1
|
Brook B, Checkervarty AK, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The BNT162b2 mRNA vaccine demonstrates reduced age-associated T H1 support in vitro and in vivo. iScience 2024; 27:111055. [PMID: 39569372 PMCID: PMC11576392 DOI: 10.1016/j.isci.2024.111055] [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: 09/05/2023] [Revised: 07/05/2024] [Accepted: 09/24/2024] [Indexed: 11/22/2024] Open
Abstract
mRNA vaccines demonstrate impaired immunogenicity and durability in vulnerable older populations. We hypothesized that human in vitro modeling and proteomics could elucidate age-specific mRNA vaccine actions. BNT162b2-stimulation changed the plasma proteome of blood samples from young (18-50Y) and older adult (≥60Y) participants, assessed by mass spectrometry, proximity extension assay, and multiplex. Young adult up-regulation (e.g., PSMC6, CPN1) contrasted reduced induction in older adults (e.g., TPM4, APOF, APOC2, CPN1, PI16). 30-85% lower TH1-polarizing cytokines and chemokines were induced in elderly blood (e.g., IFNγ, CXCL10). Analytes lower in older adult samples included human in vivo mRNA immunogenicity biomarkers (e.g., IFNγ, CXCL10, CCL4, IL-1RA). BNT162b2 also demonstrated reduced CD4+ TH1 responses in aged vs. young adult mice. Our study demonstrates the utility of human in vitro platforms modeling age-specific mRNA vaccine immunogenicity, highlights impaired support of TH1 polarization in older adults, and provides a rationale for precision mRNA vaccine adjuvantation to induce greater immunogenicity.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC V6Z 2K5, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
| | - Soumik Barman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Amy C Sherman
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lindsey R Baden
- Department of Medicine, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Paolo Palma
- Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy
- Department of Systems Medicine- Chair of Pediatrics, University of Rome, 00133 Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dheeraj Soni
- Global Investigative Toxicology, Preclinical Safety, Sanofi, Cambridge, MA 02142, USA
| | - Simon D van Haren
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Al Ozonoff
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Hanno Steen
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - David J Dowling
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Ofer Levy
- Precision Vaccines Program, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
2
|
Jung W, Abdelnour A, Kaplonek P, Herrero R, Shih-Lu Lee J, Barbati DR, Chicz TM, Levine KS, Fantin RC, Loria V, Porras C, Lauffenburger DA, Gail MH, Aparicio A, Hildesheim A, Alter G, McNamara RP. SARS-CoV-2 infection prior to vaccination amplifies Fc-mediated humoral profiles in an age-dependent manner. Cell Rep 2024; 43:114684. [PMID: 39213155 DOI: 10.1016/j.celrep.2024.114684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/24/2024] [Accepted: 08/09/2024] [Indexed: 09/04/2024] Open
Abstract
Immunity acquired by vaccination following infection, termed hybrid immunity, has been shown to confer enhanced protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) by enhancing the breadth and potency of immune responses. Here, we assess Fc-mediated humoral profiles in hybrid immunity and their association with age and vaccine type. Participants are divided into three groups: infection only, vaccination only, and vaccination following infection (i.e., hybrid immunity). Using systems serology, we profile humoral immune responses against spikes and subdomains of SARS-CoV-2 variants. We find that hybrid immunity is characterized by superior Fc receptor binding and natural killer (NK) cell-, neutrophil-, and complement-activating antibodies, which is higher than what can be expected from the sum of the vaccination and infection. These differences between hybrid immunity and vaccine-induced immunity are more pronounced in aged adults, especially for immunoglobulin (Ig)G1, IgG2, and Fcγ receptor-binding antibodies. Our findings suggest that vaccination strategies that aim to mimic hybrid immunity should consider age as an important modifier.
Collapse
Affiliation(s)
- Wonyeong Jung
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Paulina Kaplonek
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Rolando Herrero
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | | | - Domenic R Barbati
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Taras M Chicz
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Kate S Levine
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Romain Clement Fantin
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Viviana Loria
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Carolina Porras
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mitchell H Gail
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD, USA
| | - Amada Aparicio
- Caja Costarricense de Seguro Social, San José, Costa Rica
| | - Allan Hildesheim
- Agencia Costarricense de Investigaciones Biomédicas, Fundación INCIENSA, San José, Costa Rica
| | - Galit Alter
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA.
| | - Ryan P McNamara
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
3
|
Lemos-Pérez G, Barrese-Pérez Y, Chacón-Quintero Y, Uranga-Piña R, Avila-Albuerne Y, Figueroa-García I, Calderín-Marín O, Gómez-Vázquez MM, Piñera-Martínez M, Chávez-Valdés S, Martínez-Rosales R, Ávila-Díaz L, Vázquez-Arteaga A, González-Formental H, Freyre-Corrales G, Coizeau-Rodríguez E, Limonta-Fernández M, Ayala-Avila M, Martínez-Díaz E, Pimentel-Vazquez E, Guillen G. Safety and Immunogenicity of the Intranasal Vaccine Candidate Mambisa and the Intramuscular Vaccine Abdala Used as Booster Doses for COVID-19 Convalescents: A Randomized Phase 1-2 Clinical Trial. Vaccines (Basel) 2024; 12:1001. [PMID: 39340031 PMCID: PMC11435458 DOI: 10.3390/vaccines12091001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 07/23/2024] [Accepted: 08/02/2024] [Indexed: 09/30/2024] Open
Abstract
A phase 1-2, prospective, multicenter, randomized, open-label clinical trial (Code RPCEC00000382), with parallel groups, involving 1161 participants, was designed to assess the safety and immunogenicity of two Cuban COVID-19 vaccines (Mambisa and Abdala) in boosting COVID-19 immunity of convalescent adults after receiving one dose of either vaccine. The main safety outcome was severe vaccination adverse events occurring in <5% of vaccinees. Main immunogenicity success endpoints were a ≥4-fold anti-RBD IgG seroconversion or a ≥20% increase in ACE2-RBD inhibitory antibodies in >55% of vaccinees in Phase 1 and >70% in Phase 2. Neutralizing antibody titers against SARS-CoV-2 variants were evaluated. Both vaccines were safe-no deaths or severe adverse events occurred. Mild intensity adverse events were the most frequent (>73%); headaches predominated for both vaccines. Phase 1 responders were 83.3% (p = 0.0018) for Abdala. Mambisa showed similar results. Phase 2 responders were 88.6% for Abdala (p < 0.0001) and 74.2% for Mambisa (p = 0.0412). In both phases, anti-RBD IgG titers, inhibition percentages and neutralizing antibody titers increased significantly after the booster dose. Both vaccines were safe and their immunogenicity surpassed the study endpoints.
Collapse
Affiliation(s)
- Gilda Lemos-Pérez
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Yinet Barrese-Pérez
- National Coordinating Center for Clinical Trials (CENCEC), La Habana 11300, Cuba; (Y.B.-P.); (R.U.-P.); (Y.A.-A.)
| | - Yahima Chacón-Quintero
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Rolando Uranga-Piña
- National Coordinating Center for Clinical Trials (CENCEC), La Habana 11300, Cuba; (Y.B.-P.); (R.U.-P.); (Y.A.-A.)
| | - Yisel Avila-Albuerne
- National Coordinating Center for Clinical Trials (CENCEC), La Habana 11300, Cuba; (Y.B.-P.); (R.U.-P.); (Y.A.-A.)
| | | | - Osaida Calderín-Marín
- Manuel Ascunce Domenech Provincial Clinical-Surgical Teaching Hospital, Camagüey 70100, Cuba;
| | | | | | - Sheila Chávez-Valdés
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Ricardo Martínez-Rosales
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Lismary Ávila-Díaz
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Amalia Vázquez-Arteaga
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Hany González-Formental
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Giselle Freyre-Corrales
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Edelgis Coizeau-Rodríguez
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Miladys Limonta-Fernández
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | - Marta Ayala-Avila
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
| | | | | | - Gerardo Guillen
- Center for Genetic Engineering and Biotechnology (CIGB), P.O. Box 6162, La Habana 10600, Cuba; (Y.C.-Q.); (S.C.-V.); (R.M.-R.); (L.Á.-D.); (A.V.-A.); (H.G.-F.); (G.F.-C.); (E.C.-R.); (M.L.-F.); (M.A.-A.)
