to improve clinical outcomes and reduce LOS and healthcare cost among COVID-19 patients.
Collapse
Affiliation(s)
- Alpesh Amin
- Department of Medicine, School of Medicine, University of California - Irvine, Orange, CA, USA
| | - Rena Moon
- PINC AI Applied Sciences, Premier Inc., Charlotte, NC, USA,Corresponding author at: Rena Moon, MD, MPH, Principal Research Scientist, PINC AI Applied Sciences, Premier Inc., 13034 Ballantyne Corporate Place, Charlotte, NC, 28277
| | | | - Ning Rosenthal
- PINC AI Applied Sciences, Premier Inc., Charlotte, NC, USA
| | - Harold Brown
- PINC AI Applied Sciences, Premier Inc., Charlotte, NC, USA
| | | | | |
Collapse
|
9
|
Focosi D, Franchini M, Pirofski LA, Burnouf T, Paneth N, Joyner MJ, Casadevall A. COVID-19 Convalescent Plasma and Clinical Trials: Understanding Conflicting Outcomes. Clin Microbiol Rev 2022; 35:e0020021. [PMID: 35262370 PMCID: PMC9491201 DOI: 10.1128/cmr.00200-21] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Convalescent plasma (CP) recurs as a frontline treatment in epidemics because it is available as soon as there are survivors. The COVID-19 pandemic represented the first large-scale opportunity to shed light on the mechanisms of action, safety, and efficacy of CP using modern evidence-based medicine approaches. Studies ranging from observational case series to randomized controlled trials (RCTs) have reported highly variable efficacy results for COVID-19 CP (CCP), resulting in uncertainty. We analyzed variables associated with efficacy, such as clinical settings, disease severity, CCP SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) antibody levels and function, dose, timing of administration (variously defined as time from onset of symptoms, molecular diagnosis, diagnosis of pneumonia, or hospitalization, or by serostatus), outcomes (defined as hospitalization, requirement for ventilation, clinical improvement, or mortality), CCP provenance and time for collection, and criteria for efficacy. The conflicting trial results, along with both recent WHO guidelines discouraging CCP usage and the recent expansion of the FDA emergency use authorization (EUA) to include outpatient use of CCP, create confusion for both clinicians and patients about the appropriate use of CCP. A review of 30 available RCTs demonstrated that signals of efficacy (including reductions in mortality) were more likely if the CCP neutralizing titer was >160 and the time to randomization was less than 9 days. The emergence of the Omicron variant also reminds us of the benefits of polyclonal antibody therapies, especially as a bridge to the development and availability of more specific therapies.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Liise-anne Pirofski
- Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, New York, New York, USA
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Nigel Paneth
- Department of Epidemiology & Biostatistics and Pediatrics & Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
- Department of Pediatrics & Human Development, College of Human Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Michael J. Joyner
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
10
|
Lachert E, Lasocka J, Bielawski A, Sulkowska E, Guz K, Pyrc K, Dabrowska A, Wawryniuk-Malmon A, Letowska M, Tomasiewicz K, Grabarczyk P. Human Intramuscular Hyperimmune Gamma Globulin (hIHGG) Anti-SARS-CoV-2-Characteristics of Intermediates and Final Product. Viruses 2022; 14:1328. [PMID: 35746798 PMCID: PMC9227433 DOI: 10.3390/v14061328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 12/05/2022] Open
Abstract
This study aims to characterize the intermediates, and the final product (FP) obtained during the production of human intramuscular hyperimmune gamma globulin anti-SARS-CoV-2 (hIHGG anti-SARS-CoV-2) and to determine its stability. Material and methods: hIHGG anti-SARS-CoV-2 was fractionated from 270 convalescent plasma donations with the Cohn method. Prior to fractionation, the plasma was inactivated (Theraflex MB Plasma). Samples were defined using enzyme immunoassays (EIA) for anti-S1, anti-RBD S1, and anti-N antibodies, and neutralization assays with SARS-CoV-2 (VN) and pseudoviruses (PVN, decorated with SARS-CoV-2 S protein). Results were expressed as a titer (EIA) or 50% of the neutralization titer (IC50) estimated in a four-parameter nonlinear regression model. Results: Concentration of anti-S1 antibodies in plasma was similar before and after inactivation. Following fractionation, the anti-S1, anti-RBD, and anti-N (total tests) titers in FP were concentrated approximately 15-fold from 1:4 to 1:63 (1800 BAU/mL), 7-fold from 1:111 to 1:802 and from 1:13 to 1:88, respectively. During production, the IgA (anti-S1) antibody titer was reduced to an undetectable level and the IgM (anti-RBD) titer from 1:115 to 1:24. The neutralizing antibodies (nAb) titer increased in both VN (from 1:40 to 1:160) and PVN (IC50 from 63 to 313). The concentration of specific IgG in the FP did not change significantly for 14 months. Conclusions: The hIHGG anti-SARS-CoV-2 was stable, with concentration up to approximately 15-fold nAb compared to the source plasma pool.
