151
|
Gachoud D, Pillonel T, Tsilimidos G, Battolla D, Dumas D, Opota O, Fontana S, Vollenweider P, Manuel O, Greub G, Bertelli C, Rufer N. Antibody response and intra-host viral evolution after plasma therapy in COVID-19 patients pre-exposed or not to B-cell-depleting agents. Br J Haematol 2022; 199:549-559. [PMID: 36101920 PMCID: PMC9539045 DOI: 10.1111/bjh.18450] [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: 07/15/2022] [Revised: 08/19/2022] [Accepted: 08/28/2022] [Indexed: 12/16/2022]
Abstract
Administration of plasma therapy may contribute to viral control and survival of COVID-19 patients receiving B-cell-depleting agents that impair humoral immunity. However, little is known on the impact of anti-CD20 pre-exposition on the kinetics of SARS-CoV-2-specific antibodies. Here, we evaluated the relationship between anti-spike immunoglobulin G (IgG) kinetics and the clinical status or intra-host viral evolution after plasma therapy in 36 eligible hospitalized COVID-19 patients, pre-exposed or not to B-cell-depleting treatments. The majority of anti-CD20 pre-exposed patients (14/17) showed progressive declines of anti-spike IgG titres following plasma therapy, contrasting with the 4/19 patients who had not received B-cell-depleting agents (p = 0.0006). Patients with antibody decay also depicted prolonged clinical symptoms according to the World Health Organization (WHO) severity classification (p = 0.0267) and SARS-CoV-2 viral loads (p = 0.0032) before complete virus clearance. Moreover, they had higher mutation rates than patients able to mount an endogenous humoral response (p = 0.015), including three patients with one to four spike mutations, potentially associated with immune escape. No relevant differences were observed between patients treated with plasma from convalescent and/or mRNA-vaccinated donors. Our study emphasizes the need for an individualized clinical care and follow-up in the management of COVID-19 patients with B-cell lymphopenia.
Collapse
Affiliation(s)
- David Gachoud
- Department of Internal MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland,Medical Education Unit, School of Medicine, Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
| | - Trestan Pillonel
- Institute of MicrobiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Gerasimos Tsilimidos
- Division of Hematology, Department of OncologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Dunia Battolla
- Department of Internal MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Dominique Dumas
- Department of Internal MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Onya Opota
- Institute of MicrobiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Stefano Fontana
- Interregional Blood Transfusion SRCBernSwitzerland,Faculty of Biology and MedicineUniversity of LausanneLausanneSwitzerland
| | - Peter Vollenweider
- Department of Internal MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Oriol Manuel
- Infectious Diseases Service and Transplantation Center, Department of MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Gilbert Greub
- Institute of MicrobiologyLausanne University Hospital and University of LausanneLausanneSwitzerland,Infectious Diseases Service and Transplantation Center, Department of MedicineLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Claire Bertelli
- Institute of MicrobiologyLausanne University Hospital and University of LausanneLausanneSwitzerland
| | - Nathalie Rufer
- Interregional Blood Transfusion SRCEpalingesSwitzerland,Department of OncologyLausanne University Hospital and University of LausanneEpalingesSwitzerland
| |
Collapse
|
152
|
Willeit P, Kimpel J, Winner H, Harthaller T, Schäfer H, Bante D, Falkensammer B, Rössler A, Riepler L, Ower C, Sacher M, von Laer D, Borena W. Seroprevalence of SARS-CoV-2 infection in the Tyrolean district of Schwaz at the time of the rapid mass vaccination in March 2021 following B.1.351-variant outbreak. Front Public Health 2022; 10:989337. [PMID: 36159252 PMCID: PMC9500479 DOI: 10.3389/fpubh.2022.989337] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/17/2022] [Indexed: 01/26/2023] Open
Abstract
In order to curb the rapid dissemination of the B.1.351 variant of SARS-CoV-2 in the district of Schwaz and beyond, the EU allocated additional vaccine doses at the beginning of March 2021 to implement a rapid mass vaccination of the population (16+). The aim of our study was to determine the seroprevalence of SARS-CoV-2 among the adult population in the district of Schwaz at the time of the implementation. Data on previous history of infections, symptoms and immunization status were collected using a structured questionnaire. Blood samples were used to determine SARS-CoV-2 specific anti-spike, anti-nucleocapsid and neutralizing antibodies. We recruited 2,474 individuals with a median age (IQR) of 42 (31-54) years. Using the official data on distribution of age and sex, we found a standardized prevalence of undocumented infections at 15.0% (95% CI: 13.2-16.7). Taken together with the officially documented infections, we estimated that 24.0% (95% CI: 22.5-25.6) of the adult population had prior SARS-CoV-2 infection. Hence, the proportion of undocumented infections identified by our study was 55.8% (95% CI: 52.7-58.5). With a vaccination coverage of 10% among the adults population at that time, we imply that a minimum of two-thirds of the target popuation was susceptible to the circulating threat when this unique campaign started.
Collapse
Affiliation(s)
- Peter Willeit
- Clinical Epidemiology Team, Medical University of Innsbruck, Innsbruck, Austria,Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Janine Kimpel
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hannes Winner
- Department of Economics, University of Salzburg, Salzburg, Austria
| | - Teresa Harthaller
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Helena Schäfer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - David Bante
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Falkensammer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annika Rössler
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lydia Riepler
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Cornelia Ower
- Department of Surgery, University Hospital of Trauma Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Sacher
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Dorothee von Laer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria
| | - Wegene Borena
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, Innsbruck, Austria,*Correspondence: Wegene Borena
| |
Collapse
|
153
|
Nevo L, Cahen-Peretz A, Vorontsov O, Frenkel R, Kabessa M, Cohen SM, Hamrani A, Oiknine-Djian E, Lipschuetz M, Goldman-Wohl D, Walfisch A, Kovo M, Neeman M, Yagel S, Wolf DG, Beharier O. Boosting maternal and neonatal humoral immunity following SARS-CoV-2 infection using a single messenger RNA vaccine dose. Am J Obstet Gynecol 2022; 227:486.e1-486.e10. [PMID: 35430228 PMCID: PMC9008977 DOI: 10.1016/j.ajog.2022.04.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 12/04/2022]
Abstract
BACKGROUND Post-COVID-19 vaccine boosting is a potent tool in the ongoing pandemic. Relevant data regarding this approach during pregnancy are lacking, which affects vaccination policy guidance, public acceptance, and vaccine uptake during pregnancy. We aimed to investigate the dynamics of anti-SARS-CoV-2 antibody levels following SARS-CoV-2 infection during pregnancy and to characterize the effect of a single postinfection vaccine booster dose on the anti-SARS-CoV-2 antibody levels in parturients in comparison with the levels in naïve vaccinated and convalescent, nonboosted parturients. STUDY DESIGN Serum samples prospectively collected from parturients and umbilical cords at delivery at our university-affiliated urban medical center in Jerusalem, Israel, from May to October 2021, were selected and analyzed in a case-control manner. Study groups comprised the following participants: a consecutive sample of parturients with a polymerase chain reaction-confirmed history of COVID-19 during any stage of pregnancy; and comparison groups selected according to time of exposure comprising (1) convalescent, nonboosted parturients with polymerase chain reaction-confirmed COVID-19; (2) convalescent parturients with polymerase chain reaction-confirmed COVID-19 who received a single booster dose of the BNT162b2 messenger RNA vaccine; and (3) infection-naïve, fully vaccinated parturients who received 2 doses of the BNT162b2 messenger RNA vaccine. Outcomes that were determined included maternal and umbilical cord blood anti-SARS-CoV-2 antibody levels detected at delivery, the reported side effects, and pregnancy outcomes. RESULTS A total of 228 parturients aged 18 to 45 years were included. Of those, samples from 64 were studied to characterize the titer dynamics following COVID-19 at all stages of pregnancy. The boosting effect was determined by comparing (1) convalescent (n=54), (2) boosted convalescent (n=60), and (3) naïve, fully vaccinated (n=114) parturients. Anti-SARS-CoV-2 antibody levels detected on delivery showed a gradual and significant decline over time from infection to delivery (r=0.4371; P=.0003). Of the gravidae infected during the first trimester, 34.6% (9/26) tested negative at delivery, compared with 9.1% (3/33) of those infected during the second trimester (P=.023). Significantly higher anti-SARS-CoV-2 antibody levels were observed among boosted convalescent than among nonboosted convalescent (17.6-fold; P<.001) and naïve vaccinated parturients (3.2-fold; P<.001). Similar patterns were observed in umbilical cord blood. Side effects in convalescent gravidae resembled those in previous reports of mild symptoms following COVID-19 vaccination during pregnancy. CONCLUSION Postinfection maternal humoral immunity wanes during pregnancy, leading to low or undetectable protective titers for a marked proportion of patients. A single boosting dose of the BNT162b2 messenger RNA vaccine induced a robust increase in protective titers for both the mother and newborn with moderate reported side effects.
Collapse
Affiliation(s)
- Lea Nevo
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adva Cahen-Peretz
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Olesya Vorontsov
- Clinical Virology Unit, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Lautenberg Center for General and Tumor Immunology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rachelli Frenkel
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Maor Kabessa
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sarah M Cohen
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Adar Hamrani
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Esther Oiknine-Djian
- Clinical Virology Unit, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Lipschuetz
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Debra Goldman-Wohl
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Asnat Walfisch
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Michal Kovo
- Department Obstetrics and Gynecology, Wolfson Medical Center, Holon, Israel
| | - Michal Neeman
- Department of Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Simcha Yagel
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Dana G Wolf
- Clinical Virology Unit, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel; Lautenberg Center for General and Tumor Immunology, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ofer Beharier
- Department of Obstetrics and Gynecology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
154
|
Sette A, Crotty S. Immunological memory to SARS-CoV-2 infection and COVID-19 vaccines. Immunol Rev 2022; 310:27-46. [PMID: 35733376 PMCID: PMC9349657 DOI: 10.1111/imr.13089] [Citation(s) in RCA: 169] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 05/04/2022] [Indexed: 12/12/2022]
Abstract
Immunological memory is the basis of protective immunity provided by vaccines and previous infections. Immunological memory can develop from multiple branches of the adaptive immune system, including CD4 T cells, CD8 T cells, B cells, and long-lasting antibody responses. Extraordinary progress has been made in understanding memory to SARS-CoV-2 infection and COVID-19 vaccines, addressing development; quantitative and qualitative features of different cellular and anatomical compartments; and durability of each cellular component and antibodies. Given the sophistication of the measurements; the size of the human studies; the use of longitudinal samples and cross-sectional studies; and head-to-head comparisons between infection and vaccines or between multiple vaccines, the understanding of immune memory for 1 year to SARS-CoV-2 infection and vaccines already supersedes that of any other acute infectious disease. This knowledge may help inform public policies regarding COVID-19 and COVID-19 vaccines, as well as the scientific development of future vaccines against SARS-CoV-2 and other diseases.
Collapse
Affiliation(s)
- Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
155
|
Protective Immunity of COVID-19 Vaccination with ChAdOx1 nCoV-19 Following Previous SARS-CoV-2 Infection: A Humoral and Cellular Investigation. Viruses 2022; 14:v14091916. [PMID: 36146723 PMCID: PMC9504152 DOI: 10.3390/v14091916] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Infections caused by SARS-CoV-2 induce a severe acute respiratory syndrome called COVID-19 and have led to more than six million deaths worldwide. Vaccination is the most effective preventative measure, and cellular and humoral immunity is crucial to developing individual protection. Here, we aim to investigate hybrid immunity against SARS-CoV-2 triggered by the ChAadOx1 nCoV-19 vaccine in a Brazilian cohort. We investigated the immune response from ChAadOx1 nCoV-19 vaccination in naïve (noCOVID-19) and previously infected individuals (COVID-19) by analyzing levels of D-dimers, total IgG, neutralizing antibodies (Nabs), IFN-γ (interferon-γ) secretion, and immunophenotyping of memory lymphocytes. No significant differences in D-dimer levels were observed 7 or 15 days after vaccination (DAV). All vaccinated individuals presented higher levels of total IgG or Nabs with a positive correlation (R = 0.88). Individuals in the COVID-19 group showed higher levels of antibody and memory B cells, with a faster antibody response starting at 7 DAV compared to noCOVID-19 at 15 DAV. Further, ChAadOx1 nCoV-19 vaccination led to enhanced IFN-γ production (15 DAV) and an increase in activated T CD4+ naïve cells in noCOVID-19 individuals in contrast with COVID-19 individuals. Hence, our data support that hybrid immunity triggered by ChAadOx1 nCoV-19 vaccination is associated with enhanced humoral response, together with a balanced cellular response.
Collapse
|
156
|
de Michelena P, Olea B, Torres I, González‐Candelas F, Navarro D. SARS-CoV-2 RNA load in nasopharyngeal specimens from outpatients with breakthrough COVID-19 due to Omicron BA.1 and BA.2. J Med Virol 2022; 94:5836-5840. [PMID: 35986484 PMCID: PMC9537928 DOI: 10.1002/jmv.28079] [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: 07/20/2022] [Revised: 08/16/2022] [Accepted: 08/17/2022] [Indexed: 01/06/2023]
Abstract
This retrospective observational study compared severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA load in nasopharyngeal specimens (NPs) from patients with breakthrough coronavirus disease 2019 (COVID-19) caused by the Omicron BA.1 or BA.2 sublineages. The convenience sample was composed of 277 outpatients (176 female/112 male; median age, 48 years; range, 12-97) with breakthrough COVID-19 (n = 130 due to BA.1 and n = 147 due to BA.2). All participants had completed a full vaccination schedule and 56% had received a booster vaccine dose at the time of COVID-19 breakthrough microbiological diagnosis. NPs were collected within 7 days (median 2 days) after symptom onset. The TaqPath COVID-19 Combo Kit (Thermo Fisher Scientific) was used to estimate viral loads in NPs. Overall, viral RNA loads in NPs were comparable (p = 0.31) for BA.1 (median, 7.1 log10 copies/ml; range, 2.7-10.6) and BA.2 (median, 7.5 log10 copies/ml; range, 2.7-10.6), yet peak viral load appeared to be reached sooner for BA.2 than for BA.1 (Day 1 vs. Days 3-5; p = 0.002). Time elapsed since last vaccine dose had no significant impact on SARS-CoV-2 RNA loads in the upper respiratory tract (URT) for either BA.1 or BA.2. The data presented do not support that the transmissibility advantage of BA.2 over BA.1 is related to generation of higher viral loads in the URT early after infection.