- Latin American School of Medicine (ELAM), La Habana 19108, Cuba
| |
Collapse
|
4
|
Alessandra R, Sara C, Claudia P, Natasha G, Federica C, Chiara B, Tobia F, Stefano T, Eleonora R, Andrea M, Martin MN, Caterina UF, Nigel T, Stefania DSM, Lucia L, Chiara P. Immune signature in vaccinated versus non-vaccinated aged people with COVID-19 pneumonia. J Transl Med 2024; 22:755. [PMID: 39135151 PMCID: PMC11318244 DOI: 10.1186/s12967-024-05556-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND A definition of the immunological features of COVID-19 pneumonia is needed to support clinical management of aged patients. In this study, we characterized the humoral and cellular immune responses in presence or absence of SARS-CoV-2 vaccination, in aged patients admitted to the IRCCS San Raffaele Hospital (Italy) for COVID-19 pneumonia between November 2021 and March 2022. METHODS The study was approved by local authorities. Disease severity was evaluated according to WHO guidelines. We tested: (A) anti-SARS-CoV-2 humoral response (anti-RBD-S IgG, anti-S IgM, anti-N IgG, neutralizing activity against Delta, BA1, BA4/5 variants); (B) Lymphocyte B, CD4 and CD8 T-cell phenotype; (C) plasma cytokines. The impact of vaccine administration and different variants on the immunological responses was evaluated using standard linear regression models and Tobit models for censored outcomes adjusted for age, vaccine doses and gender. RESULT We studied 47 aged patients (median age 78.41), 22 (47%) female, 33 (70%) older than 70 years (elderly). At hospital admission, 36% were unvaccinated (VACno), whilst 63% had received 2 (VAC2) or 3 doses (VAC3) of vaccine. During hospitalization, WHO score > 5 was higher in unvaccinated (14% in VAC3 vs. 43% in VAC2 and 44% VACno). Independently from vaccination doses and gender, elderly had overall reduced anti-SARS-CoV-2 humoral response (IgG-RBD-S, p = 0.0075). By linear regression, the anti-RBD-S (p = 0.0060), B (p = 0.0079), CD8 (p = 0.0043) and Th2 cell counts (p = 0.0131) were higher in VAC2 + 3 compared to VACno. Delta variant was the most representative in VAC2 (n = 13/18, 72%), detected in 41% of VACno, whereas undetected in VAC3, and anti-RBD-S production was higher in VAC2 vs. VACno (p = 0.0001), alongside neutralization against Delta (p = 0141), BA1 (p = 0.0255), BA4/5 (p = 0.0162). Infections with Delta also drove an increase of pro-inflammatory cytokines (IFN-α, p = 0.0463; IL-6, p = 0.0010). CONCLUSIONS Administration of 3 vaccination doses reduces the severe symptomatology in aged and elderly. Vaccination showed a strong association with anti-SARS-CoV-2 humoral response and an expansion of Th2 T-cells populations, independently of age. Delta variants and number of vaccine doses affected the magnitude of the humoral response against the original SARS-CoV-2 and emerging variants. A systematic surveillance of the emerging variants is paramount to define future vaccination strategies.
Collapse
Affiliation(s)
- Ruggiero Alessandra
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Caldrer Sara
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Negrar, Italy
| | - Pastori Claudia
- Division of Immunology, Transplantation and Infectious Disease, Immunobiology of HIV Group, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianesini Natasha
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Cugnata Federica
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Brombin Chiara
- University Centre of Statistics in the Biomedical Sciences, Vita-Salute San Raffaele University, Milan, Italy
| | - Fantoni Tobia
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Tais Stefano
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Negrar, Italy
| | - Rizzi Eleonora
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Negrar, Italy
| | - Matucci Andrea
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Negrar, Italy
| | - Mayora-Neto Martin
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham, UK
| | | | - Temperton Nigel
- Viral Pseudotype Unit, Medway School of Pharmacy, Universities of Kent and Greenwich, Chatham, UK
| | | | - Lopalco Lucia
- Division of Immunology, Transplantation and Infectious Disease, Immunobiology of HIV Group, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Piubelli Chiara
- Department of Infectious, Tropical Diseases and Microbiology, IRCCS Sacro Cuore Don Calabria Hospital, Verona, Negrar, Italy
| |
Collapse
|
5
|
Liu HH, Xie Y, Yang BP, Wen HY, Yang PH, Lu JE, Liu Y, Chen X, Qu MM, Zhang Y, Hong WG, Li YG, Fu J, Wang FS. Safety, immunogenicity and protective effect of sequential vaccination with inactivated and recombinant protein COVID-19 vaccine in the elderly: a prospective longitudinal study. Signal Transduct Target Ther 2024; 9:129. [PMID: 38740763 DOI: 10.1038/s41392-024-01846-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/19/2024] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
The safety and efficacy of COVID-19 vaccines in the elderly, a high-risk group for severe COVID-19 infection, have not been fully understood. To clarify these issues, this prospective study followed up 157 elderly and 73 young participants for 16 months and compared the safety, immunogenicity, and efficacy of two doses of the inactivated vaccine BBIBP-CorV followed by a booster dose of the recombinant protein vaccine ZF2001. The results showed that this vaccination protocol was safe and tolerable in the elderly. After administering two doses of the BBIBP-CorV, the positivity rates and titers of neutralizing and anti-RBD antibodies in the elderly were significantly lower than those in the young individuals. After the ZF2001 booster dose, the antibody-positive rates in the elderly were comparable to those in the young; however, the antibody titers remained lower. Gender, age, and underlying diseases were independently associated with vaccine immunogenicity in elderly individuals. The pseudovirus neutralization assay showed that, compared with those after receiving two doses of BBIBP-CorV priming, some participants obtained immunological protection against BA.5 and BF.7 after receiving the ZF2001 booster. Breakthrough infection symptoms last longer in the infected elderly and pre-infection antibody titers were negatively associated with the severity of post-infection symptoms. The antibody levels in the elderly increased significantly after breakthrough infection but were still lower than those in the young. Our data suggest that multiple booster vaccinations at short intervals to maintain high antibody levels may be an effective strategy for protecting the elderly against COVID-19.
Collapse
MESH Headings
- Humans
- COVID-19/prevention & control
- COVID-19/immunology
- Female
- Male
- Aged
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/adverse effects
- COVID-19 Vaccines/administration & dosage
- SARS-CoV-2/immunology
- Prospective Studies
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/adverse effects
- Vaccines, Inactivated/administration & dosage
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Aged, 80 and over
- Adult
- Vaccination
- Longitudinal Studies
- Middle Aged
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/administration & dosage
- Immunogenicity, Vaccine/immunology
- Immunization, Secondary
Collapse
Affiliation(s)
- Hong-Hong Liu
- Out-patient Department of Day Diagnosis and Treatment, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Yunbo Xie
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China
| | - Bao-Peng Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Huan-Yue Wen
- Hunyuan County People's Hospital, Datong, 037499, Shanxi Province, China
| | - Peng-Hui Yang
- Faculty of Hepato-Pancreato-Biliary Surgery, Institute of Hepatobiliary Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jin-E Lu
- Hunyuan County People's Hospital, Datong, 037499, Shanxi Province, China
| | - Yan Liu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Xi Chen
- Out-patient Department of Day Diagnosis and Treatment, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Meng-Meng Qu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Yang Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Wei-Guo Hong
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Yong-Gang Li
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China
| | - Junliang Fu
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China.
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, 100039, China.