Collapse
Affiliation(s)
- Elzbieta Lachert
- Department of Transfusion Medicine, Institute of Hematology and Transfusion Medicine, Indiry Gandhi 14 Str., 02-776 Warsaw, Poland; (E.L.); (J.L.); (M.L.)
| | - Joanna Lasocka
- Department of Transfusion Medicine, Institute of Hematology and Transfusion Medicine, Indiry Gandhi 14 Str., 02-776 Warsaw, Poland; (E.L.); (J.L.); (M.L.)
| | - Artur Bielawski
- Biomed Company, Uniwersytecka 10 Str., 20-029 Lublin, Poland; (A.B.); (A.W.-M.)
| | - Ewa Sulkowska
- Department of Virology, Institute of Hematology and Transfusion Medicine, Chocimska 5 Str., 00-957 Warsaw, Poland;
| | - Katarzyna Guz
- Department of Immunohematology and Transfusion Medicine, Institute of Hematology and Transfusion Medicine, Chocimska 5 Str., 00-957 Warsaw, Poland;
| | - Krzysztof Pyrc
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A Str., 30-387 Krakow, Poland; (K.P.); (A.D.)
| | - Agnieszka Dabrowska
- Virogenetics Laboratory of Virology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A Str., 30-387 Krakow, Poland; (K.P.); (A.D.)
- Microbiology Department, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7A Str., 30-387 Krakow, Poland
| | | | - Magdalena Letowska
- Department of Transfusion Medicine, Institute of Hematology and Transfusion Medicine, Indiry Gandhi 14 Str., 02-776 Warsaw, Poland; (E.L.); (J.L.); (M.L.)
| | - Krzysztof Tomasiewicz
- Department of Infectious Diseases, Medical University of Lublin, Stanislawa Staszica 16 Str., 20-081 Lublin, Poland;
| | - Piotr Grabarczyk
- Department of Virology, Institute of Hematology and Transfusion Medicine, Chocimska 5 Str., 00-957 Warsaw, Poland;
| |
Collapse
|
11
|
Beraud M, Goodhue Meyer E, Lozano M, Bah A, Vassallo R, Brown BL. Lessons learned from the use of convalescent plasma for the treatment of COVID-19 and specific considerations for immunocompromised patients. Transfus Apher Sci 2022; 61:103355. [PMID: 35063360 PMCID: PMC8757642 DOI: 10.1016/j.transci.2022.103355] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) convalescent plasma (CovCP) infusions have been widely used for the treatment of hospitalized patients with COVID-19. The aims of this narrative review were to analyze the safety and efficacy of CovCP infusions in the overall population and in immunocompromised patients with COVID-19 and to identify the lessons learned concerning the use of convalescent plasma (CP) to fill treatment gaps for emerging viruses. Systematic searches (PubMed, Scopus, and COVID-19 Research) were conducted to identify peer-reviewed articles and pre-prints published between March 1, 2020 and May 1, 2021 on the use of CovCP for the treatment of patients with COVID-19. From 261 retrieved articles, 37 articles reporting robust controlled studies in the overall population of patients with COVID-19 and 9 articles in immunocompromised patients with COVID-19 were selected. While CovCP infusions are well tolerated in both populations, they do not seem to improve clinical outcomes in critically-ill patients with COVID-19 and no conclusion could be drawn concerning their potential benefits in immunocompromised patients with COVID-19. To be better prepared for future epidemics/pandemics and to evaluate potential benefits of CP treatment, only CP units with high neutralizing antibodies (NAbs) titers should be infused in patients with low NAb titers, patient eligibility criteria should be based on the disease pathophysiology, and measured clinical outcomes and methods should be comparable across studies. Even if CovCP infusions did not improve clinical outcomes in patients with COVID-19, NAb-containing CP infusions remain a safe, widely available and potentially beneficial treatment option for future epidemics/pandemics.