Collapse
Affiliation(s)
- Paula de Michelena
- Microbiology Service, Clinic University HospitalINCLIVA Health Research InstituteValenciaSpain
| | - Beatriz Olea
- Microbiology Service, Clinic University HospitalINCLIVA Health Research InstituteValenciaSpain
| | - Ignacio Torres
- Microbiology Service, Clinic University HospitalINCLIVA Health Research InstituteValenciaSpain
| | - Fernando González‐Candelas
- Joint Research Unit Infection and Public Health FISABIO‐University of ValenciaInstitute for Integrative Systems Biology (I2SysBio, UV‐CSIC) and CIBER in Epidemiology and Public HealthValenciaSpain
| | - David Navarro
- Microbiology Service, Clinic University HospitalINCLIVA Health Research InstituteValenciaSpain,Department of Microbiology, School of MedicineUniversity of ValenciaValenciaSpain
| |
Collapse
|
157
|
Tang L, Cherry S, Tuomanen EI, Kirkpatrick Roubidoux E, Lin CY, Allison KJ, Gowen A, Freiden P, Allen EK, Su Y, Gaur AH, Estepp JH, McGargill MA, Krammer F, Thomas PG, Schultz-Cherry S, Wolf J. Host Predictors of Broadly Cross-Reactive Antibodies Against Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Variants of Concern Differ Between Infection and Vaccination. Clin Infect Dis 2022; 75:e705-e714. [PMID: 34891165 PMCID: PMC8689782 DOI: 10.1093/cid/ciab996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination there is significant variability between individuals in protective antibody levels against SARS-CoV-2, and within individuals against different virus variants. However, host demographic or clinical characteristics that predict variability in cross-reactive antibody levels are not well-described. These data could inform clinicians, researchers, and policymakers on the populations most likely to require vaccine booster shots. METHODS In an institutional review board-approved prospective observational cohort study of staff at St. Jude Children's Research Hospital, we identified participants with plasma samples collected after SARS-CoV-2 infection, after mRNA vaccination, and after vaccination following infection, and quantitated immunoglobulin G (IgG) levels by enzyme-linked immunosorbent assay to the spike receptor binding domain (RBD) from 5 important SARS-CoV-2 variants (Wuhan Hu-1, B.1.1.7, B.1.351, P.1, and B.1.617.2). We used regression models to identify factors that contributed to cross-reactive IgG against 1 or multiple viral variants. RESULTS Following infection, a minority of the cohort generated cross-reactive antibodies, IgG antibodies that bound all tested variants. Those who did had increased disease severity, poor metabolic health, and were of a particular ancestry. Vaccination increased the levels of cross-reactive IgG levels in all populations, including immunocompromised, elderly, and persons with poor metabolic health. Younger people with a healthy weight mounted the highest responses. CONCLUSIONS Our findings provide important new information on individual antibody responses to infection/vaccination that could inform clinicians on populations that may require follow-on immunization.
Collapse
Affiliation(s)
- Li Tang
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Sean Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Elaine I Tuomanen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | - Chun Yang Lin
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Kim J Allison
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Ashleigh Gowen
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Pamela Freiden
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - E Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Yin Su
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Aditya H Gaur
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Jeremie H Estepp
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
- Department of Global Pediatric Medicine, St. Jude Children’s Research Hospital, Memphis, Tennessee, USAand
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Paul G Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Stacey Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Joshua Wolf
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| |
Collapse
|
158
|
Pannus P, Neven KY, De Craeye S, Heyndrickx L, Vande Kerckhove S, Georges D, Michiels J, Francotte A, Van Den Bulcke M, Zrein M, Van Gucht S, Schmickler MN, Verbrugghe M, Matagne A, Thomas I, Dierick K, Weiner JA, Ackerman ME, Goriely S, Goossens ME, Ariën KK, Desombere I, Marchant A. Poor Antibody Response to BioNTech/Pfizer Coronavirus Disease 2019 Vaccination in Severe Acute Respiratory Syndrome Coronavirus 2-Naive Residents of Nursing Homes. Clin Infect Dis 2022; 75:e695-e704. [PMID: 34864935 PMCID: PMC8690239 DOI: 10.1093/cid/ciab998] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Indexed: 12/02/2022] Open
Abstract
BACKGROUND Residents of nursing homes (NHs) are at high risk of coronavirus disease 2019 (COVID-19)-related disease and death and may respond poorly to vaccination because of old age and frequent comorbid conditions. METHODS Seventy-eight residents and 106 staff members, naive to infection or previously infected with severe acute respiratory syndrome coronavirus (SARS-CoV-2), were recruited in NHs in Belgium before immunization with 2 doses of 30 µg BNT162b2 messenger RNA (mRNA) vaccine at days 0 and 21. Binding antibodies (Abs) to SARS-CoV-2 receptor-binding domain (RBD), spike domains S1 and S2, RBD Ab avidity, and neutralizing Abs against SARS-CoV-2 wild type and B.1.351 were assessed at days 0, 21, 28, and 49. RESULTS SARS-CoV-2-naive residents had lower Ab responses to BNT162b2 mRNA vaccination than naive staff. These poor responses involved lower levels of immunoglobulin (Ig) G to all spike domains, lower avidity of RBD IgG, and lower levels of Abs neutralizing the vaccine strain. No naive residents had detectable neutralizing Abs to the B.1.351 variant. In contrast, SARS-CoV-2-infected residents had high responses to mRNA vaccination, with Ab levels comparable to those in infected staff. Cluster analysis revealed that poor vaccine responders included not only naive residents but also naive staff, emphasizing the heterogeneity of responses to mRNA vaccination in the general population. CONCLUSIONS The poor Ab responses to mRNA vaccination observed in infection-naive NH residents and in some naive staff members suggest suboptimal protection against breakthrough infection, especially with variants of concern. These data support the administration of a third dose of mRNA vaccine to further improve protection of NH residents against COVID-19.
Collapse
Affiliation(s)
- Pieter Pannus
- SD Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - Kristof Y Neven
- SD Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | | | - Leo Heyndrickx
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | | - Daphnée Georges
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université libre de Bruxelles (ULB), Gosselies, Belgium
- Laboratory of Enzymology and Protein Folding, Centre for Protein Engineering, InBioS, University of Liège, Liège, Belgium
| | - Johan Michiels
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | | | | | | | | | | | | - André Matagne
- Laboratory of Enzymology and Protein Folding, Centre for Protein Engineering, InBioS, University of Liège, Liège, Belgium
| | - Isabelle Thomas
- SD Infectious Diseases in Humans, Sciensano, Brussels, Belgium
| | | | - Joshua A Weiner
- Thayer School of Engineering, Dartmouth College, Hanover, NH 03755, USA
| | | | - Stanislas Goriely
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| | | | - Kevin K Ariën
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Arnaud Marchant
- Institute for Medical Immunology and ULB Center for Research in Immunology (U-CRI), Université libre de Bruxelles (ULB), Gosselies, Belgium
| |
Collapse
|
159
|
Walker MR, Podlekareva D, Johnsen S, Leerhøy B, Fougeroux C, Søgaard M, Salanti A, Ditlev SB, Barfod L. SARS-CoV-2 RBD-Specific Antibodies Induced Early in the Pandemic by Natural Infection and Vaccination Display Cross-Variant Binding and Inhibition. Viruses 2022; 14:1861. [PMID: 36146667 PMCID: PMC9503696 DOI: 10.3390/v14091861] [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: 07/13/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/19/2022] Open
Abstract
The development of vaccine candidates for COVID-19 has been rapid, and those that are currently approved display high efficacy against the original circulating strains. However, recently, new variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have emerged with increased transmission rates and less susceptibility to vaccine induced immunity. A greater understanding of protection mechanisms, including antibody longevity and cross-reactivity towards the variants of concern (VoCs), is needed. In this study, samples collected in Denmark early in the pandemic from paucisymptomatic subjects (n = 165) and symptomatic subjects (n = 57) infected with SARS-CoV-2 were used to assess IgG binding and inhibition in the form of angiotensin-converting enzyme 2 receptor (ACE2) competition against the wild-type and four SARS-CoV-2 VoCs (Alpha, Beta, Gamma, and Omicron). Antibodies induced early in the pandemic via natural infection were cross-reactive and inhibited ACE2 binding of the VoC, with reduced inhibition observed for the Omicron variant. When examined longitudinally, sustained cross-reactive inhibitory responses were found to exist in naturally infected paucisymptomatic subjects. After vaccination, receptor binding domain (RBD)-specific IgG binding increased by at least 3.5-fold and inhibition of ACE2 increased by at least 2-fold. When vaccination regimens were compared (two doses of Pfizer-BioNTech BNT162b2 (n = 50), or one dose of Oxford-AstraZeneca ChAdOx1 nCoV-19 followed by Pfizer-BioNTech BNT162b2 (ChAd/BNT) (n = 15)), higher levels of IgG binding and inhibition were associated with mix and match (ChAd/BNT) prime-boosting and time since vaccination. These results are particularly relevant for countries where vaccination levels are low.
Collapse
Affiliation(s)
- Melanie R. Walker
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Daria Podlekareva
- Centre for Translational Research, Bispebjerg Hospital, 2400 Copenhagen, Denmark
| | - Stine Johnsen
- Centre for Translational Research, Bispebjerg Hospital, 2400 Copenhagen, Denmark
| | - Bonna Leerhøy
- Centre for Translational Research, Bispebjerg Hospital, 2400 Copenhagen, Denmark
| | - Cyrielle Fougeroux
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Max Søgaard
- Expres2ion Biotechnologies, 2970 Hørsholm, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sisse Bolm Ditlev
- Centre for Translational Research, Bispebjerg Hospital, 2400 Copenhagen, Denmark
| | - Lea Barfod
- Centre for Medical Parasitology, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
160
|
Abstract
Despite effective spike-based vaccines and monoclonal antibodies, the SARS-CoV-2 pandemic continues more than two and a half years post-onset. Relentless investigation has outlined a causative dynamic between host-derived antibodies and reciprocal viral subversion. Integration of this paradigm into the architecture of next generation antiviral strategies, predicated on a foundational understanding of the virology and immunology of SARS-CoV-2, will be critical for success. This review aims to serve as a primer on the immunity endowed by antibodies targeting SARS-CoV-2 spike protein through a structural perspective. We begin by introducing the structure and function of spike, polyclonal immunity to SARS-CoV-2 spike, and the emergence of major SARS-CoV-2 variants that evade immunity. The remainder of the article comprises an in-depth dissection of all major epitopes on SARS-CoV-2 spike in molecular detail, with emphasis on the origins, neutralizing potency, mechanisms of action, cross-reactivity, and variant resistance of representative monoclonal antibodies to each epitope.
Collapse
Affiliation(s)
- John M Errico
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - Lucas J Adams
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - Daved H Fremont
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, United States; Department of Molecular Microbiology, Washington University School of Medicine, St Louis, MO, United States; Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St Louis, MO, United States.
| |
Collapse
|
161
|
Chang CC, Vlad G, Vasilescu ER, Li P, Husain SA, Silvia EA, Cohen DJ, Ratner LE, Sun WZ, Mohan S, Suciu-Foca N. Previous SARS-CoV-2 infection or a third dose of vaccine elicited cross-variant neutralising antibodies in vaccinated solid-organ transplant recipients. Clin Transl Immunology 2022; 11:e1411. [PMID: 35979345 PMCID: PMC9371857 DOI: 10.1002/cti2.1411] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/26/2022] [Accepted: 07/27/2022] [Indexed: 12/24/2022] Open
Abstract
Objectives The SARS‐CoV‐2 pandemic poses a great threat to global health, particularly in solid organ transplant recipients (SOTRs). A 3‐dose mRNA vaccination protocol has been implemented for the majority of SOTRs, yet their immune responses are less effective compared to healthy controls (HCs). Methods We analyzed the humoral immune responses against the vaccine strain and variants of concern (VOC), including the highly mutated‐omicron variant in 113 SOTRs, of whom 44 had recovered from COVID‐19 (recovered‐SOTRs) and 69 had not contracted the virus (COVID‐naïve). In addition, 30 HCs, 8 of whom had recovered from COVID‐19, were also studied. Results Here, we report that three doses of the mRNA vaccine had only a modest effect in eliciting anti‐viral antibodies against all viral strains in the fully vaccinated COVID‐naive SOTRs (n = 47). Only 34.0% of this group of patients demonstrated both detectable anti‐RBD IgG with neutralization activities against alpha, beta, and delta variants, and only 8.5% of them showed additional omicron neutralizing capacities. In contrast, 79.5% of the recovered‐SOTRs who received two doses of vaccine demonstrated both higher anti‐RBD IgG levels and neutralizing activities against all VOC, including omicron. Conclusion These findings illustrate a significant impact of previous infection on the development of anti‐SARS‐CoV‐2 immune responses in vaccinated SOTRs and highlight the need for alternative strategies to protect a subset of a lesser‐vaccine responsive population.
Collapse
Affiliation(s)
- Chih-Chao Chang
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - George Vlad
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Elena Rodica Vasilescu
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Ping Li
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - Syed A Husain
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Elaine A Silvia
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| | - David J Cohen
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Lloyd E Ratner
- Department of Surgery Columbia University Irving Medical Center New York NY USA
| | - Wei-Zen Sun
- Department of Anesthesiology National Taiwan University Hospital Taipei Taiwan
| | - Sumit Mohan
- Division of Nephrology, Department of Medicine Columbia University Irving Medical Center New York NY USA.,The Columbia University Renal Epidemiology (CURE) Group New York NY USA
| | - Nicole Suciu-Foca
- Department of Pathology and Cell Biology Columbia University Irving Medical Center New York NY USA
| |
Collapse
|
162
|
Bowen JE, Addetia A, Dang HV, Stewart C, Brown JT, Sharkey WK, Sprouse KR, Walls AC, Mazzitelli IG, Logue JK, Franko NM, Czudnochowski N, Powell AE, Dellota E, Ahmed K, Ansari AS, Cameroni E, Gori A, Bandera A, Posavad CM, Dan JM, Zhang Z, Weiskopf D, Sette A, Crotty S, Iqbal NT, Corti D, Geffner J, Snell G, Grifantini R, Chu HY, Veesler D. Omicron spike function and neutralizing activity elicited by a comprehensive panel of vaccines. Science 2022; 377:890-894. [PMID: 35857529 PMCID: PMC9348749 DOI: 10.1126/science.abq0203] [Citation(s) in RCA: 123] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 07/12/2022] [Indexed: 12/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant of concern comprises several sublineages, with BA.2 and BA.2.12.1 having replaced the previously dominant BA.1 and with BA.4 and BA.5 increasing in prevalence worldwide. We show that the large number of Omicron sublineage spike mutations leads to enhanced angiotensin-converting enzyme 2 (ACE2) binding, reduced fusogenicity, and severe dampening of plasma neutralizing activity elicited by infection or seven clinical vaccines relative to the ancestral virus. Administration of a homologous or heterologous booster based on the Wuhan-Hu-1 spike sequence markedly increased neutralizing antibody titers and breadth against BA.1, BA.2, BA.2.12.1, BA.4, and BA.5 across all vaccines evaluated. Our data suggest that although Omicron sublineages evade polyclonal neutralizing antibody responses elicited by primary vaccine series, vaccine boosters may provide sufficient protection against Omicron-induced severe disease.