- Chinese PLA Medical School, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
6
|
Serwanga J, Kato L, Oluka GK, Ankunda V, Sembera J, Baine C, Kitabye I, Namuyanja A, Opio S, Katende JS, Ejou P, Kaleebu P. The single-dose Janssen Ad26.COV2.S COVID-19 vaccine elicited robust and persistent anti-spike IgG antibody responses in a 12-month Ugandan cohort. Front Immunol 2024; 15:1384668. [PMID: 38779677 PMCID: PMC11109398 DOI: 10.3389/fimmu.2024.1384668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction The study investigation examined the immune response to the Janssen Ad26.COV2.S COVID-19 vaccine within a Ugandan cohort, specifically targeting antibodies directed against spike (S) and nucleocapsid (N) proteins. We aimed to examine the durability and robustness of the induced antibody response while also assessing occurrences of breakthrough infections and previous anti-Spike seropositivity to SARS-CoV-2. Methods The study included 319 specimens collected over 12 months from 60 vaccinees aged 18 to 64. Binding antibodies were quantified using a validated ELISA method to measure SARS-CoV-2-specific IgG, IgM, and IgA levels against the S and N proteins. Results The results showed that baseline seropositivity for S-IgG was high at 67%, increasing to 98% by day 14 and consistently stayed above 95% for up to 12 months. However, S-IgM responses remained suboptimal. A raised S-IgA seropositivity rate was seen that doubled from 40% at baseline to 86% just two weeks following the initial vaccine dose, indicating sustained and robust peripheral immunity. An increase in N-IgG levels at nine months post-vaccination suggested breakthrough infections in eight cases. Baseline cross-reactivity influenced spike-directed antibody responses, with individuals harbouring S-IgG antibodies showing notably higher responses. Discussion Robust and long lasting vaccine and infection-induced immune responses were observed, with significant implications for regions where administering subsequent doses poses logistical challenges.
Collapse
Affiliation(s)
- Jennifer Serwanga
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Laban Kato
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Gerald Kevin Oluka
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Violet Ankunda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Jackson Sembera
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Claire Baine
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Isaac Kitabye
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Angela Namuyanja
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Solomon Opio
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - Joseph Ssebwana Katende
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Peter Ejou
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
| | - The COVID-19 Immunoprofiling Team
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| | - Pontiano Kaleebu
- Department of Immunology, Uganda Virus Research Institute, Entebbe, Uganda
- Viral Pathogens Research Theme, Medical Research Council, Uganda Virus Research Institute and London School of Hygiene and Tropical Medicine, Uganda Research Unit, Entebbe, Uganda
| |
Collapse
|
7
|
Collins CP, Longo DL, Murphy WJ. The immunobiology of SARS-CoV-2 infection and vaccine responses: potential influences of cross-reactive memory responses and aging on efficacy and off-target effects. Front Immunol 2024; 15:1345499. [PMID: 38469293 PMCID: PMC10925677 DOI: 10.3389/fimmu.2024.1345499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Immune responses to both SARS-CoV-2 infection and its associated vaccines have been highly variable within the general population. The increasing evidence of long-lasting symptoms after resolution of infection, called post-acute sequelae of COVID-19 (PASC) or "Long COVID," suggests that immune-mediated mechanisms are at play. Closely related endemic common human coronaviruses (hCoV) can induce pre-existing and potentially cross-reactive immunity, which can then affect primary SARS-CoV-2 infection, as well as vaccination responses. The influence of pre-existing immunity from these hCoVs, as well as responses generated from original CoV2 strains or vaccines on the development of new high-affinity responses to CoV2 antigenic viral variants, needs to be better understood given the need for continuous vaccine adaptation and application in the population. Due in part to thymic involution, normal aging is associated with reduced naïve T cell compartments and impaired primary antigen responsiveness, resulting in a reliance on the pre-existing cross-reactive memory cell pool which may be of lower affinity, restricted in diversity, or of shorter duration. These effects can also be mediated by the presence of down-regulatory anti-idiotype responses which also increase in aging. Given the tremendous heterogeneity of clinical data, utilization of preclinical models offers the greatest ability to assess immune responses under a controlled setting. These models should now involve prior antigen/viral exposure combined with incorporation of modifying factors such as age on immune responses and effects. This will also allow for mechanistic dissection and understanding of the different immune pathways involved in both SARS-CoV-2 pathogen and potential vaccine responses over time and how pre-existing memory responses, including potential anti-idiotype responses, can affect efficacy as well as potential off-target effects in different tissues as well as modeling PASC.
Collapse
Affiliation(s)
- Craig P. Collins
- Graduate Program in Immunology, University of California (UC) Davis, Davis, CA, United States
| | - Dan L. Longo
- Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - William J. Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), University of California (UC) Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
8
|
Huot N, Planchais C, Rosenbaum P, Contreras V, Jacquelin B, Petitdemange C, Lazzerini M, Beaumont E, Orta-Resendiz A, Rey FA, Reeves RK, Le Grand R, Mouquet H, Müller-Trutwin M. SARS-CoV-2 viral persistence in lung alveolar macrophages is controlled by IFN-γ and NK cells. Nat Immunol 2023; 24:2068-2079. [PMID: 37919524 PMCID: PMC10681903 DOI: 10.1038/s41590-023-01661-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/26/2023] [Indexed: 11/04/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA generally becomes undetectable in upper airways after a few days or weeks postinfection. Here we used a model of viral infection in macaques to address whether SARS-CoV-2 persists in the body and which mechanisms regulate its persistence. Replication-competent virus was detected in bronchioalveolar lavage (BAL) macrophages beyond 6 months postinfection. Viral propagation in BAL macrophages occurred from cell to cell and was inhibited by interferon-γ (IFN-γ). IFN-γ production was strongest in BAL NKG2r+CD8+ T cells and NKG2Alo natural killer (NK) cells and was further increased in NKG2Alo NK cells after spike protein stimulation. However, IFN-γ production was impaired in NK cells from macaques with persisting virus. Moreover, IFN-γ also enhanced the expression of major histocompatibility complex (MHC)-E on BAL macrophages, possibly inhibiting NK cell-mediated killing. Macaques with less persisting virus mounted adaptive NK cells that escaped the MHC-E-dependent inhibition. Our findings reveal an interplay between NK cells and macrophages that regulated SARS-CoV-2 persistence in macrophages and was mediated by IFN-γ.
Collapse
Affiliation(s)
- Nicolas Huot
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France.
| | - Cyril Planchais
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Pierre Rosenbaum
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Vanessa Contreras
- Université Paris-Saclay, INSERM, CEA, Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Beatrice Jacquelin
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Caroline Petitdemange
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Marie Lazzerini
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Emma Beaumont
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Aurelio Orta-Resendiz
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| | - Félix A Rey
- Institut Pasteur, Université Paris-Cité, Structural Virology Unit, CNRS UMR3569, Paris, France
| | - R Keith Reeves
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Innate and Comparative Immunology, Center for Human Systems Immunology, Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Ragon Institute of Massachusetts General Hospital, MIT, Cambridge, MA, USA
- Duke Research and Discovery at RTP, Duke University Health System, Durham, NC, USA
| | - Roger Le Grand
- Université Paris-Saclay, INSERM, CEA, Immunologie des Maladies Virales, Auto-Immunes, Hématologiques et Bactériennes (IMVA-HB/IDMIT/UMR1184), Fontenay-aux-Roses & Kremlin Bicêtre, France
| | - Hugo Mouquet
- Institut Pasteur, Université Paris Cité, INSERM U1222, Humoral Immunology Unit, Paris, France
| | - Michaela Müller-Trutwin
- Institut Pasteur, Université Paris-Cité, HIV, Inflammation and Persistence Unit, Paris, France
| |
Collapse
|
9
|
Lopez-Gomez A, Pelaez-Prestel HF, Juarez I. Approaches to evaluate the specific immune responses to SARS-CoV-2. Vaccine 2023; 41:6434-6443. [PMID: 37770298 DOI: 10.1016/j.vaccine.2023.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
The SARS-CoV-2 pandemic has a huge impact on public health and global economy, meaning an enormous scientific, political, and social challenge. Studying how infection or vaccination triggers both cellular and humoral responses is essential to know the grade and length of protection generated in the population. Nowadays, scientists and authorities around the world are increasingly concerned about the arrival of new variants, which have a greater spread, due to the high mutation rate of this virus. The aim of this review is to summarize the different techniques available for the study of the immune responses after exposure or vaccination against SARS-CoV-2, showing their advantages and limitations, and proposing suitable combinations of different techniques to achieve extensive information in these studies. We wish that the information provided here will helps other scientists in their studies of the immune response against SARS-CoV-2 after vaccination with new vaccine candidates or infection with upcoming variants.