Collapse
Affiliation(s)
- Mickael Beraud
- Terumo Blood and Cell Technologies Europe NV, Ikaroslaan 41, 1930, Zaventem, Belgium.
| | - Erin Goodhue Meyer
- Terumo Blood and Cell Technologies, 10811 W Collins Ave, Lakewood, CO, 80215, United States.
| | - Miquel Lozano
- Department of Hemotherapy and Hemostasis, ICMHO, University Clinic Hospital, IDIBAPS, University of Barcelona, Villarroel 170, 08036, Barcelona, Catalonia, Spain.
| | - Aicha Bah
- Terumo Blood and Cell Technologies Europe NV, Ikaroslaan 41, 1930, Zaventem, Belgium.
| | - Ralph Vassallo
- Vitalant, 6210 E Oak St, Scottsdale, AZ, 85257, United States.
| | - Bethany L Brown
- American Red Cross, Biomedical Services, Holland Laboratory for the Biomedical Sciences, 15601 Crabbs Branch Way, Rockville, MD, 20855, United States.
| |
Collapse
|
12
|
Song AT, Rocha V, Mendrone-Júnior A, Calado RT, De Santis GC, Benites BD, Costa-Lima C, Vargas T, Marques LS, Fernandes JC, Breda FC, Wendel S, Fachini R, Rizzo LV, Kutner JM, Avelino-Silva VI, Machado RR, Durigon EL, Chevret S, Kallas EG. Treatment of severe COVID-19 patients with either low- or high-volume of convalescent plasma versus standard of care: A multicenter Bayesian randomized open-label clinical trial (COOP-COVID-19-MCTI). LANCET REGIONAL HEALTH. AMERICAS 2022; 10:100216. [PMID: 35308034 PMCID: PMC8923059 DOI: 10.1016/j.lana.2022.100216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background Administration of convalescent plasma may serve as an adjunct to supportive treatment to prevent COVID-19 progression and death. We aimed to evaluate the efficacy and safety of 2 volumes of intravenous convalescent plasma (CP) with high antibody titers for the treatment of severe cases of COVID-19. Methods We conducted a Bayesian, randomized, open-label, multicenter, controlled clinical trial in 7 Brazilian hospitals. Adults admitted to hospital with positive RT-PCR for SARS-CoV2, within 10 days of the symptom onset, were eligible. Patients were randomly assigned (1:1:1) to receive standard of care (SoC) alone, or in combination with 200 mL (150-300 mL) of CP (Low-volume), or 400 mL (300-600 mL) of CP (High-volume); infusion had to be performed within 24 h of randomization. Randomization was centralized, stratified by center. The primary outcome was the time until clinical improvement up to day 28, measured by the WHO ten-point scale, assessed in the intention-to-treat population. Interim and terminal analyses were performed in a Bayesian framework. Trial registered at ClinicalTrials.gov: NCT04415086. Findings Between June 2, 2020, and November 18, 2020, 129 patients were enrolled and randomly assigned to SoC (n = 42), Low-volume (n = 43) or High-volume (n = 44) CP. Donors presented a median titer of neutralizing antibodies of 1:320 (interquartile range, 1:160 to 1:1088). No evidence of any benefit of convalescent plasma was observed, with Bayesian estimate of 28-day clinical improvement of 72.7% (95%CI, 58.8 to 84.7) in the SoC versus 64.1% (95%ci, 53.8 to 73.7) in the pooled experimental groups (mean difference of -8.7%, 95%CI, -24.6 to 8.2). There was one case of cutaneous mild allergic reaction related to plasma transfusion and one case of suspected transfusion-related acute lung injury but deemed not to be related to convalescent plasma infusion. Interpretation In this prospective, randomized trial of adult hospitalized patients with severe COVID-19, convalescent plasma was not associated with clinical benefits. Funding Brazilian Ministry of Science, Technology and Innovation, Fundação de Amparo à Pesquisa do Estado de São Paulo.
