1
|
Coutant F, Touret F, Pin JJ, Alonzo M, Baronti C, Munier S, Attia M, de Lamballerie X, Ferry T, Miossec P. Neutralizing and enhancing monoclonal antibodies in SARS-CoV-2 convalescent patients: lessons from early variant infection and impact on shaping emerging variants. Emerg Microbes Infect 2024; 13:2307510. [PMID: 38240255 PMCID: PMC10829827 DOI: 10.1080/22221751.2024.2307510] [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: 10/03/2023] [Accepted: 01/16/2024] [Indexed: 02/01/2024]
Abstract
Serological studies of COVID-19 convalescent patients have identified polyclonal lineage-specific and cross-reactive antibodies (Abs), with varying effector functions against virus variants. Individual specificities of anti-SARS-CoV-2 Abs and their impact on infectivity by other variants have been little investigated to date. Here, we dissected at a monoclonal level neutralizing and enhancing Abs elicited by early variants and how they affect infectivity of emerging variants. B cells from 13 convalescent patients originally infected by D614G or Alpha variants were immortalized to isolate 445 naturally-produced anti-SARS-CoV-2 Abs. Monoclonal antibodies (mAbs) were tested for their abilities to impact the cytopathic effect of D614G, Delta, and Omicron (BA.1) variants. Ninety-eight exhibited robust neutralization against at least one of the three variant types, while 309 showed minimal or no impact on infectivity. Thirty-eight mAbs enhanced infectivity of SARS-CoV-2. Infection with D614G/Alpha variants generated variant-specific (65 neutralizing Abs, 35 enhancing Abs) and cross-reactive (18 neutralizing Abs, 3 enhancing Abs) mAbs. Interestingly, among the neutralizing mAbs with cross-reactivity restricted to two of the three variants tested, none demonstrated specific neutralization of the Delta and Omicron variants. In contrast, cross-reactive mAbs enhancing infectivity (n = 3) were found exclusively specific to Delta and Omicron variants. Notably, two mAbs that amplified in vitro the cytopathic effect of the Delta variant also exhibited neutralization against Omicron. These findings shed light on functional diversity of cross-reactive Abs generated during SARS-CoV-2 infection and illustrate how the balance between neutralizing and enhancing Abs facilitate variant emergence.
Collapse
Affiliation(s)
- Frédéric Coutant
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
- Immunology Department, Lyon-Sud Hospital, Hospices Civils of Lyon, Pierre-Bénite, France
| | - Franck Touret
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Jean-Jacques Pin
- Eurobio Scientific/Dendritics – Edouard Herriot Hospital, Lyon, France
| | - Marina Alonzo
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
| | - Cécile Baronti
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Sandie Munier
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Molecular Genetics of RNA Viruses Unit, Paris, France
| | - Mikaël Attia
- Institut Pasteur, Université Paris Cité, CNRS UMR3569, Molecular Genetics of RNA Viruses Unit, Paris, France
| | - Xavier de Lamballerie
- Unité des Virus Émergents (UVE: Aix-Marseille University - IRD 190 - Inserm 1207), Marseille, France
| | - Tristan Ferry
- Department of Infectious and Tropical Diseases, Hospices Civils of Lyon - Croix-Rousse Hospital, Lyon, France
- CIRI, Inserm U1111, CNRS, UMR5308, ENS Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Team, University of Lyon, Edouard Herriot Hospital, Lyon, France
- Department of Immunology and Rheumatology, Edouard Herriot Hospital, Lyon, France
| |
Collapse
|
2
|
Marchese AM, Fries L, Beyhaghi H, Vadivale M, Zhu M, Cloney-Clark S, Plested JS, Chung AW, Dunkle LM, Kalkeri R. Mechanisms and implications of IgG4 responses to SARS-CoV-2 and other repeatedly administered vaccines. J Infect 2024; 89:106317. [PMID: 39419185 DOI: 10.1016/j.jinf.2024.106317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Vaccine-induced immunoglobulin G (IgG) profiles can vary with respect to the predominant subclasses that characterize the response. Among IgG subclasses, IgG4 is reported to have anti-inflammatory properties, but can also exhibit reduced capacity for virus neutralization and activation of Fc-dependent effector functions. Here, we review evidence that IgG4 subclass responses can be disproportionately increased in response to some types of vaccines targeting an array of diseases, including pertussis, HIV, malaria, and COVID-19. The basis for enhanced IgG4 induction by vaccines is poorly understood but may be associated with platform- or dose regimen-specific differences in antigen exposure and/or cytokine stimulation. The clinical implications of vaccine-induced IgG4 responses remain uncertain, though collective evidence suggests that proportional increases in IgG4 might reduce vaccine antigen-specific immunity. Additional work is needed to determine underlying mechanisms and to elucidate what role IgG4 may play in modifications of vaccine-induced immunity to disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Amy W Chung