Collapse
Affiliation(s)
- John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Amin Addetia
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Ha V. Dang
- Vir Biotechnology, San Francisco, CA 94158, USA
| | - Cameron Stewart
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Jack T. Brown
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - William K. Sharkey
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Kaitlin R. Sprouse
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| | - Ignacio G. Mazzitelli
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | - Jennifer K. Logue
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - Nicholas M. Franko
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | | | | | | | - Kumail Ahmed
- Departments of Paediatrics and Child Health and Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Asefa Shariq Ansari
- Departments of Paediatrics and Child Health and Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Elisabetta Cameroni
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Andrea Gori
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milan, Milan, Italy
| | - Christine M. Posavad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Jennifer M. Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Najeeha Talat Iqbal
- Departments of Paediatrics and Child Health and Biological and Biomedical Sciences, Aga Khan University, Karachi 74800, Pakistan
| | - Davide Corti
- Humabs Biomed SA, a subsidiary of Vir Biotechnology, 6500 Bellinzona, Switzerland
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Facultad de Medicina, Buenos Aires C1121ABG, Argentina
| | | | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare “Romeo ed Enrica Invernizzi,” Milan, Italy
| | - Helen Y. Chu
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA 98195, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
163
|
Russell MW, Mestecky J. Mucosal immunity: The missing link in comprehending SARS-CoV-2 infection and transmission. Front Immunol 2022; 13:957107. [PMID: 36059541 PMCID: PMC9428579 DOI: 10.3389/fimmu.2022.957107] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/27/2022] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV-2 is primarily an airborne infection of the upper respiratory tract, which on reaching the lungs causes the severe acute respiratory disease, COVID-19. Its first contact with the immune system, likely through the nasal passages and Waldeyer's ring of tonsils and adenoids, induces mucosal immune responses revealed by the production of secretory IgA (SIgA) antibodies in saliva, nasal fluid, tears, and other secretions within 4 days of infection. Evidence is accumulating that these responses might limit the virus to the upper respiratory tract resulting in asymptomatic infection or only mild disease. The injectable systemic vaccines that have been successfully developed to prevent serious disease and its consequences do not induce antibodies in mucosal secretions of naïve subjects, but they may recall SIgA antibody responses in secretions of previously infected subjects, thereby helping to explain enhanced resistance to repeated (breakthrough) infection. While many intranasally administered COVID vaccines have been found to induce potentially protective immune responses in experimental animals such as mice, few have demonstrated similar success in humans. Intranasal vaccines should have advantage over injectable vaccines in inducing SIgA antibodies in upper respiratory and oral secretions that would not only prevent initial acquisition of the virus, but also suppress community spread via aerosols and droplets generated from these secretions.
Collapse
Affiliation(s)
- Michael W. Russell
- Department of Microbiology and Immunology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Jiri Mestecky
- Department of Microbiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
164
|
Idoko OT, Usuf E, Okomo U, Wonodi C, Jambo K, Kampmann B, Madhi S, Adetifa I. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) in Africa: Current Considerations and Future Projections. Clin Infect Dis 2022; 75:S136-S140. [PMID: 35749696 PMCID: PMC9376270 DOI: 10.1093/cid/ciac401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 01/19/2023] Open
Abstract
The burden of severe Covid-19 has been relatively low in sib-Saharan Africa compared to Europe and the Americas. However, SARS-CoV-2 sero-prevalence data has demonstrated that there has been more widespread transmission than can be deduced from reported cases. This could be attributed to under reporting due to low testing capacity or high numbers of asymptomatic SARS-CoV-2 infection in communities. Recent data indicates that prior SARS-CoV-2 exposure is protective against reinfection and that vaccination of previously SARS-CoV-2 infected individuals induces robust cross-reactive antibody responses. Considering these data, calls for a need for a re-think of the COVID-19 vaccination strategy in sub-Saharan African settings with high SARSCoV-2 population exposure but limited available vaccine doses. A potential recommendation would be to prioritize rapid and widespread vaccination of the first dose, while waiting for more vaccines to become available.
Collapse
Affiliation(s)
- Olubukola T Idoko
- Faculty of Infectious and Tropical disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Effua Usuf
- Faculty of Infectious and Tropical disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Uduak Okomo
- Faculty of Infectious and Tropical disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Chizoba Wonodi
- International Health, Health Systems Center, John Hopkins University, Baltimore, USA
| | - Kondwani Jambo
- Viral Immunology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre, Malawi
| | - Beate Kampmann
- Faculty of Infectious and Tropical disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Shabir Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Ifedayo Adetifa
- Faculty of Infectious and Tropical disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
165
|
Abella E, Trigueros M, Pradenas E, Muñoz-Lopez F, Garcia-Pallarols F, Ben Azaiz Ben Lahsen R, Trinité B, Urrea V, Marfil S, Rovirosa C, Puig T, Grau E, Chamorro A, Toledo R, Font M, Palacín D, Lopez-Segui F, Carrillo J, Prat N, Mateu L, Clotet B, Blanco J, Massanella M. Efficacy of SARS-CoV-2 vaccination in patients with monoclonal gammopathies: A cross sectional study. Life Sci Alliance 2022; 5:e202201479. [PMID: 35961779 PMCID: PMC9375155 DOI: 10.26508/lsa.202201479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/20/2022] [Accepted: 07/20/2022] [Indexed: 11/24/2022] Open
Abstract
SARS-CoV-2 vaccination is the most effective strategy to protect individuals with haematologic malignancies against severe COVID-19, while eliciting limited vaccine responses. We characterized the humoral responses following 3 mo after mRNA-based vaccines in individuals at different plasma-cell disease stages: monoclonal gammopathy of undetermined significance (MGUS), smoldering multiple myeloma (SMM), and multiple myeloma on first-line therapy (MM), compared with a healthy population. Plasma samples from uninfected MM patients showed lower SARS-CoV-2-specific antibody levels and neutralization capacity compared with MGUS, SMM, and healthy individuals. Importantly, COVID-19 recovered MM individuals presented significantly higher plasma neutralization capacity compared with their uninfected counterparts, highlighting that hybrid immunity elicit stronger immunity even in this immunocompromised population. No differences in the vaccine-induced humoral responses were observed between uninfected MGUS, SMM and healthy individuals. In conclusion, MGUS and SMM patients could be SARS-CoV-2 vaccinated following the vaccine recommendations for the general population, whereas a tailored monitoring of the vaccine-induced immune responses should be considered in uninfected MM patients.
Collapse
Affiliation(s)
- Eugenia Abella
- Department of Hematology, Hospital del Mar-IMIM, Barcelona, Spain
- Pompeu Fabra University, Barcelona, Spain
| | - Macedonia Trigueros
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Edwards Pradenas
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Francisco Muñoz-Lopez
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | | | | | - Benjamin Trinité
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Victor Urrea
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Silvia Marfil
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Carla Rovirosa
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Teresa Puig
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Eulàlia Grau
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Anna Chamorro
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Ruth Toledo
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Marta Font
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Dolors Palacín
- Direcció d'Atenció Primària-Metropolitana Nord, Sabadell, Spain
| | - Francesc Lopez-Segui
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
| | - Jorge Carrillo
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, Madrid, Spain
| | - Nuria Prat
- Direcció d'Atenció Primària-Metropolitana Nord, Sabadell, Spain
| | - Lourdes Mateu
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, CIBERES, Madrid, Spain
| | - Bonaventura Clotet
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- Fundació Lluita Contra les Infeccions, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
| | - Julià Blanco
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, Madrid, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
- Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, Badalona Barcelona, Spain
| | - Marta Massanella
- IrsiCaixa-AIDS Research Institute, Hospital Universitari Germans Trias i Pujol, Campus Can Ruti, Badalona, Spain
- University of Vic-Central University of Catalonia (UVic-UCC), 08500, Vic, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, CIBERINFEC, Madrid, Spain
| |
Collapse
|
166
|
He WT, Yuan M, Callaghan S, Musharrafieh R, Song G, Silva M, Beutler N, Lee WH, Yong P, Torres JL, Melo M, Zhou P, Zhao F, Zhu X, Peng L, Huang D, Anzanello F, Ricketts J, Parren M, Garcia E, Ferguson M, Rinaldi W, Rawlings SA, Nemazee D, Smith DM, Briney B, Safonova Y, Rogers TF, Dan JM, Zhang Z, Weiskopf D, Sette A, Crotty S, Irvine DJ, Ward AB, Wilson IA, Burton DR, Andrabi R. Broadly neutralizing antibodies to SARS-related viruses can be readily induced in rhesus macaques. Sci Transl Med 2022; 14:eabl9605. [PMID: 35947674 PMCID: PMC10069796 DOI: 10.1126/scitranslmed.abl9605] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
To prepare for future coronavirus (CoV) pandemics, it is desirable to generate vaccines capable of eliciting broadly neutralizing antibody responses to CoVs. Here, we show that immunization of macaques with SARS-CoV-2 spike (S) protein with a two-shot protocol generated potent serum receptor binding domain cross-neutralizing antibody responses to both SARS-CoV-2 and SARS-CoV-1. Furthermore, responses were equally effective against most SARS-CoV-2 variants of concern (VOCs) and some were highly effective against Omicron. This result contrasts with human infection or many two-shot vaccination protocols where responses were typically more SARS-CoV-2 specific and where VOCs were less well neutralized. Structural studies showed that cloned macaque neutralizing antibodies, particularly using a given heavy chain germline gene, recognized a relatively conserved region proximal to the angiotensin converting enzyme 2 receptor binding site (RBS), whereas many frequently elicited human neutralizing antibodies targeted more variable epitopes overlapping the RBS. B cell repertoire differences between humans and macaques appeared to influence the vaccine response. The macaque neutralizing antibodies identified a pan-SARS-related virus epitope region less well targeted by human antibodies that could be exploited in rational vaccine design.
Collapse
Affiliation(s)
- Wan-ting He
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sean Callaghan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Rami Musharrafieh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Murillo Silva
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Wen-Hsin Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Peter Yong
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mariane Melo
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Panpan Zhou
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fangzhu Zhao
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Xueyong Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Linghang Peng
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Deli Huang
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Fabio Anzanello
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - James Ricketts
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Mara Parren
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | - Stephen A. Rawlings
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - David Nemazee
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Davey M. Smith
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Bryan Briney
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Yana Safonova
- Computer Science and Engineering Department, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Thomas F. Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Jennifer M. Dan
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Alessandro Sette
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Shane Crotty
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Darrell J. Irvine
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Andrew B. Ward
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Ian A. Wilson
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Dennis R. Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Cambridge, MA 02139, USA
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA 92037, USA
- IAVI Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA 92037, USA
- Consortium for HIV/AIDS Vaccine Development (CHAVD), The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
167
|
Kim Y, Bae JY, Kwon K, Chang HH, Lee WK, Park H, Kim J, Choi I, Park MS, Kim SW. Kinetics of neutralizing antibodies against SARS-CoV-2 infection according to sex, age, and disease severity. Sci Rep 2022; 12:13491. [PMID: 35931794 PMCID: PMC9356129 DOI: 10.1038/s41598-022-17605-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 07/28/2022] [Indexed: 11/09/2022] Open
Abstract
Knowledge of the factors affecting the difference in kinetics and longevity of the neutralizing antibody (nAb) response to SARS-CoV-2 is necessary to properly prioritize vaccination. In the present study, from March to December 2020, of the 143 patients who recovered from COVID-19, 87 underwent study visits scheduled every 3 months. Patient demographics and blood samples were collected followed by a plaque reduction neutralization test to analyze nAb titers. A linear mixed model was used to compare the effects of sex, age, and disease severity over time. Results demonstrated a gradual reduction in nAb titers over time with a significant decrease from 6 to 9 months post-COVID-19 infection (p < 0.001). In time-to-sex, age, and disease severity comparisons, reduction in nAb titers over time was unaffected by sex (p = 0.167), age (p = 0.188), or disease severity (p = 0.081). Additionally, the nAb titer was 1.46 times significantly higher in those aged ≥ 50 years than in those aged < 50 years (p = 0.036) irrespective of time Moreover, the nAb titer was 2.41 times higher in the moderate or above than that in the below moderate disease severity group (p < 0.001). However, no significant differences were observed in terms of sex (p = 0.300). Given the reduction in nAbs over time, maintaining protective neutralizing antibodies regardless of sex, age, or disease severity is needed.
Collapse
Affiliation(s)
- Yoonjung Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Joon-Yong Bae
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Kitae Kwon
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Hyun-Ha Chang
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Won Kee Lee
- Department of Medical Informatics, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Heedo Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Jeonghun Kim
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Isaac Choi
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Man-Seong Park
- Department of Microbiology, Institute for Viral Diseases, Biosafety center, College of Medicine, Korea University, Seoul, Republic of Korea.
| | - Shin-Woo Kim
- Department of Internal Medicine, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, 130, Dongdeok-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
168
|
Price R, Cho J, Nelson S. SARS-CoV-2 Seroprevalence at an Urban Hospital in Haiti. Cureus 2022; 14:e27690. [PMID: 36072217 PMCID: PMC9440879 DOI: 10.7759/cureus.27690] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 11/24/2022] Open
Abstract
Background: Much to the surprise of the global community, Haiti has had far fewer COVID-19 cases and deaths than initially expected. In this study, we sought to estimate the seroprevalence of COVID-19 in a convenience cohort based in Port-au-Prince, Haiti, to elucidate potential reasons for the apparently low burden of COVID-19 in Haiti. Methods: We performed a cross-sectional analysis of SARS-CoV-2 antibody prevalence in patients aged one to 89 years old who were seen at the Haiti Adventist Hospital (HAH) laboratory between December 17, 2020, and July 3, 2021, with an order requiring a blood draw. We excluded patients outside of the age range and those who did not verbally consent to the study. We tested residual patient serum samples using the Biosys PlusTM COVID-19 IgM/IgG Rapid Test. Findings: Of 9,740 patients seen by the HAH laboratory from December 2020 to July 2021, 538 consented to have antibody testing and answer survey questions. 529 were included in the final analysis. We excluded nine participants who were aged greater than 89 (n=3), aged less than one (n=2), or had results that were not properly recorded (n=4). Three of the tested patients were repeat testers, with one who had been tested three times. These repeat results were included for a final seroprevalence analysis of 533 samples. In the final participant pool, 142 (26.6%) of 533 samples tested positive for either IgM, IgG, or both antibodies to the SARS-CoV-2 virus. Adjustment for test sensitivity resulted in an estimated seroprevalence of 28.7% (95% CI 24.9-32.9). We observed significant differences in seroprevalences among age groups, with seroprevalence increasing with age. Interpretation: The SARS-CoV-2 antibody seroprevalence in Haiti appears to be greater than the publicly reported statistics by several orders of magnitude. Furthermore, a reduced fatality rate relative to high-income countries points to uncertain factors that may confer immunologic resistance in the Haitian population.