Collapse
Affiliation(s)
- Ana Lopez-Gomez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Hector F Pelaez-Prestel
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| | - Ignacio Juarez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
10
|
Ferreira IATM, Lee CYC, Foster WS, Abdullahi A, Dratva LM, Tuong ZK, Stewart BJ, Ferdinand JR, Guillaume SM, Potts MOP, Perera M, Krishna BA, Peñalver A, Cabantous M, Kemp SA, Ceron-Gutierrez L, Ebrahimi S, Lyons P, Smith KGC, Bradley J, Collier DA, McCoy LE, van der Klaauw A, Thaventhiran JED, Farooqi IS, Teichmann SA, MacAry PA, Doffinger R, Wills MR, Linterman MA, Clatworthy MR, Gupta RK. Atypical B cells and impaired SARS-CoV-2 neutralization following heterologous vaccination in the elderly. Cell Rep 2023; 42:112991. [PMID: 37590132 DOI: 10.1016/j.celrep.2023.112991] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
Suboptimal responses to a primary vaccination course have been reported in the elderly, but there is little information regarding the impact of age on responses to booster third doses. Here, we show that individuals 70 years or older (median age 73, range 70-75) who received a primary two-dose schedule with AZD1222 and booster third dose with mRNA vaccine achieve significantly lower neutralizing antibody responses against SARS-CoV-2 spike pseudotyped virus compared with those younger than 70 (median age 66, range 54-69) at 1 month post booster. Impaired neutralization potency and breadth post third dose in the elderly is associated with circulating "atypical" spike-specific B cells expressing CD11c and FCRL5. However, when considering individuals who received three doses of mRNA vaccine, we did not observe differences in neutralization or enrichment in atypical B cells. This work highlights the finding that AdV and mRNA COVID-19 vaccine formats differentially instruct the memory B cell response.
Collapse
Affiliation(s)
- Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Colin Y C Lee
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - William S Foster
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - John R Ferdinand
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Stephane M Guillaume
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Martin O P Potts
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Benjamin A Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ana Peñalver
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Mia Cabantous
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Soraya Ebrahimi
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Paul Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Bradley
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami A Collier
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Agatha van der Klaauw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - Paul A MacAry
- National University of Singapore, Singapore, Singapore
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Mark R Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michelle A Linterman
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
11
|
Swadling L, Maini MK. Can T Cells Abort SARS-CoV-2 and Other Viral Infections? Int J Mol Sci 2023; 24:4371. [PMID: 36901802 PMCID: PMC10002440 DOI: 10.3390/ijms24054371] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Despite the highly infectious nature of the SARS-CoV-2 virus, it is clear that some individuals with potential exposure, or even experimental challenge with the virus, resist developing a detectable infection. While a proportion of seronegative individuals will have completely avoided exposure to the virus, a growing body of evidence suggests a subset of individuals are exposed, but mediate rapid viral clearance before the infection is detected by PCR or seroconversion. This type of "abortive" infection likely represents a dead-end in transmission and precludes the possibility for development of disease. It is, therefore, a desirable outcome on exposure and a setting in which highly effective immunity can be studied. Here, we describe how early sampling of a new pandemic virus using sensitive immunoassays and a novel transcriptomic signature can identify abortive infections. Despite the challenges in identifying abortive infections, we highlight diverse lines of evidence supporting their occurrence. In particular, expansion of virus-specific T cells in seronegative individuals suggests abortive infections occur not only after exposure to SARS-CoV-2, but for other coronaviridae, and diverse viral infections of global health importance (e.g., HIV, HCV, HBV). We discuss unanswered questions related to abortive infection, such as: 'Are we just missing antibodies? Are T cells an epiphenomenon? What is the influence of the dose of viral inoculum?' Finally, we argue for a refinement of the current paradigm that T cells are only involved in clearing established infection; instead, we emphasise the importance of considering their role in terminating early viral replication by studying abortive infections.
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K. Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| |
Collapse
|
12
|
Lo Tartaro D, Paolini A, Mattioli M, Swatler J, Neroni A, Borella R, Santacroce E, Di Nella A, Gozzi L, Busani S, Cuccorese M, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Piwocka K, Gibellini L, Cossarizza A, De Biasi S. Detailed characterization of SARS-CoV-2-specific T and B cells after infection or heterologous vaccination. Front Immunol 2023; 14:1123724. [PMID: 36845156 PMCID: PMC9947839 DOI: 10.3389/fimmu.2023.1123724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
The formation of a robust long-term antigen (Ag)-specific memory, both humoral and cell-mediated, is created following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. Here, by using polychromatic flow cytometry and complex data analyses, we deeply investigated the magnitude, phenotype, and functionality of SARS-CoV-2-specific immune memory in two groups of healthy subjects after heterologous vaccination compared to a group of subjects who recovered from SARS-CoV-2 infection. We find that coronavirus disease 2019 (COVID-19) recovered patients show different long-term immunological profiles compared to those of donors who had been vaccinated with three doses. Vaccinated individuals display a skewed T helper (Th)1 Ag-specific T cell polarization and a higher percentage of Ag-specific and activated memory B cells expressing immunoglobulin (Ig)G compared to those of patients who recovered from severe COVID-19. Different polyfunctional properties characterize the two groups: recovered individuals show higher percentages of CD4+ T cells producing one or two cytokines simultaneously, while the vaccinated are distinguished by highly polyfunctional populations able to release four molecules, namely, CD107a, interferon (IFN)-γ, tumor necrosis factor (TNF), and interleukin (IL)-2. These data suggest that functional and phenotypic properties of SARS-CoV-2 adaptive immunity differ in recovered COVID-19 individuals and vaccinated ones.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Julian Swatler
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- National Institute for Cardiovascular Research, Bologna, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| |
Collapse
|
13
|
Touizer E, Alrubayyi A, Ford R, Hussain N, Gerber PP, Shum HL, Rees-Spear C, Muir L, Gea-Mallorquí E, Kopycinski J, Jankovic D, Jeffery-Smith A, Pinder CL, Fox TA, Williams I, Mullender C, Maan I, Waters L, Johnson M, Madge S, Youle M, Barber TJ, Burns F, Kinloch S, Rowland-Jones S, Gilson R, Matheson NJ, Morris E, Peppa D, McCoy LE. Attenuated humoral responses in HIV after SARS-CoV-2 vaccination linked to B cell defects and altered immune profiles. iScience 2023; 26:105862. [PMID: 36590902 PMCID: PMC9788849 DOI: 10.1016/j.isci.2022.105862] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
We assessed a cohort of people living with human immunodeficiency virus (PLWH) (n = 110) and HIV negative controls (n = 64) after 1, 2 or 3 SARS-CoV-2 vaccine doses. At all timepoints, PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs). Improved neutralization breadth was seen against the Omicron variant (BA.1) after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global MBC dysfunction. In contrast, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, individuals with low or absent neutralization had detectable functional T cell responses. These PLWH had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3+CD127+CD8+T cells after two doses of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Emma Touizer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rosemarie Ford
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Noshin Hussain
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hiu-Long Shum
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Chloe Rees-Spear
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Luke Muir
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dylan Jankovic
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Christopher L. Pinder
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Thomas A. Fox
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Ian Williams
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Claire Mullender
- Institute for Global Health, University College London, London, UK
| | - Irfaan Maan
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Margaret Johnson
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sara Madge
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Michael Youle
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Tristan J. Barber
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Fiona Burns
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sabine Kinloch