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Victoria 3000, Australia
| | | | | |
Collapse
|
3
|
Tong X, Wang Q, Jung W, Chicz TM, Blanc R, Parker LJ, Barouch DH, McNamara RP. Compartment-specific antibody correlates of protection to SARS-CoV-2 Omicron in macaques. iScience 2024; 27:110174. [PMID: 39224511 PMCID: PMC11367469 DOI: 10.1016/j.isci.2024.110174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 09/04/2024] Open
Abstract
Antibodies represent a primary mediator of protection against respiratory viruses. Serum neutralizing antibodies (NAbs) are often considered a primary correlate of protection. However, detailed antibody profiles including characterization of antibody functions in different anatomic compartments are poorly understood. Here we show that antibody correlates of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are different in systemic versus mucosal compartments in rhesus macaques. In serum, NAbs were the strongest correlate of protection and linked to spike-specific binding antibodies and other extra-NAb functions that create a larger protective network. In bronchiolar lavage (BAL), antibody-dependent cellular phagocytosis (ADCP) proved the strongest correlate of protection rather than NAbs. Within BAL, ADCP was linked to mucosal spike-specific immunoglobulin (Ig)G, IgA/secretory IgA, and Fcγ-receptor binding antibodies. Our results support a model in which antibodies with different functions mediate protection at different anatomic sites.
Collapse
Affiliation(s)
- Xin Tong
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Qixin Wang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Wonyeong Jung
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Taras M. Chicz
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Ross Blanc
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Lily J. Parker
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Dan H. Barouch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ryan P. McNamara
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
4
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 DOI: 10.1016/j.cell.2024.07.021] [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: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Izadi A, Godzwon M, Söderlund Strand A, Schmidt T, Kumlien Georén S, Drosten C, Ohlin M, Nordenfelt P. Protective Non-neutralizing anti-N-terminal Domain mAb Maintains Fc-mediated Function against SARS-COV-2 Variants up to BA.2.86-JN.1 with Superfluous In Vivo Protection against JN.1 Due to Attenuated Virulence. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:678-689. [PMID: 39018495 PMCID: PMC11335326 DOI: 10.4049/jimmunol.2300675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 06/25/2024] [Indexed: 07/19/2024]
Abstract
Substantial evidence supports that Fc-mediated effector functions of anti-spike Abs contribute to anti-SARS-Cov-2 protection. We have previously shown that two non-neutralizing but opsonic mAbs targeting the receptor-binding domain and N-terminal domain (NTD), Ab81 and Ab94, respectively, are protective against lethal Wuhan SARS-CoV-2 infection in K18-hACE2 mice. In this article, we investigated whether these protective non-neutralizing Abs maintain Fc-mediated function and Ag binding against mutated SARS-CoV-2 variants. Ab81 and Ab94 retained their nanomolar affinity and Fc-mediated function toward Omicron and its subvariants, such as BA.2, BA.4, BA.5, XBB, XBB1.5, and BQ1.1. However, when encountering the more heavily mutated BA.2.86, Ab81 lost its function, whereas the 10 new mutations in the NTD did not affect Ab94. In vivo experiments with Ab94 in K18-hACE2 mice inoculated with a stringent dose of 100,000 PFU of the JN.1 variant revealed unexpected results. Surprisingly, this variant exhibited low disease manifestation in this animal model with no weight loss or death in the control group. Still, assessment of mice using a clinical scoring system showed better protection for Ab94-treated mice, indicating that Fc-mediated functions are still beneficial. Our work shows that a protective anti-receptor-binding domain non-neutralizing mAb lost reactivity when BA.2.86 emerged, whereas the anti-NTD mAb was still functional. Finally, this work adds new insight into the evolution of the SARS-CoV-2 virus by reporting that JN.1 is substantially less virulent in vivo than previous strains.