Collapse
|
169
|
Gammon R, Katz LM, Strauss D, Rowe K, Menitove J, Benjamin RJ, Goel R, Borge D, Reichenberg S, Smith R. Beyond COVID-19 and lessons learned in the United States. Transfus Med 2022; 33:6-15. [PMID: 35918741 PMCID: PMC9539268 DOI: 10.1111/tme.12896] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 06/20/2022] [Accepted: 07/02/2022] [Indexed: 12/16/2022]
Abstract
The COVID-19 pandemic severely tested the resilience of the US blood supply with wild fluctuations in blood donation and utilisation rates as community donation opportunities ebbed and hospitals post-poned elective surgery. Key stakeholders in transfusion services, blood centres, supply chains and manufacturers reviewed their experiences during the SARS-CoV-2 pandemic as well as available literature to describe successes, opportunities for improvement and lessons learned. The blood community found itself in uncharted territory responding to restriction of its access to donors (approximately 20% decrease) and some supplies; environmental adjustments to address staff and donor concerns about coronavirus transmission; and the development of a new product (COVID-19 convalescent plasma [CCP]). In assuring that the needs of the patients were paramount, the donation process was safe, that clinicians had access to CCP, and vendor relationships aligned, the blood banking community relearned its primary focus: improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | - Jay Menitove
- Kansas University Medical CenterKansas CityKansasUSA
| | | | | | - Dayand Borge
- Memorial Sloan Kettering Cancer Center, Center for Laboratory MedicineNew YorkNew York StateUSA
| | | | | |
Collapse
|
170
|
Wilhelm A, Widera M, Grikscheit K, Toptan T, Schenk B, Pallas C, Metzler M, Kohmer N, Hoehl S, Marschalek R, Herrmann E, Helfritz FA, Wolf T, Goetsch U, Ciesek S. Limited neutralisation of the SARS-CoV-2 Omicron subvariants BA.1 and BA.2 by convalescent and vaccine serum and monoclonal antibodies. EBioMedicine 2022; 82:104158. [PMID: 35834885 DOI: 10.1101/2021.12.07.21267432] [Citation(s) in RCA: 160] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 05/21/2023] Open
Abstract
BACKGROUND In recent months, Omicron variants of SARS-CoV-2 have become dominant in many regions of the world, and case numbers with Omicron subvariants BA.1 and BA.2 continue to increase. Due to numerous mutations in the spike protein, the efficacy of currently available vaccines, which are based on Wuhan-Hu 1 isolate of SARS-CoV-2, is reduced, leading to breakthrough infections. Efficacy of monoclonal antibody therapy is also likely impaired. METHODS In our in vitro study using A549-AT cells constitutively expressing ACE2 and TMPRSS2, we determined and compared the neutralizing capacity of vaccine-elicited sera, convalescent sera and monoclonal antibodies against authentic SARS-CoV-2 Omicron BA.1 and BA.2 compared with Delta. FINDINGS Almost no neutralisation of Omicron BA.1 and BA.2 was observed using sera from individuals vaccinated with two doses 6 months earlier, regardless of the type of vaccine taken. Shortly after the booster dose, most sera from triple BNT162b2-vaccinated individuals were able to neutralise both Omicron variants. In line with waning antibody levels three months after the booster, only weak residual neutralisation was observed for BA.1 (26%, n = 34, 0 median NT50) and BA.2 (44%, n = 34, 0 median NT50). In addition, BA.1 but not BA.2 was resistant to the neutralising monoclonal antibodies casirivimab/imdevimab, while BA.2 exhibited almost a complete evasion from the neutralisation induced by sotrovimab. INTERPRETATION Both SARS-CoV-2 Omicron subvariants BA.1 and BA.2 escape antibody-mediated neutralisation elicited by vaccination, previous infection with SARS-CoV-2, and monoclonal antibodies. Waning immunity renders the majority of tested sera obtained three months after booster vaccination negative in BA.1 and BA.2 neutralisation. Omicron subvariant specific resistance to the monoclonal antibodies casirivimab/imdevimab and sotrovimab emphasizes the importance of genotype-surveillance and guided application. FUNDING This study was supported in part by the Goethe-Corona-Fund of the Goethe University Frankfurt (M.W.) and the Federal Ministry of Education and Research (COVIDready; grant 02WRS1621C (M.W.).
Collapse
Affiliation(s)
- Alexander Wilhelm
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany.
| | - Katharina Grikscheit
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Tuna Toptan
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Barbara Schenk
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Christiane Pallas
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Melinda Metzler
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Sebastian Hoehl
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Eva Herrmann
- Institute of Biostatistics and Mathematical Modelling, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Fabian A Helfritz
- Bürgerhospital Frankfurt, Nibelungenallee 37-41, 60318 Frankfurt am Main, Germany
| | - Timo Wolf
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Udo Goetsch
- Health Protection Authority of the City of Frankfurt am Main, 60313 Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany; German Centre for Infection Research (DZIF), partner site Frankfurt, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
171
|
Antibody and T-cellular response to COVID-19 booster vaccine in SARS-CoV-1 survivors. Clin Immunol 2022; 244:109103. [PMID: 36049602 PMCID: PMC9423872 DOI: 10.1016/j.clim.2022.109103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022]
Abstract
The severe acute respiratory syndrome coronavirus 1 (SARS-CoV-1) survivors are more likely to produce a potent immune response to SARS-CoV-2 after booster vaccination. We assessed humoral and T cell responses against SARS-CoV-2 in previously vaccinated SARS-CoV-1 survivors and naïve healthy individuals (NHIs) after a booster Ad5-nCoV dose. Boosted SARS-CoV-1 survivors had a high neutralization of SARS-CoV-2 Wuhan-Hu-1 (WA1), Beta, and Delta but is limited to Omicron subvariants (BA.1, BA.2, BA.2.12.1, and BA.4/BA.5). Most boosted SARS-CoV-1 survivors had robust SARS-CoV-2-specific CD4+ and CD8+ T cell responses. While booster vaccination in NHIs elicited less or ineffective neutralization of WA1, Beta, and Delta, and none of them induced neutralizing antibodies against Omicron subvariants. However, they developed comparable SARS-CoV-2-specific T cell responses compared to boosted SARS-CoV-1 survivors. These findings suggest that boosted Ad5-nCoV would not elicit effective neutralizing antibodies against Omicron subvariants in SARS-CoV-1 survivors and NHIs but induced comparable robust T cell responses. Achieving a high antibody titer in SARS-CoV-1 survivors and NHIs is desirable to generate broad neutralization.
Collapse
|
172
|
Abstract
The SARS-CoV-2 pandemic has demonstrated the importance of studying antiviral immunity within sites of infection to gain insights into mechanisms for immune protection and disease pathology. As SARS-CoV-2 is tropic to the respiratory tract, many studies of airway washes, lymph node aspirates, and postmortem lung tissue have revealed site-specific immune dynamics that are associated with the protection or immunopathology but are not readily observed in circulation. This review summarizes the growing body of work identifying immune processes in tissues and their interplay with immune responses in circulation during acute SARS-CoV-2 infection, severe disease, and memory persistence. Establishment of tissue resident immunity also may have implications for vaccination and the durability of immune memory and protection.
Collapse
Affiliation(s)
- Ksenia Rybkina
- Department of Microbiology and ImmunologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Julia Davis‐Porada
- Department of Microbiology and ImmunologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
| | - Donna L. Farber
- Department of Microbiology and ImmunologyColumbia University Vagelos College of Physicians and SurgeonsNew YorkNew YorkUSA
- Department of SurgeryColumbia University Irving Medical CenterNew YorkNew YorkUSA
| |
Collapse
|
173
|
Wilhelm A, Widera M, Grikscheit K, Toptan T, Schenk B, Pallas C, Metzler M, Kohmer N, Hoehl S, Marschalek R, Herrmann E, Helfritz FA, Wolf T, Goetsch U, Ciesek S. Limited neutralisation of the SARS-CoV-2 Omicron subvariants BA.1 and BA.2 by convalescent and vaccine serum and monoclonal antibodies. EBioMedicine 2022; 82:104158. [PMID: 35834885 PMCID: PMC9271884 DOI: 10.1016/j.ebiom.2022.104158] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In recent months, Omicron variants of SARS-CoV-2 have become dominant in many regions of the world, and case numbers with Omicron subvariants BA.1 and BA.2 continue to increase. Due to numerous mutations in the spike protein, the efficacy of currently available vaccines, which are based on Wuhan-Hu 1 isolate of SARS-CoV-2, is reduced, leading to breakthrough infections. Efficacy of monoclonal antibody therapy is also likely impaired. METHODS In our in vitro study using A549-AT cells constitutively expressing ACE2 and TMPRSS2, we determined and compared the neutralizing capacity of vaccine-elicited sera, convalescent sera and monoclonal antibodies against authentic SARS-CoV-2 Omicron BA.1 and BA.2 compared with Delta. FINDINGS Almost no neutralisation of Omicron BA.1 and BA.2 was observed using sera from individuals vaccinated with two doses 6 months earlier, regardless of the type of vaccine taken. Shortly after the booster dose, most sera from triple BNT162b2-vaccinated individuals were able to neutralise both Omicron variants. In line with waning antibody levels three months after the booster, only weak residual neutralisation was observed for BA.1 (26%, n = 34, 0 median NT50) and BA.2 (44%, n = 34, 0 median NT50). In addition, BA.1 but not BA.2 was resistant to the neutralising monoclonal antibodies casirivimab/imdevimab, while BA.2 exhibited almost a complete evasion from the neutralisation induced by sotrovimab. INTERPRETATION Both SARS-CoV-2 Omicron subvariants BA.1 and BA.2 escape antibody-mediated neutralisation elicited by vaccination, previous infection with SARS-CoV-2, and monoclonal antibodies. Waning immunity renders the majority of tested sera obtained three months after booster vaccination negative in BA.1 and BA.2 neutralisation. Omicron subvariant specific resistance to the monoclonal antibodies casirivimab/imdevimab and sotrovimab emphasizes the importance of genotype-surveillance and guided application. FUNDING This study was supported in part by the Goethe-Corona-Fund of the Goethe University Frankfurt (M.W.) and the Federal Ministry of Education and Research (COVIDready; grant 02WRS1621C (M.W.).
Collapse
Affiliation(s)
- Alexander Wilhelm
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Marek Widera
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany.
| | - Katharina Grikscheit
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Tuna Toptan
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Barbara Schenk
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Christiane Pallas
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Melinda Metzler
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Niko Kohmer
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Sebastian Hoehl
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Eva Herrmann
- Institute of Biostatistics and Mathematical Modelling, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Fabian A Helfritz
- Bürgerhospital Frankfurt, Nibelungenallee 37-41, 60318 Frankfurt am Main, Germany
| | - Timo Wolf
- Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany
| | - Udo Goetsch
- Health Protection Authority of the City of Frankfurt am Main, 60313 Frankfurt am Main, Germany
| | - Sandra Ciesek
- Institute for Medical Virology, University Hospital Frankfurt, Goethe University Frankfurt, 60596 Frankfurt am Main, Germany; German Centre for Infection Research (DZIF), partner site Frankfurt, Germany; Fraunhofer Institute for Molecular Biology and Applied Ecology, Branch Translational Medicine and Pharmacology, 60596 Frankfurt am Main, Germany.
| |
Collapse
|
174
|
Nguyen DC, Lamothe PA, Woodruff MC, Saini AS, Faliti CE, Sanz I, Lee FE. COVID-19 and plasma cells: Is there long-lived protection? Immunol Rev 2022; 309:40-63. [PMID: 35801537 PMCID: PMC9350162 DOI: 10.1111/imr.13115] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ankur S. Saini
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Caterina E. Faliti
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ignacio Sanz
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Frances Eun‐Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|
175
|
Singh VK, Chaurasia H, Mishra R, Srivastava R, Yadav AK, Dwivedi J, Singh P, Singh RK. COVID-19: Pathophysiology, transmission, and drug development for therapeutic treatment and vaccination strategies. Curr Pharm Des 2022; 28:2211-2233. [PMID: 35909276 DOI: 10.2174/1381612828666220729093340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/04/2022] [Indexed: 11/22/2022]
Abstract
COVID-19, a dreaded and highly contagious pandemic, is flagrantly known for its rapid prevalence across the world. Till date, none of the treatments are distinctly accessible for this life-threatening disease. Under the prevailing conditions of medical emergency, one creative strategy for the identification of novel and potential antiviral agents gaining momentum in research institutions and progressively being leveraged by pharmaceutical companies is target-based drug repositioning/repurposing. A continuous monitoring and recording of results offer an anticipation that this strategy may help to reveal new medications for viral infections. This review recapitulates the neoteric illation of COVID-19, its genomic dispensation, molecular evolution via phylogenetic assessment, drug targets, the most frequently worldwide used repurposed drugs and their therapeutic applications, and a recent update on vaccine management strategies. The available data from solidarity trials exposed that the treatment with several known drugs, viz. lopinavir-ritonavir, chloroquine, hydroxychloroquine, etc had displayed various antagonistic effects along with no impactful result in diminution of mortality rate. The drugs like remdesivir, favipiravir, and ribavirin proved to be quite safer therapeutic options for treatment against COVID-19. Similarly, dexamethasone, convalescent plasma therapy and oral administration of 2DG are expected to reduce the mortality rate of COVID-19 patients.
Collapse
Affiliation(s)
- Vishal Kumar Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Himani Chaurasia
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Richa Mishra
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Ritika Srivastava
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Aditya K Yadav
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Jayati Dwivedi
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Prashant Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| | - Ramendra K Singh
- Bioorganic Research Laboratory, Department of Chemistry, University of Allahabad, Prayagraj- 211002, India
| |
Collapse
|
176
|
Li T, Luo KQ. Recipients of COVID-19 vaccines face challenges of SARS-CoV-2 variants. Int J Biol Sci 2022; 18:4642-4647. [PMID: 35874950 PMCID: PMC9305267 DOI: 10.7150/ijbs.72424] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/02/2022] [Indexed: 01/15/2023] Open
Abstract
The coronavirus disease 19 (COVID-19) has been rampant since 2019, severely affecting global public health, and causing 5.75 million deaths worldwide. So far, many vaccines have been developed to prevent the infection of SARS-CoV-2 virus. However, the emergence of new variants may threat vaccine recipients as they might evade immunological surveillance that depends on the using of anti-SARS-CoV-2 antibody to neutralize the viral particles. Recent studies have found that recipients who received two doses of vaccination plus an additional booster shoot were able to quickly elevate neutralization response and immune response against wild-type SARS-CoV-2 virus and some initially appeared viral variants. In this review, we assessed the real-world effectiveness of different COVID-19 vaccines by population studies and neutralization assays and compared neutralization responses of booster vaccines in vitro. Finally, as the efficacy of COVID-19 vaccine is expected to decline over time, continued vaccination should be considered to achieve a long-term immune protection against coronavirus.