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | | | - Richard Gilson
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Emma Morris
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Laura E. McCoy
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
14
|
Brook B, Fatou B, Kumar Checkervarty A, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The mRNA vaccine BNT162b2 demonstrates impaired T H1 immunogenicity in human elders in vitro and aged mice in vivo. RESEARCH SQUARE 2022:rs.3.rs-2395118. [PMID: 36597547 PMCID: PMC9810224 DOI: 10.21203/rs.3.rs-2395118/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
mRNA vaccines have been key to addressing the SARS-CoV-2 pandemic but have impaired immunogenicity and durability in vulnerable older populations. We evaluated the mRNA vaccine BNT162b2 in human in vitro whole blood assays with supernatants from adult (18-50 years) and elder (≥60 years) participants measured by mass spectrometry and proximity extension assay proteomics. BNT162b2 induced increased expression of soluble proteins in adult blood (e.g., C1S, PSMC6, CPN1), but demonstrated reduced proteins in elder blood (e.g., TPM4, APOF, APOC2, CPN1, and PI16), including 30-85% lower induction of TH1-polarizing cytokines and chemokines (e.g., IFNγ, and CXCL10). Elder TH1 impairment was validated in mice in vivo and associated with impaired humoral and cellular immunogenicity. Our study demonstrates the utility of a human in vitro platform to model age-specific mRNA vaccine activity, highlights impaired TH1 immunogenicity in older adults, and provides rationale for developing enhanced mRNA vaccines with greater immunogenicity in vulnerable populations.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC, Canada
| | - Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Amy C Sherman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Bambino Gesù Children's Hospital, Rome, Italy
- Chair of Pediatrics, University of Rome, Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
15
|
Mendez-Cortina Y, Rodriguez-Perea AL, Chvatal-Medina M, Lopera TJ, Alvarez-Mesa N, Rodas-Marín JK, Moncada DC, Rugeles MT, Velilla PA. Dynamics of humoral immune response in SARS-CoV-2 infected individuals with different clinical stages. Front Immunol 2022; 13:1007068. [DOI: 10.3389/fimmu.2022.1007068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
BackgroundThe COVID-19 pandemic remains a global health problem. As in other viral infections, the humoral immune response against SARS-CoV-2 is thought to be crucial for controlling the infection. However, the dynamic of B cells in the clinical spectrum of this disease is still controversial. This study aimed to characterize B cell subsets and neutralizing responses in COVID-19 patients according to disease severity through a one-month follow-up.MethodsA cohort of 71 individuals with SARS-CoV-2 infection confirmed by RT-PCR were recruited and classified into four groups: i) asymptomatic; ii) symptomatic outpatients; iii) hospitalized in ward, and iv) intensive care unit patients (ICU). Samples were taken at days 0 (inclusion to the study), 7 and 30. B cell subsets and neutralizing antibodies were assessed using multiparametric flow cytometry and plaque reduction neutralization, respectively.ResultsOlder age, male gender and body mass index over 25 were common factors among hospitalized and ICU patients, compared to those with milder clinical presentations. In addition, those requiring hospitalization had more comorbidities. A significant increase in the frequencies of CD19+ cells at day 0 was observed in hospitalized and ICU patients compared to asymptomatic and symptomatic groups. Likewise, the frequency of plasmablasts was significantly increased at the first sample in the ICU group compared to the asymptomatic group, but then waned over time. The frequency of naïve B cells decreased at days 7 and 30 compared to day 0 in hospitalized and ICU patients. The neutralizing antibody titers were higher as the severity of COVID-19 increased; in asymptomatic individuals, it was strongly correlated with the percentage of IgM+ switched memory B cells, and a moderate correlation was found with plasmablasts.ConclusionThe humoral immune response is variable among SARS-CoV-2 infected people depending on the severity and time of clinical evolution. In severe COVID-19 patients, a higher plasmablast frequency and neutralizing antibody response were observed, suggesting that, despite having a robust humoral immunity, this response could be late, having a low impact on disease outcome.
Collapse
|
16
|
Touizer E, Alrubbayi A, Ford R, Hussain N, Gerber PP, Shum HL, Rees-Spear C, Muir L, Gea-Mallorquí E, Kopycinski J, Jankovic D, Pinder C, Fox TA, Williams I, Mullender C, Maan I, Waters L, Johnson M, Madge S, Youle M, Barber T, Burns F, Kinloch S, Rowland-Jones S, Gilson R, Matheson NJ, Morris E, Peppa D, McCoy LE. Attenuated humoral responses in HIV infection after SARS-CoV-2 vaccination are linked to global B cell defects and cellular immune profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.11.516111. [PMID: 36380764 PMCID: PMC9665338 DOI: 10.1101/2022.11.11.516111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
People living with HIV (PLWH) on suppressive antiretroviral therapy (ART) can have residual immune dysfunction and often display poorer responses to vaccination. We assessed in a cohort of PLWH (n=110) and HIV negative controls (n=64) the humoral and spike-specific B-cell responses following 1, 2 or 3 SARS-CoV-2 vaccine doses. PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls at all studied timepoints. Moreover, their neutralization breadth was reduced with fewer individuals developing a neutralizing response against the Omicron variant (BA.1) relative to controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs) and pronounced B cell dysfunction. Improved neutralization breadth was seen after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global, but not spike-specific, MBC dysfunction. In contrast to the inferior antibody responses, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, a subset of PLWH with low or absent neutralization had detectable functional T cell responses. These individuals had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3 + CD127 + CD8 + T cells after two doses of SARS-CoV-2 vaccination, which may compensate for sub-optimal serological responses in the event of infection. Therefore, normalisation of B cell homeostasis could improve serological responses to vaccines in PLWH and evaluating T cell immunity could provide a more comprehensive immune status profile in these individuals and others with B cell imbalances.
Collapse
Affiliation(s)
- Emma Touizer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Aljawharah Alrubbayi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- Nuffield Department of Medicine, University of Oxford, UK
| | - Rosemarie Ford
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Noshin Hussain
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, UK
| | - Hiu-Long Shum
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Chloe Rees-Spear
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Luke Muir
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | | | | | - Dylan Jankovic
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Christopher Pinder
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Thomas A Fox
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Ian Williams
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
| | | | - Irfaan Maan
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
| | - Margaret Johnson
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Sara Madge
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Michael Youle
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Tristan Barber
- Institute for Global Health, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Fiona Burns
- Institute for Global Health, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Sabine Kinloch
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | | | - Richard Gilson
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Emma Morris
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Laura E McCoy
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| |
Collapse
|
17
|
Mountz JD, Gao M, Ponder DM, Liu S, Sun CW, Alduraibi F, Sullivan K, Pat B, Dell'Italia LJ, Hsu HC. IL-4 receptor blockade is a global repressor of naïve B cell development and responses in a dupilumab-treated patient. Clin Immunol 2022; 244:109130. [PMID: 36189576 PMCID: PMC9741950 DOI: 10.1016/j.clim.2022.109130] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 09/14/2022] [Accepted: 09/14/2022] [Indexed: 12/14/2022]
Abstract
Here, we report a case of atopic dermatitis (AD) in a patient who received biweekly doses of dupilumab, an antibody against the IL-4 receptor α chain (IL-4Rα). Single cell RNA-sequencing showed that naïve B cells expressed the highest levels of IL4R compared to other B cell subpopulations. Compared to controls, the dupilumab-treated patient exhibited diminished percentages of IL4R+IGHD+ naïve B cells and down-regulation of IL4R, FCER2 (CD23), and IGHD. Dupilumab treatment resulted in upregulation of genes associated with apoptosis and inhibition of B cell receptor signaling and down-regulation of class-switch and memory B cell development genes. The dupilumab-treated patient exhibited a rapid decline in COVID-19 anti-spike and anti-receptor binding domain antibodies between 4 and 8 and 11 months post COVID-19 vaccination. Our data suggest that intact and persistent IL-4 signaling is necessary for maintaining robust survival and development of naïve B cells, and maintaining a long term vaccine response.