Collapse
Affiliation(s)
- Arman Izadi
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Anna Söderlund Strand
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden
| | - Tobias Schmidt
- Department of Clinical Sciences Lund, Division of Pediatrics, Faculty of Medicine, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | | | - Christian Drosten
- German Center for Infection Research, Berlin, Germany
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Mats Ohlin
- Department of Immunotechnology, Lund University, Lund, Sweden
- SciLifeLab Drug Discovery and Development, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden
| |
Collapse
|
6
|
Jiang B, Elkashif A, Coulter JA, Dunne NJ, McCarthy HO. Immunotherapy for HPV negative head and neck squamous cell carcinoma. Biochim Biophys Acta Rev Cancer 2024; 1879:189138. [PMID: 38889878 DOI: 10.1016/j.bbcan.2024.189138] [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: 03/22/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Head and neck cancer (HNSCC) is the 8th most common cancer in the UK, with incidence increasing due to lifestyle factors such as tobacco and alcohol abuse. HNSCC is an immune-suppressive disease characterised by impaired cytokine secretion and dysregulation of immune infiltrate. As such, immunotherapy is a potential treatment option, with therapeutic cancer vaccination demonstrating the greatest potential. The success of cancer vaccination is dependent on informed antigen selection: an ideal antigen must be either tumour-specific or tumour-associated, as well as highly immunogenic. Stratification of the patient population for antigen expression and validated biomarkers are also vital. This review focuses on the latest developments in immunotherapy, specifically the development of therapeutic vaccines, and highlights successes, potential drawbacks and areas for future development. Immunotherapy approaches considered for HNSCC include monoclonal antibodies (mAb), Oncolytic viral (OV) therapies, Immune Checkpoint Inhibitors (ICIs) and cancer vaccines.
Collapse
Affiliation(s)
- Binyumeng Jiang
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Ahmed Elkashif
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Jonathan A Coulter
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, UK.
| |
Collapse
|
7
|
Izadi A, Nordenfelt P. Protective non-neutralizing SARS-CoV-2 monoclonal antibodies. Trends Immunol 2024; 45:609-624. [PMID: 39034185 DOI: 10.1016/j.it.2024.06.003] [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: 06/17/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/23/2024]
Abstract
Recent studies show an important role for non-neutralizing anti-spike antibodies, including monoclonal antibodies (mAbs), in robustly protecting against SARS-CoV-2 infection. These mAbs use Fc-mediated functions such as complement activation, phagocytosis, and cellular cytotoxicity. There is an untapped potential for using non-neutralizing mAbs in durable antibody treatments; because of their available conserved epitopes, they may not be as sensitive to virus mutations as neutralizing mAbs. Here, we discuss evidence of non-neutralizing mAb-mediated protection against SARS-CoV-2 infection. We explore how non-neutralizing mAb Fc-mediated functions can be enhanced via novel antibody-engineering techniques. Important questions remain to be answered regarding the characteristics of protective non-neutralizing mAbs, including the models and assays used for study, the risks of ensuing detrimental inflammation, as well as the durability and mechanisms of protection.