Collapse
Affiliation(s)
- Alice T.W. Song
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Vanderson Rocha
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Rodrigo T. Calado
- Hospital das Clínicas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Gil C. De Santis
- Hospital das Clínicas, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Bruno D. Benites
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Carolina Costa-Lima
- Centro de Hematologia e Hemoterapia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Taiani Vargas
- Hospital Nossa Senhora da Conceição, Grupo Hospitalar Conceição, Porto Alegre, Brazil
| | - Leonardo S. Marques
- Hospital Nossa Senhora da Conceição, Grupo Hospitalar Conceição, Porto Alegre, Brazil
| | | | | | | | | | - Luiz V. Rizzo
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | | | - Vivian I. Avelino-Silva
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rafael R.G. Machado
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Edison L. Durigon
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Sylvie Chevret
- Biostatistical Department, Hôpital Saint-Louis, Paris University, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Esper G. Kallas
- Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
13
|
Fodor E, Müller V, Iványi Z, Berki T, Kuten Pella O, Hornyák I, Ambrus M, Sárkány Á, Skázel Á, Madár Á, Kardos D, Kemenesi G, Földes F, Nagy S, Matusovits A, János N, Tordai A, Jakab F, Lacza Z. Early Transfusion of Convalescent Plasma Improves the Clinical Outcome in Severe SARS-CoV2 Infection. Infect Dis Ther 2022; 11:293-304. [PMID: 34817840 PMCID: PMC8611245 DOI: 10.1007/s40121-021-00514-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/26/2021] [Indexed: 01/10/2023] Open
Abstract
INTRODUCTION Plasma harvested from convalescent COVID-19 patients (CCP) has been applied as first-line therapy in the early phase of the SARS-CoV2 pandemic through clinical studies using various protocols. METHODS We present data from a cohort of 267 hospitalized severe COVID-19 patients who received CCP. No transfusion-related complications were reported, indicating the overall safety of CCP therapy. RESULTS Patients who eventually died from COVID-19 received CCP significantly later (3.95 versus 5.22 days after hospital admission) and had higher interleukin 6 (IL-6) levels (28.9 pg/ml versus 102.5 pg/ml) than those who survived. In addition, CCP transfusion caused a significant reduction in the overall inflammatory status of the patients regardless of the severity of disease or outcome, as evidenced by decreasing C-reactive protein, IL6 and ferritin levels. CONCLUSION We conclude that CCP transfusion is a safe and effective supplementary treatment modality for hospitalized COVID-19 patients characterized by better expected outcome if applied as early as possible. We also observed that IL-6 may be a suitable laboratory parameter for patient selection and monitoring of CCP therapy effectiveness.
Collapse
Affiliation(s)
- Eszter Fodor
- Orthosera Kft, Budapest, 1149 Hungary
- Univesity of Physical Education, Budapest, 1223 Hungary
| | - Veronika Müller
- Department of Pulmonology, Semmelweis University, Budapest, 1083 Hungary
| | - Zsolt Iványi
- Department of Anesthesiology and Intensive Therapy, Semmelweis University, Budapest, 1082 Hungary
| | - Tímea Berki
- Department of Immunology and Biotechnology, University of Pécs, Budapest, 7643 Hungary
| | | | - István Hornyák
- Instute of Translational Medicine, Semmelweis University, Budapest, Hungary
| | - Mira Ambrus
- Univesity of Physical Education, Budapest, 1223 Hungary
| | - Ágnes Sárkány
- Szent György University Teaching Hospital, Székesfehérvár, 8000 Hungary
| | - Árpád Skázel
- Szent György University Teaching Hospital, Székesfehérvár, 8000 Hungary
| | - Ágnes Madár
- Univesity of Physical Education, Budapest, 1223 Hungary
| | | | - Gábor Kemenesi
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Fanni Földes
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Sándor Nagy
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Andrea Matusovits
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Nacsa János
- Hungarian National Blood Transfusion Service, Budapest, 1113 Hungary
| | - Attila Tordai
- Department of Transfusiology, Semmelweis University, Budapest, 1089 Hungary
| | - Ferenc Jakab