Collapse
Affiliation(s)
- John D Mountz
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA; Department of Veterans Affairs Health Care System, Birmingham, AL, USA.
| | - Min Gao
- Informatics Institute, The University of Alabama at Birmingham, USA
| | - David M Ponder
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Shanrun Liu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Chiao-Wang Sun
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Fatima Alduraibi
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Kathryn Sullivan
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA
| | - Betty Pat
- Department of Veterans Affairs Health Care System, Birmingham, AL, USA; Department of Medicine, Division of Cardiovascular Disease, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Louis J Dell'Italia
- Department of Veterans Affairs Health Care System, Birmingham, AL, USA; Department of Medicine, Division of Cardiovascular Disease, the University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hui-Chen Hsu
- Division of Clinical Immunology and Rheumatology, Department of Medicine, The University of Alabama at Birmingham, USA.
| |
Collapse
|
18
|
Bates TA, Lu P, Kang YJ, Schoen D, Thornton M, McBride SK, Park C, Kim D, Messer WB, Curlin ME, Tafesse FG, Lu LL. BNT162b2-induced neutralizing and non-neutralizing antibody functions against SARS-CoV-2 diminish with age. Cell Rep 2022; 41:111544. [PMID: 36252569 PMCID: PMC9533669 DOI: 10.1016/j.celrep.2022.111544] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 08/12/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022] Open
Abstract
Each severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant renews concerns about decreased vaccine neutralization weakening efficacy. However, while prevention of infection varies, protection from disease remains and implicates immunity beyond neutralization in vaccine efficacy. Polyclonal antibodies function through Fab domains that neutralize virus and Fc domains that induce non-neutralizing responses via engagement of Fc receptors on immune cells. To understand how vaccines promote protection, we leverage sera from 51 SARS-CoV-2 uninfected individuals after two doses of the BNT162b2 mRNA vaccine. We show that neutralizing activities against clinical isolates of wild-type and five SARS-CoV-2 variants, including Omicron BA.2, link to FcγRIIIa/CD16 non-neutralizing effector functions. This is associated with post-translational afucosylation and sialylation of vaccine-specific antibodies. Further, polyfunctional neutralizing and non-neutralizing breadth, magnitude, and coordination diminish with age. Thus, studying Fc functions in addition to Fab-mediated neutralization provides greater insight into vaccine efficacy for vulnerable populations, such as the elderly, against SARS-CoV-2 and novel variants.
Collapse
Affiliation(s)
- Timothy A Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ye Jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Devin Schoen
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Micah Thornton
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Savannah K McBride
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Chanhee Park
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Daehwan Kim
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - William B Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Marcel E Curlin
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR 97239, USA.
| | - Fikadu G Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR 97239, USA.
| | - Lenette L Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA; Department of Immunology, UT Southwestern Medical Center, Dallas, TX 75390, USA; Parkland Health & Hospital System, Dallas, TX 75235, USA.
| |
Collapse
|
19
|
Diani S, Leonardi E, Cavezzi A, Ferrari S, Iacono O, Limoli A, Bouslenko Z, Natalini D, Conti S, Mantovani M, Tramonte S, Donzelli A, Serravalle E. SARS-CoV-2-The Role of Natural Immunity: A Narrative Review. J Clin Med 2022; 11:6272. [PMID: 36362500 PMCID: PMC9655392 DOI: 10.3390/jcm11216272] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 10/17/2022] [Accepted: 10/20/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Both natural immunity and vaccine-induced immunity to COVID-19 may be useful to reduce the mortality/morbidity of this disease, but still a lot of controversy exists. AIMS This narrative review analyzes the literature regarding these two immunitary processes and more specifically: (a) the duration of natural immunity; (b) cellular immunity; (c) cross-reactivity; (d) the duration of post-vaccination immune protection; (e) the probability of reinfection and its clinical manifestations in the recovered patients; (f) the comparisons between vaccinated and unvaccinated as to the possible reinfections; (g) the role of hybrid immunity; (h) the effectiveness of natural and vaccine-induced immunity against Omicron variant; (i) the comparative incidence of adverse effects after vaccination in recovered individuals vs. COVID-19-naïve subjects. MATERIAL AND METHODS through multiple search engines we investigated COVID-19 literature related to the aims of the review, published since April 2020 through July 2022, including also the previous articles pertinent to the investigated topics. RESULTS nearly 900 studies were collected, and 246 pertinent articles were included. It was highlighted that the vast majority of the individuals after suffering from COVID-19 develop a natural immunity both of cell-mediated and humoral type, which is effective over time and provides protection against both reinfection and serious illness. Vaccine-induced immunity was shown to decay faster than natural immunity. In general, the severity of the symptoms of reinfection is significantly lower than in the primary infection, with a lower degree of hospitalizations (0.06%) and an extremely low mortality. CONCLUSIONS this extensive narrative review regarding a vast number of articles highlighted the valuable protection induced by the natural immunity after COVID-19, which seems comparable or superior to the one induced by anti-SARS-CoV-2 vaccination. Consequently, vaccination of the unvaccinated COVID-19-recovered subjects may not be indicated. Further research is needed in order to: (a) measure the durability of immunity over time; (b) evaluate both the impacts of Omicron BA.5 on vaccinated and healed subjects and the role of hybrid immunity.
Collapse
Affiliation(s)
- Sara Diani
- School of Musictherapy, Université Européenne Jean Monnet, 35129 Padova, Italy
| | | | | | | | - Oriana Iacono
- Physical Medicine and Rehabilitation Department, Mirandola Hospital, 41037 Mirandola, Italy
| | - Alice Limoli
- ARPAV (Regional Agency for the Environment Protection), 31100 Treviso, Italy
| | - Zoe Bouslenko
- Cardiology Department, Valdese Hospital, 10100 Torino, Italy
| | | | | | | | - Silvano Tramonte
- Environment and Health Commission, National Bioarchitecture Institute, 20121 Milano, Italy
| | | | | |
Collapse
|
20
|
Mullender C, da Costa KAS, Alrubayyi A, Pett SL, Peppa D. SARS-CoV-2 immunity and vaccine strategies in people with HIV. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac005. [PMID: 36846557 PMCID: PMC9452103 DOI: 10.1093/oxfimm/iqac005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/24/2022] [Accepted: 08/02/2022] [Indexed: 12/15/2022] Open
Abstract
Current severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccines, based on the ancestral Wuhan strain, were developed rapidly to meet the needs of a devastating global pandemic. People living with Human Immunodeficiency Virus (PLWH) have been designated as a priority group for SARS-CoV-2 vaccination in most regions and varying primary courses (two- or three-dose schedule) and additional boosters are recommended depending on current CD4+ T cell count and/or detectable HIV viraemia. From the current published data, licensed vaccines are safe for PLWH, and stimulate robust responses to vaccination in those well controlled on antiretroviral therapy and with high CD4+ T cell counts. Data on vaccine efficacy and immunogenicity remain, however, scarce in PLWH, especially in people with advanced disease. A greater concern is a potentially diminished immune response to the primary course and subsequent boosters, as well as an attenuated magnitude and durability of protective immune responses. A detailed understanding of the breadth and durability of humoral and T cell responses to vaccination, and the boosting effects of natural immunity to SARS-CoV-2, in more diverse populations of PLWH with a spectrum of HIV-related immunosuppression is therefore critical. This article summarizes focused studies of humoral and cellular responses to SARS-CoV-2 infection in PLWH and provides a comprehensive review of the emerging literature on SARS-CoV-2 vaccine responses. Emphasis is placed on the potential effect of HIV-related factors and presence of co-morbidities modulating responses to SARS-CoV-2 vaccination, and the remaining challenges informing the optimal vaccination strategy to elicit enduring responses against existing and emerging variants in PLWH.