Collapse
Affiliation(s)
- Arman Izadi
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden; Karolinska University Hospital, Stockholm, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Division of Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden; Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden.
| |
Collapse
|
8
|
Richardson SI, Mzindle N, Motlou T, Manamela NP, van der Mescht MA, Lambson BE, Everatt J, Amoako DG, Balla S, von Gottberg A, Wolter N, de Beer Z, de Villiers TR, Bodenstein A, van den Berg G, Abdullah F, Rossouw TM, Boswell MT, Ueckermann V, Bhiman JN, Moore PL. SARS-CoV-2 BA.4/5 infection triggers more cross-reactive FcγRIIIa signaling and neutralization than BA.1, in the context of hybrid immunity. J Virol 2024; 98:e0067824. [PMID: 38953380 PMCID: PMC11265454 DOI: 10.1128/jvi.00678-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
SARS-CoV-2 variants of concern (VOCs) differentially trigger neutralizing and antibody-dependent cellular cytotoxic (ADCC) antibodies with variable cross-reactivity. Omicron BA.4/5 was approved for inclusion in bivalent vaccination boosters, and therefore the antigenic profile of antibodies elicited by this variant is critical to understand. Here, we investigate the ability of BA.4/5-elicited antibodies following the first documented (primary) infection (n = 13) or breakthrough infection after vaccination (n = 9) to mediate neutralization and FcγRIIIa signaling across multiple SARS-CoV-2 variants including XBB.1.5 and BQ.1. Using a pseudovirus neutralization assay and a FcγRIIIa crosslinking assay to measure ADCC potential, we show that unlike SARS-CoV-2 Omicron BA.1, BA.4/5 infection triggers highly cross-reactive functional antibodies. Cross-reactivity was observed both in the absence of prior vaccination and in breakthrough infections following vaccination. However, BQ.1 and XBB.1.5 neutralization and FcγRIIIa signaling were significantly compromised compared to other VOCs, regardless of prior vaccination status. BA.4/5 triggered FcγRIIIa signaling was significantly more resilient against VOCs (<10-fold decrease in magnitude) compared to neutralization (10- to 100-fold decrease). Overall, this study shows that BA.4/5 triggered antibodies are highly cross-reactive compared to those triggered by other variants. Although this is consistent with enhanced neutralization and FcγRIIIa signaling breadth of BA.4/5 vaccine boosters, the reduced activity against XBB.1.5 supports the need to update vaccines with XBB sublineage immunogens to provide adequate coverage of these highly antibody evasive variants. IMPORTANCE The continued evolution of SARS-CoV-2 has resulted in a number of variants of concern. Of these, the Omicron sublineage is the most immune evasive. Within Omicron, the BA.4/5 sublineage drove the fifth wave of infection in South Africa prior to becoming the dominant variant globally. As a result this spike sequence was approved as part of a bivalent vaccine booster, and rolled out worldwide. We aimed to understand the cross-reactivity of neutralizing and Fc mediated cytotoxic functions elicited by BA.4/5 infection following infection or breakthrough infection. We find that, in contrast to BA.1 which triggered fairly strain-specific antibodies, BA.4/5 triggered antibodies that are highly cross-reactive for neutralization and antibody-dependent cellular cytotoxicity potential. Despite this cross-reactivity, these antibodies are compromised against highly resistant variants such as XBB.1.5 and BQ.1. This suggests that next-generation vaccines will require XBB sublineage immunogens in order to protect against these evasive variants.