- Szentágothai Research Center, National Laboratory of Virology, Univesity of Pécs, Pécs, 7622 Hungary
| | - Zsombor Lacza
- Orthosera Kft, Budapest, 1149 Hungary
- Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary
- Univesity of Physical Education, Budapest, 1223 Hungary
| |
Collapse
|
14
|
Focosi D, Franchini M, Pirofski LA, Maggi F, Casadevall A. Is SARS-CoV-2 viral clearance in nasopharyngeal swabs an appropriate surrogate marker for clinical efficacy of neutralising antibody-based therapeutics? Rev Med Virol 2021; 32:e2314. [PMID: 34861088 DOI: 10.1002/rmv.2314] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 12/15/2022]
Abstract
Viral clearance is likely the best way to assess the efficacy of antibody-based therapies. Although antibodies can mediate a variety of effects that include modulation of inflammation, the demonstration of viral clearance provides an accessible and measurable parameter that can be used to evaluate efficacy and determine dosing. Therefore, it is important to ascertain the ability of monoclonal antibodies and convalescent plasma to effect viral clearance. For COVID-19, which is caused by the respiratory virus SARS-CoV-2, the most common assay to assess viral clearance is via a nasopharyngeal swab (NPS). However, assessment of antibody efficacy by sampling this site may be misleading because it may not be as accessible to serum antibodies as respiratory secretions or circulating blood. Adding to the complexity of assessing the efficacy of administered antibody, particularly in randomised controlled trials (RCTs) that enroled patients at different times after the onset of COVID-19 symptoms, viral clearance may also be mediated by endogenous antibody. In this article we critically review available data on viral clearance in RCTs, matched control studies, case series and case reports of antibody therapies in an attempt to identify variables that contribute to antibody efficacy and suggest optimal strategies for future studies.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Liise-Anne Pirofski
- Division of Infectious Diseases, Departments of Medicine, Microbiology and Immunology, Albert Einstein College of Medicine and Montefiore Medical Center, New York City, New York, USA
| | - Fabrizio Maggi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy.,Laboratory of Microbiology, ASST Sette Laghi, Varese, Italy
| | - Arturo Casadevall
- Department of Medicine, Johns Hopkins School of Public Health and School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
15
|
Körper S, Appl T, Jahrsdörfer B, Lotfi R, Rojewski M, Wuchter P, Tonn T, Bakchoul T, Karatas M, Schmidt M, Klüter H, Seifried E, Schrezenmeier H. Randomisierte Studien zum Einsatz von Rekonvaleszentenplasma bei COVID-19: eine Standortbestimmung. TRANSFUSIONSMEDIZIN 2021. [DOI: 10.1055/a-1521-7884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
ZusammenfassungPlasma von genesenen Spendern mit COVID-19 (COVID-19 Convalescent Plasma, CCP) wurde als Behandlungsoption für Patienten mit COVID-19 in Betracht gezogen. In der ersten Phase der Pandemie wurden mehrere Fallberichte und Fallkontrollstudien mit Hinweisen auf eine therapeutische Wirkung veröffentlicht. Inzwischen liegen die Ergebnisse zahlreicher randomisierter Studien vor. Die Studien unterscheiden sich in vielen Aspekten, u. a. in den Patientenpopulationen, die von ambulanten Patienten mit mildem COVID-19 bis zu kritisch Kranken reichten, wie auch den Endpunkten. Ebenso war der Behandlungsstandard innerhalb der klinischen Studien sehr unterschiedlich. Vor allem aber unterschied sich das Prüfpräparat CCP erheblich in Bezug auf das Behandlungsschema, das Volumen und den Gehalt an Antikörpern. Im Folgenden werden wir die Ergebnisse der bisher publizierten randomisierten Studien diskutieren. Aus den bisher veröffentlichten Ergebnissen lässt sich eine Wirksamkeit von CCP
ableiten, sofern es sehr hohe Titer neutralisierender Antikörper enthält und früh im Krankheitsverlauf verabreicht wird. COVID-19-Rekonvaleszenten-Plasma ist noch keine Routinebehandlung und sollte möglichst weiter in klinischen Studien untersucht werden. Neu konzipierte Studien sollten sich auf die frühe Anwendung von CCP mit einem hohen Gehalt an neutralisierenden Antikörpern bei Patienten mit hohem Risiko für eine Progression zu einer schweren COVID-19-Erkrankung konzentrieren und wichtige Begleitmedikationen kontrollieren.