Collapse
Affiliation(s)
- Claire Mullender
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
| | - Kelly A S da Costa
- Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Pett
- Centre for Clinical Research in Infection and Sexual Health, Institute for Global Health, University College London Institute for Global Health, London, UK
- Medical Research Council Clinical Trials Unit, Institute of Clinical Trials and Methodology, London, UK
| | - Dimitra Peppa
- Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
21
|
Bates TA, Lu P, Kang YJ, Schoen D, Thornton M, McBride SK, Park C, Kim D, Messer WB, Curlin ME, Tafesse FG, Lu LL. BNT162b2 induced neutralizing and non-neutralizing antibody functions against SARSCoV-2 diminish with age. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2022.08.12.22278726. [PMID: 36032979 PMCID: PMC9413715 DOI: 10.1101/2022.08.12.22278726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Each novel SARS-CoV-2 variant renews concerns about decreased vaccine efficacy caused by evasion of vaccine induced neutralizing antibodies. However, accumulating epidemiological data show that while vaccine prevention of infection varies, protection from severe disease and death remains high. Thus, immune responses beyond neutralization could contribute to vaccine efficacy. Polyclonal antibodies function through their Fab domains that neutralize virus directly, and Fc domains that induce non-neutralizing host responses via engagement of Fc receptors on immune cells. To understand how vaccine induced neutralizing and non-neutralizing activities synergize to promote protection, we leverage sera from 51 SARS-CoV-2 uninfected health-care workers after two doses of the BNT162b2 mRNA vaccine. We show that BNT162b2 elicits antibodies that neutralize clinical isolates of wildtype and five variants of SARS-CoV-2, including Omicron BA.2, and, critically, induce Fc effector functions. FcγRIIIa/CD16 activity is linked to neutralizing activity and associated with post-translational afucosylation and sialylation of vaccine specific antibodies. Further, neutralizing and non-neutralizing functions diminish with age, with limited polyfunctional breadth, magnitude and coordination observed in those ≥65 years old compared to <65. Thus, studying Fc functions in addition to Fab mediated neutralization provides greater insight into vaccine efficacy for vulnerable populations such as the elderly against SARS-CoV-2 and novel variants.
Collapse
Affiliation(s)
- Timothy A. Bates
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Ye jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
| | - Devin Schoen
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR
| | - Micah Thornton
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Savannah K. McBride
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Chanhee Park
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - Daehwan Kim
- Lyda Hill Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX
| | - William B. Messer
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Marcel E. Curlin
- Department of Occupational Health, Oregon Health and Sciences University, Portland, OR
| | - Fikadu G. Tafesse
- Department of Molecular Microbiology and Immunology, Oregon Health and Sciences University, Portland, OR
| | - Lenette L. Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX
- Department of Immunology, UT Southwestern Medical Center, Dallas, TX
- Parkland Health & Hospital System
| |
Collapse
|
22
|
Balloux F, Tan C, Swadling L, Richard D, Jenner C, Maini M, van Dorp L. The past, current and future epidemiological dynamic of SARS-CoV-2. OXFORD OPEN IMMUNOLOGY 2022; 3:iqac003. [PMID: 35872966 PMCID: PMC9278178 DOI: 10.1093/oxfimm/iqac003] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/11/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
SARS-CoV-2, the agent of the COVID-19 pandemic, emerged in late 2019 in China, and rapidly spread throughout the world to reach all continents. As the virus expanded in its novel human host, viral lineages diversified through the accumulation of around two mutations a month on average. Different viral lineages have replaced each other since the start of the pandemic, with the most successful Alpha, Delta and Omicron variants of concern (VoCs) sequentially sweeping through the world to reach high global prevalence. Neither Alpha nor Delta was characterized by strong immune escape, with their success coming mainly from their higher transmissibility. Omicron is far more prone to immune evasion and spread primarily due to its increased ability to (re-)infect hosts with prior immunity. As host immunity reaches high levels globally through vaccination and prior infection, the epidemic is expected to transition from a pandemic regime to an endemic one where seasonality and waning host immunization are anticipated to become the primary forces shaping future SARS-CoV-2 lineage dynamics. In this review, we consider a body of evidence on the origins, host tropism, epidemiology, genomic and immunogenetic evolution of SARS-CoV-2 including an assessment of other coronaviruses infecting humans. Considering what is known so far, we conclude by delineating scenarios for the future dynamic of SARS-CoV-2, ranging from the good-circulation of a fifth endemic 'common cold' coronavirus of potentially low virulence, the bad-a situation roughly comparable with seasonal flu, and the ugly-extensive diversification into serotypes with long-term high-level endemicity.
Collapse
Affiliation(s)
- François Balloux
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Cedric Tan
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
- Genome Institute of Singapore, Agency for Science, Technology and Research (A*STAR), 138672 Singapore, Singapore
| | - Leo Swadling
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Damien Richard
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Charlotte Jenner
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| | - Mala Maini
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - Lucy van Dorp
- UCL Genetics Institute, University College London, London WC1E 6BT, UK
| |
Collapse
|
23
|
The Effect of Waning on Antibody Levels and Memory B Cell Recall following SARS-CoV-2 Infection or Vaccination. Vaccines (Basel) 2022; 10:vaccines10050696. [PMID: 35632452 PMCID: PMC9143792 DOI: 10.3390/vaccines10050696] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 02/04/2023] Open
Abstract
In order to longitudinally track SARS-CoV-2 antibody levels after vaccination or infection, we assessed anti-RBD antibody levels in over 1000 people and found no significant decrease in antibody levels during the first 14 months after infection in unvaccinated participants, however, a significant waning of antibody levels was observed following vaccination. Participants who were pre-immune to SARS-CoV-2 prior to vaccination seroconverted to higher antibody levels, which were maintained at higher levels than in previously infected, unvaccinated participants. Older participants exhibited lower level of antibodies after vaccination, but a higher level after infection than younger people. The rate of antibody waning was not affected by pre-immunity or age. Participants who received a third dose of an mRNA vaccine not only increased their antibody levels ~14-fold, but also had ~3 times more antibodies compared to when they received their primary vaccine series. PBMC-derived memory B cells from 13 participants who lost all circulating antibodies were differentiated into antibody secreting cells (ASCs). There was a significant recall of memory B cell ASCs in the absence of serum antibodies in 5–8 of the 10 vaccinated participants, but not in any of the 3 infected participants, suggesting a strong connection between antibody levels and the effectiveness of memory B cell recall.
Collapse
|
24
|
Forgacs D, Moraes VS, Hanley HB, Gattiker JL, Jefferson AM, Ross TM. The effect of waning on antibody levels and memory B cell recall following SARS-CoV-2 infection or vaccination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.03.16.484099. [PMID: 35313572 PMCID: PMC8936119 DOI: 10.1101/2022.03.16.484099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As of March 2022, there have been over 450 million reported SARS-CoV-2 cases worldwide, and more than 4 billion people have received their primary series of a COVID-19 vaccine. In order to longitudinally track SARS-CoV-2 antibody levels in people after vaccination or infection, a large-scale COVID-19 sero-surveillance progam entitled SPARTA (SeroPrevalence and Respiratory Tract Assessment) was established early in the pandemic. Anti-RBD antibody levels were tracked in more than 1,000 people. There was no significant decrease in antibody levels during the first 14 months after infection in unvaccinated participants, however, significant waning of antibody levels was observed following vaccination, regardless of previous infection status. Moreover, participants who were pre-immune to SARS-CoV-2 prior to vaccination seroconverted to significantly higher antibody levels, and antibodies were maintained at significantly higher levels than in previously infected, unvaccinated participants. This pattern was entirely due to differences in the magnitude of the initial seroconversion event, and the rate of antibody waning was not significantly different based on the pre-immune status. Participants who received a third (booster) dose of an mRNA vaccine not only increased their anti-RBD antibody levels ∼14-fold, but they also had ∼3 times more anti-RBD antibodies compared to the peak of their antibody levels after receiving their primary vaccine series. In order to ascertain whether the presence of serum antibodies is important for long-term seroprotection, PBMCs from 13 participants who lost all detectable circulating antibodies after vaccination or infection were differentiated into memory cells in vitro . There was a significant recall of memory B cells in the absence of serum antibodies in 70% of the vaccinated participants, but not in any of the infected participants. Therefore, there is a strong connection between anti-RBD antibody levels and the effectiveness of memory B cell recall.