Collapse
Affiliation(s)
- Tianhong Li
- Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Kathy Qian Luo
- Faculty of Health Sciences, University of Macau, Macao SAR, China.,Ministry of Education-Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macao SAR, China
| |
Collapse
|
177
|
Dhanya CR, Shailaja A, Mary AS, Kandiyil SP, Savithri A, Lathakumari VS, Veettil JT, Vandanamthadathil JJ, Madhavan M. RNA Viruses, Pregnancy and Vaccination: Emerging Lessons from COVID-19 and Ebola Virus Disease. Pathogens 2022; 11:800. [PMID: 35890044 PMCID: PMC9322689 DOI: 10.3390/pathogens11070800] [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: 05/26/2022] [Revised: 07/10/2022] [Accepted: 07/11/2022] [Indexed: 02/01/2023] Open
Abstract
Pathogenic viruses with an RNA genome represent a challenge for global human health since they have the tremendous potential to develop into devastating pandemics/epidemics. The management of the recent COVID-19 pandemic was possible to a certain extent only because of the strong foundations laid by the research on previous viral outbreaks, especially Ebola Virus Disease (EVD). A clear understanding of the mechanisms of the host immune response generated upon viral infections is a prime requisite for the development of new therapeutic strategies. Hence, we present here a comparative study of alterations in immune response upon SARS-CoV-2 and Ebola virus infections that illustrate many common features. Vaccination and pregnancy are two important aspects that need to be studied from an immunological perspective. So, we summarize the outcomes and immune responses in vaccinated and pregnant individuals in the context of COVID-19 and EVD. Considering the significance of immunomodulatory approaches in combating both these diseases, we have also presented the state of the art of such therapeutics and prophylactics. Currently, several vaccines against these viruses have been approved or are under clinical trials in various parts of the world. Therefore, we also recapitulate the latest developments in these which would inspire researchers to look for possibilities of developing vaccines against many other RNA viruses. We hope that the similar aspects in COVID-19 and EVD open up new avenues for the development of pan-viral therapies.
Collapse
Affiliation(s)
| | - Aswathy Shailaja
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Aarcha Shanmugha Mary
- Department of Microbiology, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610105, India;
| | | | - Ambili Savithri
- Department of Biochemistry, Sree Narayana College, Kollam 691001, India;
| | | | | | | | - Maya Madhavan
- Department of Biochemistry, Government College for Women, Thiruvananthapuram 695014, India
| |
Collapse
|
178
|
Verstegen NJM, Hagen RR, van den Dijssel J, Kuijper LH, Kreher C, Ashhurst T, Kummer LYL, Steenhuis M, Duurland M, de Jongh R, de Jong N, van der Schoot CE, Bos AV, Mul E, Kedzierska K, van Dam KPJ, Stalman EW, Boekel L, Wolbink G, Tas SW, Killestein J, van Kempen ZLE, Wieske L, Kuijpers TW, Eftimov F, Rispens T, van Ham SM, ten Brinke A, van de Sandt CE. Immune dynamics in SARS-CoV-2 experienced immunosuppressed rheumatoid arthritis or multiple sclerosis patients vaccinated with mRNA-1273. eLife 2022; 11:e77969. [PMID: 35838348 PMCID: PMC9337853 DOI: 10.7554/elife.77969] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background Patients affected by different types of autoimmune diseases, including common conditions such as multiple sclerosis (MS) and rheumatoid arthritis (RA), are often treated with immunosuppressants to suppress disease activity. It is not fully understood how the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific humoral and cellular immunity induced by infection and/or upon vaccination is affected by immunosuppressants. Methods The dynamics of cellular immune reactivation upon vaccination of SARS-CoV-2 experienced MS patients treated with the humanized anti-CD20 monoclonal antibody ocrelizumab (OCR) and RA patients treated with methotrexate (MTX) monotherapy were analyzed at great depth via high-dimensional flow cytometry of whole blood samples upon vaccination with the SARS-CoV-2 mRNA-1273 (Moderna) vaccine. Longitudinal B and T cell immune responses were compared to SARS-CoV-2 experienced healthy controls (HCs) before and 7 days after the first and second vaccination. Results OCR-treated MS patients exhibit a preserved recall response of CD8+ T central memory cells following first vaccination compared to HCs and a similar CD4+ circulating T follicular helper 1 and T helper 1 dynamics, whereas humoral and B cell responses were strongly impaired resulting in absence of SARS-CoV-2-specific humoral immunity. MTX treatment significantly delayed antibody levels and B reactivation following the first vaccination, including sustained inhibition of overall reactivation marker dynamics of the responding CD4+ and CD8+ T cells. Conclusions Together, these findings indicate that SARS-CoV-2 experienced MS-OCR patients may still benefit from vaccination by inducing a broad CD8+ T cell response which has been associated with milder disease outcome. The delayed vaccine-induced IgG kinetics in RA-MTX patients indicate an increased risk after the first vaccination, which might require additional shielding or alternative strategies such as treatment interruptions in vulnerable patients. Funding This research project was supported by ZonMw (The Netherlands Organization for Health Research and Development, #10430072010007), the European Union's Horizon 2020 research and innovation program under the Marie Skłodowska-Curie grant agreement (#792532 and #860003), the European Commission (SUPPORT-E, #101015756) and by PPOC (#20_21 L2506), the NHMRC Leadership Investigator Grant (#1173871).
Collapse
Affiliation(s)
- Niels JM Verstegen
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Ruth R Hagen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Jet van den Dijssel
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Lisan H Kuijper
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Christine Kreher
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Thomas Ashhurst
- Sydney Cytometry Core Research Facility, Charles Perkins Centre, Centenary Institute, and The University of SydneySydneyAustralia
- School of Medical Sciences, Faculty of Medicine and Health, The University of SydneySydneyAustralia
| | - Laura YL Kummer
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, University of AmsterdamAmsterdamNetherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Mariel Duurland
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Rivka de Jongh
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Nina de Jong
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner LaboratoryAmsterdamNetherlands
| | - Amélie V Bos
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Erik Mul
- Department of Research Facilities, Sanquin ResearchAmsterdamNetherlands
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido UniversitySapporoJapan
| | - Koos PJ van Dam
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, University of AmsterdamAmsterdamNetherlands
| | - Eileen W Stalman
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, University of AmsterdamAmsterdamNetherlands
| | - Laura Boekel
- Department of Rheumatology, Amsterdam Rheumatology and immunology CenterAmsterdamNetherlands
| | - Gertjan Wolbink
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
- Department of Rheumatology, Amsterdam Rheumatology and immunology CenterAmsterdamNetherlands
| | - Sander W Tas
- Amsterdam Rheumatology and immunology Center, Department of Rheumatology and Clinical Immunology, University of AmsterdamAmsterdamNetherlands
| | - Joep Killestein
- Amsterdam UMC, Vrije Universiteit, Department of NeurologyAmsterdamNetherlands
| | - Zoé LE van Kempen
- Amsterdam UMC, Vrije Universiteit, Department of NeurologyAmsterdamNetherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, University of AmsterdamAmsterdamNetherlands
- Department of Clinical Neurophysiology, St Antonius HospitalNieuwegeinNetherlands
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, University of AmsterdamAmsterdamNetherlands
| | - Filip Eftimov
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, University of AmsterdamAmsterdamNetherlands
| | - Theo Rispens
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
- Swammerdam Institute for Life Sciences, University of AmsterdamAmsterdamNetherlands
| | - Anja ten Brinke
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
| | - Carolien E van de Sandt
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, University of AmsterdamAmsterdamNetherlands
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| |
Collapse
|
179
|
Neutralizing antibody activity against 21 SARS-CoV-2 variants in older adults vaccinated with BNT162b2. Nat Microbiol 2022; 7:1180-1188. [PMID: 35836002 PMCID: PMC9352594 DOI: 10.1038/s41564-022-01163-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
SARS-CoV-2 variants may threaten the effectiveness of vaccines and antivirals to mitigate serious COVID-19 disease. This is of most concern in clinically vulnerable groups such as older adults. We analysed 72 sera samples from 37 individuals, aged 70–89 years, vaccinated with two doses of BNT162b2 (Pfizer–BioNTech) 3 weeks apart, for neutralizing antibody responses to wildtype SARS-CoV-2. Between 3 and 20 weeks after the second vaccine dose, neutralizing antibody titres fell 4.9-fold to a median titre of 21.3 (neutralization dose 80%), with 21.6% of individuals having no detectable neutralizing antibodies at the later time point. Next, we examined neutralization of 21 distinct SARS-CoV-2 variant spike proteins with these sera, and confirmed substantial antigenic escape, especially for the Omicron (B.1.1.529, BA.1/BA.2), Beta (B.1.351), Delta (B.1.617.2), Theta (P.3), C.1.2 and B.1.638 spike variants. By combining pseudotype neutralization with specific receptor-binding domain (RBD) enzyme-linked immunosorbent assays, we showed that changes to position 484 in the spike RBD were mainly responsible for SARS-CoV-2 neutralizing antibody escape. Nineteen sera from the same individuals boosted with a third dose of BNT162b2 contained higher neutralizing antibody titres, providing cross-protection against Omicron BA.1 and BA.2. Despite SARS-CoV-2 immunity waning over time in older adults, booster vaccines can elicit broad neutralizing antibodies against a large number of SARS-CoV-2 variants in this clinically vulnerable cohort. Analysis of the neutralizing antibody activity from sera of vaccinated individuals aged between 70 and 89 reveals a reduction of antibody titres against SARS-CoV-2 wildtype and antigenic escape of various variants of concern that links to specific mutations within the RBD. A booster vaccination helps increasing neutralizing antibody titres against the Omicron BA.1 and BA.2 variants in older adults.
Collapse
|
180
|
Barbeau DJ, Martin JM, Carney E, Dougherty E, Doyle JD, Dermody TS, Hoberman A, Williams JV, Michaels MG, Alcorn JF, Paul Duprex W, McElroy AK. Comparative analysis of human immune responses following SARS-CoV-2 vaccination with BNT162b2, mRNA-1273, or Ad26.COV2.S. NPJ Vaccines 2022; 7:77. [PMID: 35794181 PMCID: PMC9258461 DOI: 10.1038/s41541-022-00504-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 06/22/2022] [Indexed: 12/30/2022] Open
Abstract
SARS-CoV-2 vaccines BNT162b2, mRNA-1273, and Ad26.COV2.S received emergency use authorization by the U.S. Food and Drug Administration in 2020/2021. Individuals being vaccinated were invited to participate in a prospective longitudinal comparative study of immune responses elicited by the three vaccines. In this observational cohort study, immune responses were evaluated using a SARS-CoV-2 spike protein receptor-binding domain ELISA, SARS-CoV-2 virus neutralization assays and an IFN- γ ELISPOT assay at various times over six months following initial vaccination. mRNA-based vaccines elicited higher magnitude humoral responses than Ad26.COV2.S; mRNA-1273 elicited the most durable humoral response, and all humoral responses waned over time. Neutralizing antibodies against the Delta variant were of lower magnitude than the wild-type strain for all three vaccines. mRNA-1273 initially elicited the greatest magnitude of T cell response, but this declined by 6 months. Declining immunity over time supports the use of booster dosing, especially in the setting of emerging variants.
Collapse
Affiliation(s)
- Dominique J Barbeau
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Judith M Martin
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| | - Emily Carney
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emily Dougherty
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Joshua D Doyle
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Terence S Dermody
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alejandro Hoberman
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - John V Williams
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Marian G Michaels
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - John F Alcorn
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - W Paul Duprex
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anita K McElroy
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Center for Vaccine Research, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
181
|
Impact of Prior Infection on SARS-CoV-2 Antibody Responses in Vaccinated Long-Term Care Facility Staff. mSphere 2022; 7:e0016922. [PMID: 35862798 PMCID: PMC9429942 DOI: 10.1128/msphere.00169-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in 2019 and has resulted in millions of deaths worldwide. Certain populations are at higher risk for infection, especially staff and residents at long-term care facilities (LTCF), due to the congregant living setting and high proportions of residents with many comorbidities. Prior to vaccine availability, these populations represented large fractions of total coronavirus disease 2019 (COVID-19) cases and deaths in the United States. Due to the high-risk setting and outbreak potential, staff and residents were among the first groups to be vaccinated. To define the impact of prior infection on the response to vaccination, we measured antibody responses in a cohort of staff members at an LTCF, many of whom were previously infected by SARS-CoV-2. We found that neutralizing, receptor-binding domain (RBD)-binding, and nucleoprotein (NP)-binding antibody levels were significantly higher after the full vaccination course in individuals that were previously infected and that NP antibody levels could discriminate individuals with prior infection from vaccinated individuals. While an anticipated antibody titer increase was observed after a vaccine booster dose in naive individuals, a boost response was not observed in individuals with previous COVID-19 infection. We observed a strong relationship between neutralizing antibodies and RBD-binding antibodies postvaccination across all groups, whereas no relationship was observed between NP-binding and neutralizing antibodies. One individual with high levels of neutralizing and binding antibodies experienced a breakthrough infection (prior to the introduction of Omicron), demonstrating that the presence of antibodies is not always sufficient for complete protection against infection. These results highlight that a history of COVID-19 exposure significantly increases SARS-CoV-2 antibody responses following vaccination. IMPORTANCE Long-term care facilities (LTCFs) have been disproportionately impacted by COVID-19, due to their communal nature, the high-risk profile of residents, and the vulnerability of residents to respiratory pathogens. In this study, we analyzed the role of prior natural immunity to SARS-CoV-2 in postvaccination antibody responses. The LTCF in our cohort experienced a large outbreak, with almost 40% of staff members becoming infected. We found that individuals that were infected prior to vaccination had higher levels of neutralizing and binding antibodies postvaccination. Importantly, the second vaccine dose significantly boosted antibody levels in those that were immunologically naive prior to vaccination, but not in those that had prior immunity. Regardless of the prevaccination immune status, the levels of binding and neutralizing antibodies were highly correlated. The presence of NP-binding antibodies could be used to identify individuals that were previously infected when prevaccination immune status was not known. Our results reveal that vaccination antibody responses differ depending on prior natural immunity.
Collapse
|
182
|
Qi H, Liu B, Wang X, Zhang L. The humoral response and antibodies against SARS-CoV-2 infection. Nat Immunol 2022; 23:1008-1020. [PMID: 35761083 DOI: 10.1038/s41590-022-01248-5] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/22/2022] [Indexed: 12/14/2022]
Abstract
Two and a half years into the COVID-19 pandemic, we have gained many insights into the human antibody response to the causative SARS-CoV-2 virus. In this Review, we summarize key observations of humoral immune responses in people with COVID-19, discuss key features of infection- and vaccine-induced neutralizing antibodies, and consider vaccine designs for inducing antibodies that are broadly protective against different variants of the SARS-CoV-2 virus.