Collapse
Affiliation(s)
- Sixten Körper
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Thomas Appl
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Bernd Jahrsdörfer
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Ramin Lotfi
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Markus Rojewski
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| | - Patrick Wuchter
- Institut für Transfusionsmedizin und Immunologie, Medizinische Fakultät Mannheim, Universität Heidelberg; DRK-Blutspendedienst Baden-Württemberg – Hessen, Deutschland
| | - Torsten Tonn
- Experimentelle Transfusionsmedizin, Technische Universität Dresden, Deutsches Rotes Kreuz Bluttransfusionsdienst Nord-Ost gGmbH, Dresden, Deutschland
| | - Tamam Bakchoul
- Institut für klinische und experimentelle Transfusionsmedizin, Universitätsklinikum Tübingen, Tübingen, Deutschland
| | - Mesut Karatas
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Michael Schmidt
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Harald Klüter
- Institut für Transfusionsmedizin und Immunologie, Medizinische Fakultät Mannheim, Universität Heidelberg; DRK-Blutspendedienst Baden-Württemberg – Hessen, Deutschland
| | - Erhard Seifried
- Institut für Transfusionsmedizin und Immunhämatologie, DRK-Bluttransfusionsdienst Baden-Württemberg – Hessen, Frankfurt, Deutschland
| | - Hubert Schrezenmeier
- Institut für Klinische Transfusionsmedizin und Immungenetik Ulm, DRK-Blutspendedienst Baden-Württemberg-Hessen und Institut für Transfusionsmedizin, Universität Ulm
| |
Collapse
|
16
|
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: 6.7] [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
|
17
|
Arnold Egloff SA, Junglen A, Restivo JS, Wongskhaluang M, Martin C, Doshi P, Schlauch D, Fromell G, Sears LE, Correll M, Burris HA, LeMaistre CF. Convalescent plasma associates with reduced mortality and improved clinical trajectory in patients hospitalized with COVID-19. J Clin Invest 2021; 131:e151788. [PMID: 34464352 DOI: 10.1172/jci151788] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/26/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUNDEvidence supporting convalescent plasma (CP), one of the first investigational treatments for coronavirus disease 2019 (COVID-19), has been inconclusive, leading to conflicting recommendations. The primary objective was to perform a comparative effectiveness study of CP for all-cause, in-hospital mortality in patients with COVID-19.METHODSThe multicenter, electronic health records-based, retrospective study included 44,770 patients hospitalized with COVID-19 in one of 176 HCA Healthcare-affiliated community hospitals. Coarsened exact matching (1:k) was employed, resulting in a sample of 3774 CP and 10,687 comparison patients.RESULTSExamination of mortality using a shared frailty model, controlling for concomitant medications, date of admission, and days from admission to transfusion, demonstrated a significant association of CP with lower mortality risk relative to the comparison group (adjusted hazard ratio [aHR] = 0.71; 95% CI, 0.59-0.86; P < 0.001). Examination of patient risk trajectories, represented by 400 clinico-demographic features from our real-time risk model (RTRM), indicated that patients who received CP recovered more quickly. The stratification of days to transfusion revealed that CP within 3 days after admission, but not within 4 to 7 days, was associated with a significantly lower mortality risk (aHR = 0.53; 95% CI, 0.47-0.60; P < 0.001). CP serology level was inversely associated with mortality when controlling for its interaction with days to transfusion (HR = 0.998; 95% CI, 0.997-0.999; P = 0.013), yet it did not reach univariable significance.CONCLUSIONSThis large, diverse, multicenter cohort study demonstrated that CP, compared with matched controls, is significantly associated with reduced risk of in-hospital mortality. These observations highlight the utility of real-world evidence and suggest the need for further evaluation prior to abandoning CP as a viable therapy for COVID-19.FUNDINGThis research was supported in whole by HCA Healthcare and/or an HCA Healthcare-affiliated entity, including Sarah Cannon and Genospace.