Collapse
Affiliation(s)
- David Forgacs
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Vanessa S. Moraes
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Hannah B. Hanley
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Jasper L. Gattiker
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - Ted M. Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA, USA
| |
Collapse
|
25
|
Mohn KGI, Bredholt G, Zhou F, Madsen A, Onyango TB, Fjelltveit EB, Jalloh SL, Brokstad KA, Cantoni D, Mayora-Neto M, Temperton N, Langeland N, Cox RJ. Durable T-cellular and humoral responses in SARS-CoV-2 hospitalized and community patients. PLoS One 2022; 17:e0261979. [PMID: 35192617 PMCID: PMC8863217 DOI: 10.1371/journal.pone.0261979] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/14/2021] [Indexed: 12/13/2022] Open
Abstract
Background Neutralizing antibodies are important for protection against the pandemic SARS-CoV-2 virus, and long-term memory responses determine the risk of re-infection or boosting after vaccination. T-cellular responses are considered important for partial protection against novel variants of concern. Methods A prospective cohort of hospitalized (n = 14) and community (n = 38) patients with rt-PCR confirmed SARS-CoV-2 infection were recruited. Blood samples and clinical data were collected when diagnosed and at 6 months. Serum samples were analyzed for SARS-CoV-2-spike specific antibodies using ELISA (IgG, IgA, IgM), pseudotype neutralization and microneutralization assays. Peripheral blood mononuclear cells were investigated for virus-specific T-cell responses in the interferon-γ and interleukin-2 fluorescent-linked immunosorbent spot (FluroSpot) assay. Results We found durable SARS-CoV-2 spike- and internal protein specific T-cellular responses in patients with persistent antibodies at 6 months. Significantly higher IL-2 and IFN-γ secreting T-cell responses as well as SARS-CoV-2 specific IgG and neutralizing antibodies were detected in hospitalized compared to community patients. The immune response was impacted by age, gender, comorbidity and severity of illness, reflecting clinical observations. Conclusions SARS-CoV-2 specific T-cellular and antibody responses persisted for 6 months post confirmed infection. In previously infected patients, re-exposure or vaccination will boost long-term immunity, possibly providing protection against re-infection with variant viruses.
Collapse
Affiliation(s)
- Kristin G.-I. Mohn
- Influenza Centre, Bergen, Norway
- Department of Medicine, Bergen, Norway
- * E-mail:
| | | | - Fan Zhou
- Influenza Centre, Bergen, Norway
| | | | | | | | | | - Karl A. Brokstad
- Department of Clinical Science, Broegelmann Research Laboratory, University of Bergen, Bergen, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Nina Langeland
- Influenza Centre, Bergen, Norway
- Department of Medicine, Bergen, Norway
- National Advisory Unit for Tropical Infectious Diseases, Bergen, Norway
| | - Rebecca J. Cox
- Influenza Centre, Bergen, Norway
- Department of Microbiology, Bergen, Norway
| | | |
Collapse
|
26
|
Swadling L, Diniz MO, Schmidt NM, Amin OE, Chandran A, Shaw E, Pade C, Gibbons JM, Le Bert N, Tan AT, Jeffery-Smith A, Tan CCS, Tham CYL, Kucykowicz S, Aidoo-Micah G, Rosenheim J, Davies J, Johnson M, Jensen MP, Joy G, McCoy LE, Valdes AM, Chain BM, Goldblatt D, Altmann DM, Boyton RJ, Manisty C, Treibel TA, Moon JC, van Dorp L, Balloux F, McKnight Á, Noursadeghi M, Bertoletti A, Maini MK. Pre-existing polymerase-specific T cells expand in abortive seronegative SARS-CoV-2. Nature 2022; 601:110-117. [PMID: 34758478 PMCID: PMC8732273 DOI: 10.1038/s41586-021-04186-8] [Citation(s) in RCA: 261] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022]
Abstract
Individuals with potential exposure to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) do not necessarily develop PCR or antibody positivity, suggesting that some individuals may clear subclinical infection before seroconversion. T cells can contribute to the rapid clearance of SARS-CoV-2 and other coronavirus infections1-3. Here we hypothesize that pre-existing memory T cell responses, with cross-protective potential against SARS-CoV-2 (refs. 4-11), would expand in vivo to support rapid viral control, aborting infection. We measured SARS-CoV-2-reactive T cells, including those against the early transcribed replication-transcription complex (RTC)12,13, in intensively monitored healthcare workers (HCWs) who tested repeatedly negative according to PCR, antibody binding and neutralization assays (seronegative HCWs (SN-HCWs)). SN-HCWs had stronger, more multispecific memory T cells compared with a cohort of unexposed individuals from before the pandemic (prepandemic cohort), and these cells were more frequently directed against the RTC than the structural-protein-dominated responses observed after detectable infection (matched concurrent cohort). SN-HCWs with the strongest RTC-specific T cells had an increase in IFI27, a robust early innate signature of SARS-CoV-2 (ref. 14), suggesting abortive infection. RNA polymerase within RTC was the largest region of high sequence conservation across human seasonal coronaviruses (HCoV) and SARS-CoV-2 clades. RNA polymerase was preferentially targeted (among the regions tested) by T cells from prepandemic cohorts and SN-HCWs. RTC-epitope-specific T cells that cross-recognized HCoV variants were identified in SN-HCWs. Enriched pre-existing RNA-polymerase-specific T cells expanded in vivo to preferentially accumulate in the memory response after putative abortive compared to overt SARS-CoV-2 infection. Our data highlight RTC-specific T cells as targets for vaccines against endemic and emerging Coronaviridae.
Collapse
Affiliation(s)
- Leo Swadling
- Division of Infection and Immunity, University College London, London, UK.
| | - Mariana O Diniz
- Division of Infection and Immunity, University College London, London, UK
| | - Nathalie M Schmidt
- Division of Infection and Immunity, University College London, London, UK
| | - Oliver E Amin
- Division of Infection and Immunity, University College London, London, UK
| | - Aneesh Chandran
- Division of Infection and Immunity, University College London, London, UK
| | - Emily Shaw
- Division of Infection and Immunity, University College London, London, UK
| | - Corinna Pade
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Joseph M Gibbons
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nina Le Bert
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
| | - Anthony T Tan
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
| | - Anna Jeffery-Smith
- Division of Infection and Immunity, University College London, London, UK
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Cedric C S Tan
- UCL Genetics Institute, University College London, London, UK
| | - Christine Y L Tham
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
| | | | | | - Joshua Rosenheim
- Division of Infection and Immunity, University College London, London, UK
| | - Jessica Davies
- Division of Infection and Immunity, University College London, London, UK
| | - Marina Johnson
- Great Ormond Street Institute of Child Health NIHR Biomedical Research Centre, University College London, London, UK
| | - Melanie P Jensen
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Department of Cellular Pathology, Northwest London Pathology, Imperial College London NHS Trust, London, UK
| | - George Joy
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Laura E McCoy
- Division of Infection and Immunity, University College London, London, UK
| | - Ana M Valdes
- Academic Rheumatology, Clinical Sciences, Nottingham City Hospital, Nottingham, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
| | - Benjamin M Chain
- Division of Infection and Immunity, University College London, London, UK
| | - David Goldblatt
- Great Ormond Street Institute of Child Health NIHR Biomedical Research Centre, University College London, London, UK
| | - Daniel M Altmann
- Department of Immunology and Inflammation, Imperial College London, London, UK
| | - Rosemary J Boyton
- Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, UK
- Lung Division, Royal Brompton & Harefield Hospitals, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Charlotte Manisty
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Thomas A Treibel
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - James C Moon
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Institute of Cardiovascular Science, University College London, London, UK
| | - Lucy van Dorp
- UCL Genetics Institute, University College London, London, UK
| | | | - Áine McKnight
- Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK
| | - Antonio Bertoletti
- Emerging Infectious Diseases Program, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, A*STAR, Singapore, Singapore
| | - Mala K Maini
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|