Collapse
Affiliation(s)
- Hai Qi
- Tsinghua-Peking Center for Life Sciences, Beijing, China. .,Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China. .,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China. .,NexVac Research Center, Tsinghua University, Beijing, China. .,Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing, China. .,Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
| | - Bo Liu
- Tsinghua-Peking Center for Life Sciences, Beijing, China.,Laboratory of Dynamic Immunobiology, Institute for Immunology, Tsinghua University, Beijing, China.,Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Xinquan Wang
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Linqi Zhang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China. .,NexVac Research Center, Tsinghua University, Beijing, China. .,Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China. .,Comprehensive AIDS Research Center, Center for Global Health and Infectious Diseases, Tsinghua University, Beijing, China.
| |
Collapse
|
183
|
Gallian P, Amroun A, Laperche S, Le Cam S, Brisbarre N, Malard L, Nurtop E, Isnard C, Richard P, Morel P, Tiberghien P, de Lamballerie X. Reduced neutralizing antibody potency of COVID-19 convalescent vaccinated plasma against SARS-CoV-2 Omicron variant. Vox Sang 2022; 117:971-975. [PMID: 35577569 PMCID: PMC9347889 DOI: 10.1111/vox.13279] [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: 02/04/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND OBJECTIVE The SARS-CoV-2 Omicron variant displays increased infectiveness as well as mutations resulting in reduced neutralizing activity of antibodies acquired after vaccination or infection involving earlier strains. To assess the ability of vaccinated COVID-19 convalescent plasma (CCP-V) collected before November 2021 to seroneutralize Omicron, we compared neutralizing antibody (nAb) titres of 63 samples against Omicron and earlier B.1 (D614G) strains. METHODS AND FINDINGS Relationship between anti-Omicron titres and IgG anti-S1 levels (binding arbitrary unit: BAU/ml) was studied. Although correlated, anti-Omicron titres were significantly lower than anti-B.1 titres (median = 80 [10-1280] vs. 1280 [160-10,240], p < 0.0001). Omicron nAb titres and IgG anti-S1 levels were correlated (Spearman's rank correlation coefficient = 0.67). Anti-S1 IgG threshold at 7000 BAU/ml may allow to discard CCP-V without anti-Omicron activity (nAb titre <40). Conversely, only those with highest titres (≥160) had systematically anti-S1 IgG levels >7000 BAU/ml. CONCLUSION A fraction of CCP-V collected before November 2021 retains anti-Omicron seroneutralizing activity that may be selected by quantitative anti-IgG assays, but such assays do not easily allow the identification of 'high-titre' CCP-V. However, collecting plasma from vaccinated donors recently infected with Omicron may be the best option to provide optimal CCP-V for immunocompromised patients infected with this variant.
Collapse
Affiliation(s)
- Pierre Gallian
- Etablissement Français du SangLa Plaine Saint DenisFrance
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
| | - Abdennour Amroun
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
| | - Syria Laperche
- Etablissement Français du SangLa Plaine Saint DenisFrance
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
| | - Sophie Le Cam
- Etablissement Français du SangLa Plaine Saint DenisFrance
| | - Nadège Brisbarre
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
- Etablissement français du Sang Provence Alpes Côte d'Azur et CorseMarseilleFrance
| | - Lucile Malard
- Etablissement Français du SangLa Plaine Saint DenisFrance
| | - Elif Nurtop
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
| | - Christine Isnard
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
- Etablissement français du Sang Provence Alpes Côte d'Azur et CorseMarseilleFrance
| | | | - Pascal Morel
- Etablissement Français du SangLa Plaine Saint DenisFrance
- UMR RIGHT 1098, Inserm, Etablissement Français du SangUniversity of Franche‐ComtéBesançonFrance
| | - Pierre Tiberghien
- Etablissement Français du SangLa Plaine Saint DenisFrance
- UMR RIGHT 1098, Inserm, Etablissement Français du SangUniversity of Franche‐ComtéBesançonFrance
| | - Xavier de Lamballerie
- Unité des Virus Emergents (UVE: Aix‐Marseille Univ, IRD 190, Inserm 1207)MarseilleFrance
| |
Collapse
|
184
|
Bhattacharya M, Sharma AR, Dhama K, Agoramoorthy G, Chakraborty C. Hybrid immunity against COVID-19 in different countries with a special emphasis on the Indian scenario during the Omicron period. Int Immunopharmacol 2022; 108:108766. [PMID: 35413676 PMCID: PMC8986476 DOI: 10.1016/j.intimp.2022.108766] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/01/2022] [Accepted: 04/04/2022] [Indexed: 12/25/2022]
Abstract
Hybrid immunity has been accepted as the most robust immunity to fight against SARS-CoV-2. The hybrid immunity against the virus is produced in individuals who have contracted the disease and received the COVID-19 vaccine. This happens due to the cumulative effect of natural and acquired (vaccine) immunity, which provides higher antibody responses compared to natural and vaccine-produced immunity alone. Scientists have noted that it provides about 25 to 100 times higher antibody responses than natural and vaccine-produced immunity alone. Here, we have tried to illustrate the molecular basis of hybrid immunity against various SARS-CoV-2 variants. We have described hybrid immunity under different headings, which are as follows: an overview of hybrid immunity; a comparison between herd immunity and hybrid immunity against SARS-CoV-2; hybrid immunity in different countries; hybrid immunity and different SARS-CoV-2 variants; the molecular basis of hybrid immunity; and hybrid immunity in Indian scenario. India’s large population has recovered from SARS-CoV-2, and data shows that over 1000 million of the population received at least one dose of the vaccine. Besides, many infected individuals who have recovered also received at least one dose of the vaccine leading to hybrid immunity with a less severe third wave compared to the first and second waves. Based on the available data, we hypothesize that people's hybrid immunity could be a major cause of the less severe third wave.
Collapse
Affiliation(s)
- Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore 756020, Odisha, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si 24252, Gangwon-do, Republic of Korea
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly 243122, Uttar Pradesh, India
| | - Govindasamy Agoramoorthy
- College of Pharmacy and Health Care, Tajen University, Yanpu, Pingtung 907, Taiwan; Swami Vivekananda Yoga Anusandhana Samsthana (S-VYASA), Bengaluru, India
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, West Bengal 700126, India.
| |
Collapse
|
185
|
Xiang Y, Huang W, Liu H, Sang Z, Nambulli S, Tubiana J, Williams KL, Duprex WP, Schneidman-Duhovny D, Wilson IA, Taylor DJ, Shi Y. Superimmunity by pan-sarbecovirus nanobodies. Cell Rep 2022; 39:111004. [PMID: 35738279 PMCID: PMC9174178 DOI: 10.1016/j.celrep.2022.111004] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/09/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Abstract
Vaccine boosters and infection can facilitate the development of SARS-CoV-2 antibodies with improved potency and breadth. Here, we observe superimmunity in a camelid extensively immunized with the SARS-CoV-2 receptor-binding domain (RBD). We rapidly isolate a large repertoire of specific ultra-high-affinity nanobodies that bind strongly to all known sarbecovirus clades using integrative proteomics. These pan-sarbecovirus nanobodies (psNbs) are highly effective against SARS-CoV and SARS-CoV-2 variants, including Omicron, with the best median neutralization potency at single-digit nanograms per milliliter. A highly potent, inhalable, and bispecific psNb (PiN-31) is also developed. Structural determinations of 13 psNbs with the SARS-CoV-2 spike or RBD reveal five epitope classes, providing insights into the mechanisms and evolution of their broad activities. The highly evolved psNbs target small, flat, and flexible epitopes that contain over 75% of conserved RBD surface residues. Their potencies are strongly and negatively correlated with the distance of the epitopes from the receptor binding sites.
Collapse
Affiliation(s)
- Yufei Xiang
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wei Huang
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Zhe Sang
- The University of Pittsburgh and Carnegie Mellon University Program for Computational Biology, Pittsburgh, PA 15213, USA
| | - Sham Nambulli
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jérôme Tubiana
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel; Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Kevin L Williams
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - W Paul Duprex
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dina Schneidman-Duhovny
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA 92037, USA; Skaggs Institute for Chemical Biology, Scripps Research, La Jolla, CA 92037, USA
| | - Derek J Taylor
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH 44106, USA; Department of Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yi Shi
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; The University of Pittsburgh and Carnegie Mellon University Program for Computational Biology, Pittsburgh, PA 15213, USA; Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
186
|
Shalash AO, Toth I, Skwarczynski M. The potential of developing a protective peptide-based vaccines against SARS-CoV-2. Drug Dev Res 2022; 83:1251-1256. [PMID: 35751566 PMCID: PMC9349783 DOI: 10.1002/ddr.21969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/20/2022] [Accepted: 06/14/2022] [Indexed: 12/11/2022]
Abstract
COVID‐19 pandemic has been the deadliest infectious disease outbreak since Spanish flu. The emerging variant lineages, decay of neutralizing antibodies, and occur of reinfections require the development of highly protective and safe vaccines. As currently approved COVID‐19 vaccines that utilize virus‐related genetic material are less than ideal, other vaccine types have been also widely investigated. Among them, peptide‐based vaccines hold great promise in countering COVID‐19 as they may overcome most of the shortcomings of RNA/DNA and protein vaccines. Two basic types of potential peptide vaccines can be developed. The first type are those which rely on cytotoxic T‐cell (CTL) responses to kill infected host cells and stop the replication via employing CTL‐epitopes as vaccine antigens. The second type of peptide vaccines are those that rely on B‐cell peptide epitopes to trigger humoral response via generating SARS‐CoV‐2‐specific antibodies to neutralize and/or opsonize the virus. We propose that combining both cellular and humoral immune responses would be highly protective. Here we discuss opportunities and challenges in the development of an effective and safe peptide‐based vaccine against COVID‐19.
Collapse
Affiliation(s)
- Ahmed O Shalash
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia.,School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St. Lucia, Queensland, Australia
| |
Collapse
|
187
|
Cervantes-Luevano K, Espino-Vazquez AN, Flores-Acosta G, Bernaldez-Sarabia J, Cabanillas-Bernal O, Gasperin-Bulbarela J, Gonzalez-Sanchez R, Comas-Garcia A, Licea-Navarro AF. Neutralizing antibodies levels are increased in individuals with heterologous vaccination and hybrid immunity with Ad5-nCoV in the north of Mexico. PLoS One 2022; 17:e0269032. [PMID: 35749390 PMCID: PMC9231729 DOI: 10.1371/journal.pone.0269032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/12/2022] [Indexed: 11/24/2022] Open
Abstract
The coordinated efforts to stop the spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) include massive immunization of the population at a global scale. The humoral immunity against COVID-19 is conferred by neutralizing antibodies (NAbs) that occur during the post-infection period and upon vaccination. Here, we provide robust data showing that potent neutralizing antibodies are induced in convalescent patients of SARS-CoV-2 infection who have been immunized with different types of vaccines, and patients with no previous history of COVID-19 immunized with a mixed vaccination schedule regardless of the previous infection. More importantly, we showed that a heterologous prime-boost in individuals with Ad5-nCoV (Cansino) vaccine induces higher NAbs levels in comparison to a single vaccination scheme alone.
Collapse
Affiliation(s)
- Karla Cervantes-Luevano
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Astrid N. Espino-Vazquez
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Gonzalo Flores-Acosta
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Johanna Bernaldez-Sarabia
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Olivia Cabanillas-Bernal
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Jahaziel Gasperin-Bulbarela
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Ricardo Gonzalez-Sanchez
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| | - Andreu Comas-Garcia
- Facultad de Medicina y Centro de Investigación en Ciencias de la Salud y Biomedicina, UASLP, San Luis Potosi, Mexico
| | - Alexei F. Licea-Navarro
- Department of Biomedical Innovation, Center for Scientific Research and Higher Education at Ensenada, Baja California, Mexico
| |
Collapse
|
188
|
Abstract
Global population immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is accumulating through heterogeneous combinations of infection and vaccination. Vaccine distribution in low- and middle-income countries has been variable and reliant on diverse vaccine platforms. We studied B-cell immunity in Mexico, a middle-income country where five different vaccines have been deployed to populations with high SARS-CoV-2 incidences. Levels of antibodies that bound a stabilized prefusion spike trimer, neutralizing antibody titers, and memory B-cell expansion correlated with each other across vaccine platforms. Nevertheless, the vaccines elicited variable levels of B-cell immunity, and the majority of recipients had undetectable neutralizing activity against the recently emergent omicron variant. SARS-CoV-2 infection, experienced before or after vaccination, potentiated B-cell immune responses and enabled the generation of neutralizing activity against omicron and SARS-CoV for all vaccines in nearly all individuals. These findings suggest that broad population immunity to SARS-CoV-2 will eventually be achieved but by heterogeneous paths.
Collapse
|
189
|
Astakhova EA, Byazrova MG, Yusubalieva GM, Kulemzin SV, Kruglova NA, Prilipov AG, Baklaushev VP, Gorchakov AA, Taranin AV, Filatov AV. Functional Profiling of In Vitro Reactivated Memory B Cells Following Natural SARS-CoV-2 Infection and Gam-COVID-Vac Vaccination. Cells 2022; 11:1991. [PMID: 35805076 PMCID: PMC9265778 DOI: 10.3390/cells11131991] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Both SARS-CoV-2 infection and vaccination have previously been demonstrated to elicit robust, yet somewhat limited immunity against the evolving variants of SARS-CoV-2. Nevertheless, reports performing side-by-side comparison of immune responses following infection vs. vaccination have been relatively scarce. The aim of this study was to compare B-cell response to adenovirus-vectored vaccination in SARS-CoV-2-naive individuals with that observed in the COVID-19 convalescent patients six months after the first encounter with the viral antigens. We set out to use a single analytical platform and performed comprehensive analysis of serum levels of receptor binding domain (RBD)-specific and virus-neutralizing antibodies, frequencies of RBD-binding circulating memory B cells (MBCs), MBC-derived antibody-secreting cells, as well as RBD-specific and virus-neutralizing activity of MBC-derived antibodies after Gam-COVID-Vac (Sputnik V) vaccination and/or natural SARS-CoV-2 infection. Overall, natural immunity was superior to Gam-COVID-Vac vaccination. The levels of neutralizing MBC-derived antibodies in the convalescent patients turned out to be significantly higher than those found following vaccination. Our results suggest that after six months, SARS-CoV-2-specific MBC immunity is more robust in COVID-19 convalescent patients than in Gam-COVID-Vac recipients. Collectively, our data unambiguously indicate that natural immunity outperforms Gam-COVID-Vac-induced immunity six months following recovery/vaccination, which should inform healthcare and vaccination decisions.