Collapse
Affiliation(s)
- Shanna A Arnold Egloff
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Angela Junglen
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Joseph Sa Restivo
- HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | | | - Casey Martin
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Pratik Doshi
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Daniel Schlauch
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Gregg Fromell
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Lindsay E Sears
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Mick Correll
- Sarah Cannon, Nashville, Tennessee, USA.,Genospace, Boston, Massachusetts, USA
| | - Howard A Burris
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| | - Charles F LeMaistre
- Sarah Cannon, Nashville, Tennessee, USA.,HCA Healthcare, HCA Research Institute (HRI), Nashville, Tennessee, USA
| |
Collapse
|
18
|
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: 1.0] [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
|
19
|
Ye F, Lin X, Chen Z, Yang F, Lin S, Yang J, Chen H, Sun H, Wang L, Wen A, Zhang X, Dai Y, Cao Y, Yang J, Shen G, Yang L, Li J, Wang Z, Wang W, Wei X, Lu G. S19W, T27W, and N330Y mutations in ACE2 enhance SARS-CoV-2 S-RBD binding toward both wild-type and antibody-resistant viruses and its molecular basis. Signal Transduct Target Ther 2021; 6:343. [PMID: 34531369 PMCID: PMC8444507 DOI: 10.1038/s41392-021-00756-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023] Open
Abstract
SARS-CoV-2 recognizes, via its spike receptor-binding domain (S-RBD), human angiotensin-converting enzyme 2 (ACE2) to initiate infection. Ecto-domain protein of ACE2 can therefore function as a decoy. Here we show that mutations of S19W, T27W, and N330Y in ACE2 could individually enhance SARS-CoV-2 S-RBD binding. Y330 could be synergistically combined with either W19 or W27, whereas W19 and W27 are mutually unbeneficial. The structures of SARS-CoV-2 S-RBD bound to the ACE2 mutants reveal that the enhanced binding is mainly contributed by the van der Waals interactions mediated by the aromatic side-chains from W19, W27, and Y330. While Y330 and W19/W27 are distantly located and devoid of any steric interference, W19 and W27 are shown to orient their side-chains toward each other and to cause steric conflicts, explaining their incompatibility. Finally, using pseudotyped SARS-CoV-2 viruses, we demonstrate that these residue substitutions are associated with dramatically improved entry-inhibition efficacy toward both wild-type and antibody-resistant viruses. Taken together, our biochemical and structural data have delineated the basis for the elevated S-RBD binding associated with S19W, T27W, and N330Y mutations in ACE2, paving the way for potential application of these mutants in clinical treatment of COVID-19.
Collapse
Affiliation(s)
- Fei Ye
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xi Lin
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zimin Chen
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fanli Yang
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Sheng Lin
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jing Yang
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hua Chen
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Honglu Sun
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lingling Wang
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ao Wen
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xindan Zhang
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yushan Dai
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Cao
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Disaster Medicine Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingyun Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guobo Shen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Yang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiong Li
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenling Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Guangwen Lu
- West China Hospital Emergency Department (WCHED), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
20
|
Direct Comparison of Clinical Characteristics, Outcomes, and Risk Prediction in Patients with COVID-19 and Controls-A Prospective Cohort Study. J Clin Med 2021; 10:jcm10122672. [PMID: 34204453 PMCID: PMC8234319 DOI: 10.3390/jcm10122672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 01/08/2023] Open
Abstract
Most studies investigating early risk predictors in coronavirus disease 19 (COVID-19) lacked comparison with controls. We aimed to assess and directly compare outcomes and risk predictors at time of emergency department (ED) presentation in COVID-19 and controls. Consecutive patients presenting to the ED with suspected COVID-19 were prospectively enrolled. COVID-19-patients were compared with (i) patients tested negative (overall controls) and (ii) patients tested negative, who had a respiratory infection (respiratory controls). Primary outcome was the composite of intensive care unit (ICU) admission and death at 30 days. Among 1081 consecutive cases, 191 (18%) were tested positive for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and 890 (82%) were tested negative (overall controls), of which 323 (30%) had a respiratory infection (respiratory controls). Incidence of the composite outcome was significantly higher in COVID-19 (23%) as compared with the overall control group (10%, adjusted-HR 2.45 (95%CI, 1.61–3.74), p < 0.001) or the respiratory control group (10%, adjusted-HR 2.93 (95%CI, 1.66–5.17), p < 0.001). Blood oxygen saturation, age, high-sensitivity troponin, c-reactive protein, and lactate dehydrogenase were identified as the strongest predictors of poor outcome available at time of ED presentation in COVID-19 with highly comparable prognostic utility in overall and respiratory controls. In conclusion, patients presenting to the ED with COVID-19 have a worse outcome than controls, even after adjustment for differences in baseline characteristics. Most predictors of poor outcome in COVID-19 were not restricted to COVID-19, but of comparable prognostic utility in controls and therefore generalizable to unselected patients with suspected COVID-19.
Collapse
|
21
|
Corti D, Purcell LA, Snell G, Veesler D. Tackling COVID-19 with neutralizing monoclonal antibodies. Cell 2021; 184:3086-3108. [PMID: 34087172 PMCID: PMC8152891 DOI: 10.1016/j.cell.2021.05.005] [Citation(s) in RCA: 240] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies (mAbs) have revolutionized the treatment of several human diseases, including cancer and autoimmunity and inflammatory conditions, and represent a new frontier for the treatment of infectious diseases. In the last 20 years, innovative methods have allowed the rapid isolation of mAbs from convalescent subjects, humanized mice, or libraries assembled in vitro and have proven that mAbs can be effective countermeasures against emerging pathogens. During the past year, an unprecedentedly large number of mAbs have been developed to fight coronavirus disease 2019 (COVID-19). Lessons learned from this pandemic will pave the way for the development of more mAb-based therapeutics for other infectious diseases. Here, we provide an overview of SARS-CoV-2-neutralizing mAbs, including their origin, specificity, structure, antiviral and immunological mechanisms of action, and resistance to circulating variants, as well as a snapshot of the clinical trials of approved or late-stage mAb therapeutics.
Collapse
MESH Headings
- Angiotensin-Converting Enzyme 2/chemistry
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/metabolism
- Animals
- Antibodies, Monoclonal/chemistry
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Neutralizing/chemistry
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
- Antibodies, Viral/chemistry
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- COVID-19/pathology
- COVID-19/virology
- Humans
- SARS-CoV-2/immunology
- SARS-CoV-2/isolation & purification
- SARS-CoV-2/metabolism
- Spike Glycoprotein, Coronavirus/immunology
- COVID-19 Drug Treatment
Collapse
Affiliation(s)
- Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland.
| | | | | | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
22
|
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: 15.3] [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
|
23
|
A Phase II Study on the Use of Convalescent Plasma for the Treatment of Severe COVID-19- A Propensity Score-Matched Control Analysis. Microorganisms 2021; 9:microorganisms9040806. [PMID: 33920489 PMCID: PMC8069820 DOI: 10.3390/microorganisms9040806] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 04/01/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
COVID-19 is a global pandemic associated with increased morbidity and mortality. Convalescent plasma (CP) infusion is a strategy of potential therapeutic benefit. We conducted a multicenter phase II study to evaluate the efficacy and safety of CP in patients with COVID-19, grade 4 or higher. To evaluate the efficacy of CP, a matched propensity score analysis was used comparing the intervention (n = 59) to a control group (n = 59). Sixty patients received CP within a median time of 7 days from symptom onset. During a median follow-up of 28.5 days, 56/60 patients fully recovered and 1 patient remained in the ICU. The death rate in the CP group was 3.4% vs. 13.6% in the control group. By multivariate analysis, CP recipients demonstrated a significantly reduced risk of death [HR: 0.04 (95% CI: 0.004–0.36), p: 0.005], significantly better overall survival by Kaplan–Meir analysis (p < 0.001), and increased probability of extubation [OR: 30.3 (95% CI: 2.64–348.9), p: 0.006]. Higher levels of antibodies in the CP were independently associated with significantly reduced risk of death. CP infusion was safe with only one grade 3 adverse event (AE), which easily resolved. CP used early may be a safe and effective treatment for patients with severe COVID-19 (trial number NCT04408209).
Collapse
|