Collapse
Affiliation(s)
- Ekaterina A. Astakhova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Department of Immunology, Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Laboratory of Cell Technology, Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of Russia, 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
| | - Sergey V. Kulemzin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Natalia A. Kruglova
- Laboratory of Gene Therapy of Socially Significant Diseases, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology of the Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Alexey G. Prilipov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Laboratory of Molecular Genetics, N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Vladimir P. Baklaushev
- Laboratory of Cell Technology, Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of Russia, 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
| | - Andrey A. Gorchakov
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Alexander V. Taranin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Alexander V. Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
190
|
Li J, Wu J, Long Q, Wu Y, Hu X, He Y, Jiang M, Li J, Zhao L, Yang S, Chen X, Wang M, Zheng J, Wu F, Wu R, Ren L, Bu L, Wang H, Li K, Fu L, Zhang G, Zheng Y, Gao Z. Comprehensive Humoral and Cellular Immune Responses to SARS-CoV-2 Variants in Diverse Chinese Population. RESEARCH (WASHINGTON, D.C.) 2022; 2022:9873831. [PMID: 35935138 PMCID: PMC9275105 DOI: 10.34133/2022/9873831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/01/2022] [Indexed: 12/26/2022]
Abstract
The SARS-CoV-2 variants have been emerging and have made great challenges to current vaccine and pandemic control strategies. It is urgent to understand the current immune status of various Chinese populations given that the preexisting immunity has been established by national vaccination or exposure to past variants. Using sera from 85 individuals (including 21 convalescents of natural infection, 15 cases which suffered a breakthrough infection after being fully vaccinated, and 49 healthy vaccinees), we showed significantly enhanced neutralizing activities against SRAS-CoV-2 variants in convalescent sera, especially those who had been fully vaccinated. The neutralizing antibodies against Omicron were detectable in 75% of convalescents and 44.9% of healthy vaccinees (p = 0.006), with a GMT of 289.5, 180.9-463.3, and 42.6, 31.3-59, respectively. However, the neutralizing activities were weaker in young convalescents (aged < 18 y), with a detectable rate of 50% and a GMT of 46.4 against Omicron. We also examined and found no pan-sarbecovirus neutralizing activities in vaccinated SARS-CoV-1 survivors. A booster dose could further increase the breadth and magnitude of neutralization against WT and variants of concern (VOCs) to different degrees. In addition, we showed that COVID-19-inactivated vaccines can elicit Omicron-specific T-cell responses. The positive rates of ELISpot reactions were 26.7% (4/15) and 43.8% (7/16) in the full vaccination group and the booster vaccination group, respectively, although without statistically significant difference. The neutralizing antibody titers declined while T-cell responses remain consistent over 6 months. These findings will inform the optimization of public health vaccination and intervention strategies to protect diverse populations against SARS-CoV-2 variants. Advances. Breakthrough infection significantly boosted neutralizing activities against SARS-CoV-2 variants as compared to booster immunization with inactivated vaccine. Vaccine-induced virus-specific T-cell immunity, on the other hand, may compensate for the shortfall. Furthermore, the public health system should target the most vulnerable group due to a poorer protective serological response in both infected and vaccinated adolescents.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jing Wu
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Qiuyue Long
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yan'an Wu
- Department of Clinical Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaoyi Hu
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yukun He
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Mingzheng Jiang
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Jia Li
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Lili Zhao
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| | - Shuoqi Yang
- School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaoyong Chen
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Minghui Wang
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jianshi Zheng
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Fangfang Wu
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ruiliang Wu
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Lihong Ren
- Department of Pediatrics, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Liang Bu
- Department of Thoracic Surgery, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Houzhao Wang
- Department of Clinical Laboratory, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Ke Li
- Department of Critical Care Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Lijuan Fu
- Department of Infectious Diseases, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Guojun Zhang
- Cancer Research Center and The Department of Breast-Thyroid-Surgery, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yali Zheng
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
| | - Zhancheng Gao
- Department of Respiratory, Critical Care and Sleep Medicine, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, China
- Department of Respiratory and Critical Care Medicine, Peking University People's Hospital, Beijing, China
| |
Collapse
|
191
|
Das S, Singh J, Shaman H, Singh B, Anantharaj A, Sharanabasava P, Pandey R, Lodha R, Pandey AK, Medigeshi GR. Pre-existing antibody levels negatively correlate with antibody titers after a single dose of BBV152 vaccination. Nat Commun 2022; 13:3451. [PMID: 35705548 PMCID: PMC9199457 DOI: 10.1038/s41467-022-31170-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 06/06/2022] [Indexed: 12/13/2022] Open
Abstract
Many adults in India have received at least one dose of COVID-19 vaccine with or without a prior history SARS-CoV-2 infection. However, there is limited information on the effect of prior immunity on antibody response upon vaccination in India. As immunization of individuals continues, we aimed to assess whether pre-existing antibodies are further boosted by a single dose of BBV152, an inactivated SARS-CoV-2 vaccine, and, if these antibodies can neutralize SARS-CoV-2 Delta and Omicron variants. Here we show that natural infection during the second wave in 2021 led to generation of neutralizing antibodies against other lineages of SARS-CoV-2 including the Omicron variant, albeit at a significantly lower level for the latter. A single dose of BBV152 boosted antibody titers against the Delta and the Omicron variants but the antibody levels remained low against the Omicron variant. Boosting of antibodies showed negative correlation with baseline neutralizing antibody titers.
Collapse
Affiliation(s)
- Suman Das
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, India
| | - Janmejay Singh
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Heena Shaman
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Balwant Singh
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Anbalagan Anantharaj
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Patil Sharanabasava
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
- Serum Institute of India, Pune, India
| | - Rajesh Pandey
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) Laboratory, CSIR-Institute of Genomics and Integrative Biology, Delhi, India
| | - Rakesh Lodha
- All India Institute of Medical Sciences, New Delhi, India
| | - Anil Kumar Pandey
- Employees State Insurance Corporation Medical College and Hospital, Faridabad, Haryana, India
| | | |
Collapse
|
192
|
Abstract
Preexisting immunity to Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) was nonexistent in humans, which coupled with high transmission rates of certain SARS-CoV-2 variants and limited vaccine uptake or availability, has collectively resulted in an ongoing global pandemic. The identification and establishment of one or multiple correlates of protection (CoP) against infectious pathogens is challenging, but beneficial from both the patient care and public health perspectives. Multiple studies have shown that neutralizing antibodies, whether generated following SARS-CoV-2 infection, vaccination, or a combination of both (i.e., hybrid immunity), as well as adaptive cellular immune responses, serve as CoPs for COVID-19. However, the diverse number and type of serologic assays, alongside the lack of cross-assay standardization and emergence of new SARS-CoV-2 variants with immune evasive characteristics, have collectively posed challenges to determining a robust CoP 'threshold' and for the routine utilization of these assays to document 'immunity,' as is commonly done for other vaccine preventable diseases. Here, we discuss what CoPs are, review our current understanding of infection-induced, vaccine-elicited and hybrid immunity to COVID-19 and summarize the current and potential future utility of SARS-CoV-2 serologic testing.
Collapse
Affiliation(s)
- Anisha Misra
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elitza S. Theel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
193
|
Nayrac M, Dubé M, Sannier G, Nicolas A, Marchitto L, Tastet O, Tauzin A, Brassard N, Lima-Barbosa R, Beaudoin-Bussières G, Vézina D, Gong SY, Benlarbi M, Gasser R, Laumaea A, Prévost J, Bourassa C, Gendron-Lepage G, Medjahed H, Goyette G, Ortega-Delgado GG, Laporte M, Niessl J, Gokool L, Morrisseau C, Arlotto P, Richard J, Bélair J, Prat A, Tremblay C, Martel-Laferrière V, Finzi A, Kaufmann DE. Temporal associations of B and T cell immunity with robust vaccine responsiveness in a 16-week interval BNT162b2 regimen. Cell Rep 2022; 39:111013. [PMID: 35732172 PMCID: PMC9189142 DOI: 10.1016/j.celrep.2022.111013] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/27/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Spacing of BNT162b2 mRNA doses beyond 3 weeks raises concerns about vaccine efficacy. We longitudinally analyze B cell, T cell, and humoral responses to two BNT162b2 mRNA doses administered 16 weeks apart in 53 SARS-CoV-2 naive and previously infected donors. This regimen elicits robust RBD-specific B cell responses whose kinetics differs between cohorts, the second dose leading to increased magnitude in naive participants only. While boosting does not increase magnitude of CD4+ T cell responses further compared with the first dose, unsupervised clustering of single-cell features reveals phenotypic and functional shifts over time and between cohorts. Integrated analysis shows longitudinal immune component-specific associations, with early T helper responses post first dose correlating with B cell responses after the second dose, and memory T helper generated between doses correlating with CD8 T cell responses after boosting. Therefore, boosting elicits a robust cellular recall response after the 16-week interval, indicating functional immune memory.
Collapse
Affiliation(s)
- Manon Nayrac
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Mathieu Dubé
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | - Gérémy Sannier
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Alexandre Nicolas
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Lorie Marchitto
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Olivier Tastet
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | - Alexandra Tauzin
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | | | | | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Dani Vézina
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | - Shang Yu Gong
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada
| | - Mehdi Benlarbi
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | - Romain Gasser
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Annemarie Laumaea
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Jérémie Prévost
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | | | | | | | | | | | | | - Julia Niessl
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Laurie Gokool
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada
| | | | | | - Jonathan Richard
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Justin Bélair
- Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Alexandre Prat
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Départment of Neurosciences, Faculty of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Cécile Tremblay
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Valérie Martel-Laferrière
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada
| | - Andrés Finzi
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada; Department of Microbiology and Immunology, McGill University, Montréal, QC H3A 2B4, Canada.
| | - Daniel E Kaufmann
- Centre de Recherche du CHUM, Montréal, QC H2X 0A9, Canada; Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, QC H2X 0A9, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada.
| |
Collapse
|
194
|
Papaneophytou C, Nicolaou A, Pieri M, Nicolaidou V, Galatou E, Sarigiannis Y, Pantelidou M, Panayi P, Thoma T, Stavraki A, Argyrou X, Kalogiannis T, Yiannoukas K, Petrou CC, Felekkis K. Seroprevalence of immunoglobulin G antibodies against SARS-CoV-2 in Cyprus. PLoS One 2022; 17:e0269885. [PMID: 35696396 PMCID: PMC9191710 DOI: 10.1371/journal.pone.0269885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/27/2022] [Indexed: 12/26/2022] Open
Abstract
Monitoring the levels of IgG antibodies against the SARS-CoV-2 is important during the coronavirus disease 2019 (COVID-19) pandemic, to plan an adequate and evidence-based public health response. After this study we report that the plasma levels of IgG antibodies against SARS-CoV-2 spike protein were higher in individuals with evidence of prior infection who received at least one dose of either an mRNA-based vaccine (Comirnaty BNT162b2/Pfizer-BioNTech or Spikevax mRNA-1273/Moderna) or an adenoviral-based vaccine (Vaxzervia ChAdOx1 nCoV-19 /Oxford-Astra Zeneca) (n = 39) compared to i) unvaccinated individuals with evidence of prior infection with SARS-CoV-2 (n = 109) and ii) individuals without evidence of prior infection with SARS-CoV-2 who received one or two doses of one of the aforementioned vaccines (n = 342). Our analysis also revealed that regardless of the vaccine technology (mRNA-based and adenoviral vector-based) two doses achieved high anti- SARS-CoV-2 IgG responses. Our results indicate that vaccine-induced responses lead to higher levels of IgG antibodies compared to those produced following infection with the virus. Additionally, in agreement with previous studies, our results suggest that among individuals previously infected with SARS-CoV-2, even a single dose of a vaccine is adequate to elicit high levels of antibody response.
Collapse
Affiliation(s)
- Christos Papaneophytou
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Andria Nicolaou
- Yiannoukas Medical Laboratories/ Bioiatriki Group, Nicosia, Cyprus
| | - Myrtani Pieri
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Vicky Nicolaidou
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Eleftheria Galatou
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Yiannis Sarigiannis
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | | | - Pavlos Panayi
- Yiannoukas Medical Laboratories/ Bioiatriki Group, Nicosia, Cyprus
| | - Theklios Thoma
- Yiannoukas Medical Laboratories/ Bioiatriki Group, Nicosia, Cyprus
| | - Antonia Stavraki
- Yiannoukas Medical Laboratories/ Bioiatriki Group, Nicosia, Cyprus
| | - Xenia Argyrou
- Yiannoukas Medical Laboratories/ Bioiatriki Group, Nicosia, Cyprus
| | | | | | - Christos C. Petrou
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| | - Kyriacos Felekkis
- Department of Life and Health Science, School of Sciences and Engineering, University of Nicosia, Nicosia, Cyprus
| |
Collapse
|
195
|
Busch MP, Stramer SL, Stone M, Yu EA, Grebe E, Notari E, Saa P, Ferg R, Manrique IM, Weil N, Fink RV, Levy M, Green V, Cyrus S, Williamson PC, Haynes J, Groves J, Krysztof D, Custer B, Kleinman S, Biggerstaff BJ, Opsomer JD, Jones JM. Population-Weighted Seroprevalence From Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection, Vaccination, and Hybrid Immunity Among US Blood Donations From January to December 2021. Clin Infect Dis 2022; 75:S254-S263. [PMID: 35684973 PMCID: PMC9214177 DOI: 10.1093/cid/ciac470] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019 (COVID-19) vaccination, independently and combined ("hybrid immunity"), result in partial protection from subsequent infection and strong protection from severe disease. Proportions of the US population who have been infected, vaccinated, or have hybrid immunity remain unclear, posing a challenge for assessing effective pandemic mitigation strategies. METHODS In this serial cross-sectional study, nationwide blood donor specimens collected during January-December 2021 were tested for anti-spike and anti-nucleocapsid antibodies, and donor COVID-19 vaccination history of ≥1 dose was collected. Monthly seroprevalence induced from SARS-CoV-2 infection, COVID-19 vaccination, or both, were estimated. Estimates were weighted to account for demographic differences from the general population and were compared temporally and by demographic factors. RESULTS Overall, 1 123 855 blood samples were assayed. From January to December 2021, the weighted percentage of donations with seropositivity changed as follows: seropositivity due to vaccination without previous infection, increase from 3.5% (95% confidence interval, 3.4%-3.7%) to 64.0%, (63.5%-64.5%); seropositivity due to previous infection without vaccination, decrease from 15.6% (15.2%-16.0%) to 11.7% (11.4%-12.0%); and seropositivity due to hybrid immunity, increase from 0.7% (0.6%-0.7%) to 18.9% (18.5%-19.3%). Combined seroprevalence from infection, vaccination, or both increased from 19.8% (19.3%-20.2%) to 94.5% (93.5%-94.0%). Infection- and vaccination-induced antibody responses varied significantly by age, race-ethnicity, and region, but not by sex. CONCLUSIONS Our results indicate substantial increases in population humoral immunity from SARS-CoV-2 infection, COVID-19 vaccination, and hybrid immunity during 2021. These findings are important to consider in future COVID-19 studies and long-term pandemic mitigation efforts.
Collapse
Affiliation(s)
- Michael P. Busch
- Corresponding authors Michael P. Busch, MD. PhD Vitalant Research Institute 270 Masonic Avenue San Francisco, CA 94118
| | - Susan L. Stramer
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | - Mars Stone
- Vitalant Research Institute, San Francisco, California, USA,University of California San Francisco, San Francisco, California, USA
| | - Elaine A. Yu
- Vitalant Research Institute, San Francisco, California, USA,University of California San Francisco, San Francisco, California, USA
| | - Eduard Grebe
- Vitalant Research Institute, San Francisco, California, USA,University of California San Francisco, San Francisco, California, USA
| | - Edward Notari
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | - Paula Saa
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | | | | | | | | | | | | | | | | | - James Haynes
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | - Jamel Groves
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | - David Krysztof
- American Red Cross, Scientific Affairs, Gaithersburg and Rockville, Maryland, USA
| | - Brian Custer
- Vitalant Research Institute, San Francisco, California, USA,University of California San Francisco, San Francisco, California, USA
| | - Steve Kleinman
- Vitalant Research Institute, San Francisco, California, USA
| | | | | | | |
Collapse
|
196
|
Goldberg Y, Mandel M, Bar-On YM, Bodenheimer O, Freedman LS, Ash N, Alroy-Preis S, Huppert A, Milo R. Protection and Waning of Natural and Hybrid Immunity to SARS-CoV-2. N Engl J Med 2022; 386:2201-2212. [PMID: 35613036 PMCID: PMC9165562 DOI: 10.1056/nejmoa2118946] [Citation(s) in RCA: 249] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) provides natural immunity against reinfection. Recent studies have shown waning of the immunity provided by the BNT162b2 vaccine. The time course of natural and hybrid immunity is unknown. METHODS Using the Israeli Ministry of Health database, we extracted data for August and September 2021, when the B.1.617.2 (delta) variant was predominant, on all persons who had been previously infected with SARS-CoV-2 or who had received coronavirus 2019 vaccine. We used Poisson regression with adjustment for confounding factors to compare the rates of infection as a function of time since the last immunity-conferring event. RESULTS The number of cases of SARS-CoV-2 infection per 100,000 person-days at risk (adjusted rate) increased with the time that had elapsed since vaccination with BNT162b2 or since previous infection. Among unvaccinated persons who had recovered from infection, this rate increased from 10.5 among those who had been infected 4 to less than 6 months previously to 30.2 among those who had been infected 1 year or more previously. Among persons who had received a single dose of vaccine after previous infection, the adjusted rate was low (3.7) among those who had been vaccinated less than 2 months previously but increased to 11.6 among those who had been vaccinated at least 6 months previously. Among previously uninfected persons who had received two doses of vaccine, the adjusted rate increased from 21.1 among those who had been vaccinated less than 2 months previously to 88.9 among those who had been vaccinated at least 6 months previously. CONCLUSIONS Among persons who had been previously infected with SARS-CoV-2 (regardless of whether they had received any dose of vaccine or whether they had received one dose before or after infection), protection against reinfection decreased as the time increased since the last immunity-conferring event; however, this protection was higher than that conferred after the same time had elapsed since receipt of a second dose of vaccine among previously uninfected persons. A single dose of vaccine after infection reinforced protection against reinfection.
Collapse
Affiliation(s)
- Yair Goldberg
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Micha Mandel
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Yinon M Bar-On
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Omri Bodenheimer
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Laurence S Freedman
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Nachman Ash
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Sharon Alroy-Preis
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Amit Huppert
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| | - Ron Milo
- From the Faculty of Industrial Engineering and Management, Technion-Israel Institute of Technology, Haifa (Y.G.), the Department of Statistics and Data Science, Hebrew University of Jerusalem (M.M.), and the Israeli Ministry of Health (O.B., N.A., S.A.-P.), Jerusalem, the Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot (Y.M.B.-O., R.M.), the Biostatistics and Biomathematics Unit, Gertner Institute for Epidemiology and Health Policy Research, Sheba Medical Center, Ramat Gan (L.S.F., A.H.), and the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv (A.H.) - all in Israel
| |
Collapse
|
197
|
Pollet J, Strych U, Chen WH, Versteeg L, Keegan B, Zhan B, Wei J, Liu Z, Lee J, Kundu R, Adhikari R, Poveda C, Jose Villar M, Rani Thimmiraju S, Lopez B, Gillespie PM, Ronca S, Kimata JT, Reers M, Paradkar V, Hotez PJ, Elena Bottazzi M. Receptor-binding domain recombinant protein on alum-CpG induces broad protection against SARS-CoV-2 variants of concern. Vaccine 2022; 40:3655-3663. [PMID: 35568591 PMCID: PMC9080055 DOI: 10.1016/j.vaccine.2022.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 03/18/2022] [Accepted: 05/03/2022] [Indexed: 01/17/2023]
Abstract
We conducted preclinical studies in mice using a yeast-produced SARS-CoV-2 RBD subunit vaccine candidate formulated with aluminum hydroxide (alum) and CpG deoxynucleotides. This formulation is equivalent to the CorbevaxTM vaccine that recently received emergency use authorization by the Drugs Controller General ofIndia. We compared the immune response of mice vaccinated with RBD/alum to mice vaccinated with RBD/alum + CpG. We also evaluated mice immunized with RBD/alum + CpG and boosted with RBD/alum. Mice were immunized twice intramuscularly at a 21-day interval. Compared to two doses of the /alum formulation, the RBD/alum + CpG vaccine induced a stronger and more balanced Th1/Th2 cellular immune response, with high levels of neutralizing antibodies against the original Wuhan isolate of SARS-CoV-2 as well as the B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617.2 and (Delta) variants. Neutralizing antibody titers against the B.1.1.529 (BA.1, Omicron) variant exceeded those in human convalescent plasma after Wuhan infection but were lower than against the other variants. Interestingly, the second dose did not benefit from the addition of CpG, possibly allowing dose-sparing of the adjuvant in the future. The data reported here reinforces that the RBD/alum + CpG vaccine formulation is suitable for inducing broadly neutralizing antibodies against SARS-CoV-2, including variants of concern.
Collapse
Affiliation(s)
- Jeroen Pollet
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Wen-Hsiang Chen
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Leroy Versteeg
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Brian Keegan
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Bin Zhan
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Junfei Wei
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Zhuyun Liu
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Jungsoon Lee
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Rahki Kundu
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Rakesh Adhikari
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Cristina Poveda
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Maria Jose Villar
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | | | - Brianna Lopez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Portia M Gillespie
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA
| | - Shannon Ronca
- Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jason T Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA; James A. Baker III Institute for Public Policy, Rice University, Houston, TX, USA
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, Houston, TX, USA; Department of Pediatrics and Molecular Virology & Microbiology, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA; Department of Biology, Baylor University, Waco, TX, USA.
| |
Collapse
|
198
|
He WT, Musharrafieh R, Song G, Dueker K, Tse LV, Martinez DR, Schäfer A, Callaghan S, Yong P, Beutler N, Torres JL, Volk RM, Zhou P, Yuan M, Liu H, Anzanello F, Capozzola T, Parren M, Garcia E, Rawlings SA, Smith DM, Wilson IA, Safonova Y, Ward AB, Rogers TF, Baric RS, Gralinski LE, Burton DR, Andrabi R. Targeted isolation of diverse human protective broadly neutralizing antibodies against SARS-like viruses. Nat Immunol 2022; 23:960-970. [PMID: 35654851 PMCID: PMC10083051 DOI: 10.1038/s41590-022-01222-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/20/2022] [Indexed: 01/09/2023]
Abstract
The emergence of current severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) and potential future spillovers of SARS-like coronaviruses into humans pose a major threat to human health and the global economy. Development of broadly effective coronavirus vaccines that can mitigate these threats is needed. Here, we utilized a targeted donor selection strategy to isolate a large panel of human broadly neutralizing antibodies (bnAbs) to sarbecoviruses. Many of these bnAbs are remarkably effective in neutralizing a diversity of sarbecoviruses and against most SARS-CoV-2 VOCs, including the Omicron variant. Neutralization breadth is achieved by bnAb binding to epitopes on a relatively conserved face of the receptor-binding domain (RBD). Consistent with targeting of conserved sites, select RBD bnAbs exhibited protective efficacy against diverse SARS-like coronaviruses in a prophylaxis challenge model in vivo. These bnAbs provide new opportunities and choices for next-generation antibody prophylactic and therapeutic applications and provide a molecular basis for effective design of pan-sarbecovirus vaccines.
Collapse
Affiliation(s)
- Wan-Ting He
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Rami Musharrafieh
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Ge Song
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Katharina Dueker
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Longping V Tse
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - David R Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sean Callaghan
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Peter Yong
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Nathan Beutler
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Reid M Volk
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Panpan Zhou
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Hejun Liu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Fabio Anzanello
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Tazio Capozzola
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
| | - Mara Parren
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Elijah Garcia
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
| | - Stephen A Rawlings
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Davey M Smith
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ian A Wilson
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Yana Safonova
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, USA
| | - Andrew B Ward
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Thomas F Rogers
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA
- Division of Infectious Diseases, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Departments of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Lisa E Gralinski
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Dennis R Burton
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA.
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA.
| | - Raiees Andrabi
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, USA.
- International AIDS Vaccine Initiative Neutralizing Antibody Center, The Scripps Research Institute, La Jolla, CA, USA.
- Consortium for HIV/AIDS Vaccine Development, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
199
|
Doke P, Gothankar JS, Doke PP, Kulkarni MM, Khalate KK, Shrivastava S, Patil JR, Arankalle VA. Time dependent decline of neutralizing antibody titers in COVID-19 patients from Pune, India and evidence of reinfection. Microbes Infect 2022; 24:104979. [PMID: 35452812 PMCID: PMC9020493 DOI: 10.1016/j.micinf.2022.104979] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 03/09/2022] [Accepted: 04/07/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE To assess modulation of neutralizing antibody titers in COVID-19 patients and understand association of variables such as age, presence of comorbidity, BMI and gender with antibody titers. METHODS Patients (n = 100) diagnosed from 20th March 2020 to 17th August 2020 and treated at two large hospitals from Pune, India were included and followed up (clinical and serologic) for varied periods. IgG-anti-SARS-CoV-2 (Spike protein-based ELISA) and neutralizing antibody titers (NAb, PRNT) were determined in all the samples. RESULTS Of the 100 patients enrolled initially (median 60 days of diagnosis), follow up samples were collected from 70 patients (median 106 days of diagnosis). Overall, NAb titers reduced significantly (p < 0.001) and as early as 3-4 months. During two visits, 20% and 7.1% patients reported some symptoms. At the first visit, NAb titers were higher in patients with severe disease (p < 0.001), comorbidities (p < 0.005), age <50 years (p < 0.05) and male gender (p < 0.05). Multivariate analysis identified older age (p < 0.001), duration post-diagnosis and female gender as independent variables influencing NAb titers (negative correlation, p < 0.05). During the follow-up, reduction in NAb titers was recorded in patients with comorbidity (p < 0.05), mild disease (p < 0.05), age <50 years (p < 0.05), higher BMI (p < 0.05) and male gender (p < 0.001). Serology identified six cases of asymptomatic reinfections. CONCLUSIONS Decline of NAb titers was associated with age <50 years, mild disease, comorbidities, higher BMI and male gender. At the time of follow up, 8/70 (11.4%) patients lacked neutralizing antibodies. Evidence of 6 probable asymptomatic reinfections suggests waning of immunity, but, probable protection from clinical disease needing hospitalization.
Collapse
Affiliation(s)
- Purwa Doke
- Department of Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Jayshree Sachin Gothankar
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Prakash Prabhakarrao Doke
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | | | | | - Shubham Shrivastava
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Katraj, Pune, Maharashtra, India
| | - Jayesh Rangrao Patil
- Department of Community Medicine, Bharati Vidyapeeth Deemed University Medical College, Pune-Satara Road, Katraj, Pune 411043, Maharashtra, India
| | - Vidya Avinash Arankalle
- Department of Communicable Diseases, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune-Satara Road, Katraj, Pune, Maharashtra, India,Corresponding author
| |
Collapse
|
200
|
Chang MR, Ke H, Coherd CD, Wang Y, Mashima K, Kastrunes GM, Huang CY, Marasco WA. Analysis of a SARS-CoV-2 convalescent cohort identified a common strategy for escape of vaccine-induced anti-RBD antibodies by Beta and Omicron variants. EBioMedicine 2022; 80:104025. [PMID: 35533497 PMCID: PMC9073271 DOI: 10.1016/j.ebiom.2022.104025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/27/2022] [Accepted: 04/11/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Evolutionary pressure has led to the emergence of SARS-CoV-2 variants, with the most recent Omicron variant containing an unparalleled 30 mutations in the spike protein. Many of these mutations are expected to increase immune evasion, thus making breakthrough cases and re-infection more common. METHODS From June 2020 to December 2021 serial blood samples (initial post recovery, 6 months, 12 months) were collected from a COVID-19 convalescent cohort in Boston, MA. Plasma was isolated for use in Mesoscale Discovery based antibody binding assays. Unvaccinated donors or those vaccinated prior to the primary blood draw were excluded from this analysis, as were those who did not have at least two blood draws. Wilcoxon signed rank tests were used to compare pre- and post-vaccination titers and antibody response against different variants, while McNemar tests were used to compare the proportions of achieving ≥ 4 fold increases against different variants. FINDINGS Forty-eight COVID convalescent donors with post-infection vaccination (hybrid immunity) were studied to evaluate the levels of cross-reactive antibodies pre- and post- vaccination against various SARS-CoV-2 Spike and receptor binding domain (RBD) proteins. Vaccination with BNT162b2, mRNA-1273 or Ad26.COV2.S led to a 6·3 to 7·8 fold increase in anti-Spike antibody titers and a 7·0 to 7·4 fold increase in anti-WT, Alpha and Delta RBD antibody. However, a lower response was observed for Beta and Omicron RBDs with only 7/48 (15%) and 15/48 (31%) donors having a ≥4 fold increase in post-vaccination titers against Beta and Omicron RBDs. Structural analysis of the Beta and Omicron RBDs reveal a shared immune escape strategy involving residues K417-E484-N501 that is exploited by these variants of concern. INTERPRETATION Through mutations of the K417-E484-N501 triad, SARS-CoV-2 has evolved to evade neutralization by the class I/II anti-RBD antibody fraction of hybrid immunity plasma as the polyclonal antibody response post-vaccination shows limitations in the ability to solve the structural requirements to bind the mutant RBDs. FUNDING Massachusetts Consortium on Pathogen Readiness (280870.5116709.0016) and the National Institute of Allergy and Infectious Diseases (1R01AI161152-01A1).
Collapse
Affiliation(s)
- Matthew R Chang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Hanzhong Ke
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Christian D Coherd
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Kiyomi Mashima
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Gabriella M Kastrunes
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Chiung-Yu Huang
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, United States
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|