Collapse
Affiliation(s)
- Simone I. Richardson
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Nonkululeko Mzindle
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Thopisang Motlou
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Nelia P. Manamela
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Mieke A. van der Mescht
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Bronwen E. Lambson
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Josie Everatt
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
| | - Daniel Gyamfi Amoako
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- School of Health Sciences, College of Health Sciences, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
| | - Sashkia Balla
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Anne von Gottberg
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nicole Wolter
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | | | | | - Fareed Abdullah
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Theresa M. Rossouw
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Michael T. Boswell
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Veronica Ueckermann
- Division for Infectious Diseases, Department of Internal Medicine, Steve Biko Academic Hospital and University of Pretoria, Pretoria, South Africa
| | - Jinal N. Bhiman
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Penny L. Moore
- SAMRC Antibody Immunity Research Unit, School of Pathology, University of the Witwatersrand, Johannesburg, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
| |
Collapse
|
9
|
Davis SK, Jia F, Wright QG, Islam MT, Bean A, Layton D, Williams DT, Lynch SE. Defining correlates of protection for mammalian livestock vaccines against high-priority viral diseases. Front Immunol 2024; 15:1397780. [PMID: 39100679 PMCID: PMC11294087 DOI: 10.3389/fimmu.2024.1397780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/28/2024] [Indexed: 08/06/2024] Open
Abstract
Enhancing livestock biosecurity is critical to safeguard the livelihoods of farmers, global and local economies, and food security. Vaccination is fundamental to the control and prevention of exotic and endemic high-priority infectious livestock diseases. Successful implementation of vaccination in a biosecurity plan is underpinned by a strong understanding of correlates of protection-those elements of the immune response that can reliably predict the level of protection from viral challenge. While correlates of protection have been successfully characterized for many human viral vaccines, for many high-priority livestock viral diseases, including African swine fever and foot and mouth disease, they remain largely uncharacterized. Current literature provides insights into potential correlates of protection that should be assessed during vaccine development for these high-priority mammalian livestock viral diseases. Establishment of correlates of protection for biosecurity purposes enables immune surveillance, rationale for vaccine development, and successful implementation of livestock vaccines as part of a biosecurity strategy.
Collapse
Affiliation(s)
- Samantha K. Davis
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Australian Centre for Disease Preparedness, Geelong, VIC, Australia
| | | | | | | | | | | | | | | |
Collapse
|
10
|
McGee JE, Kirsch JR, Kenney D, Cerbo F, Chavez EC, Shih TY, Douam F, Wong WW, Grinstaff MW. Complete substitution with modified nucleotides in self-amplifying RNA suppresses the interferon response and increases potency. Nat Biotechnol 2024:10.1038/s41587-024-02306-z. [PMID: 38977924 DOI: 10.1038/s41587-024-02306-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 06/04/2024] [Indexed: 07/10/2024]
Abstract
The use of modified nucleotides to suppress the interferon response and maintain translation of self-amplifying RNA (saRNA), which has been achieved for mRNA, has not yet succeeded. We identify modified nucleotides that, when substituted at 100% in saRNA, confer innate immune evasion and robust long-term protein expression, and when formulated as a vaccine, protect against lethal SARS-CoV-2 challenge in mice. This discovery advances saRNA therapeutics by enabling prolonged protein expression at low doses.
Collapse
Affiliation(s)
- Joshua E McGee
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
- Biological Design Center, Boston University, Boston, MA, USA
| | - Jack R Kirsch
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Devin Kenney
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Faith Cerbo
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Elizabeth C Chavez
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA
| | - Ting-Yu Shih
- Department of Chemistry, Boston University, Boston, MA, USA
| | - Florian Douam
- Department of Virology, Immunology and Microbiology, Boston University School of Medicine, Boston, MA, USA.
- National Emerging Infectious Diseases Laboratories (NEIDL), Boston University, Boston, MA, USA.
| | - Wilson W Wong
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Biological Design Center, Boston University, Boston, MA, USA.
| | - Mark W Grinstaff
- Department of Biomedical Engineering, Boston University, Boston, MA, USA.
- Department of Chemistry, Boston University, Boston, MA, USA.
| |
Collapse
|
11
|
Motsoeneng BM, Bhiman JN, Richardson SI, Moore PL. SARS-CoV-2 humoral immunity in people living with HIV-1. Trends Immunol 2024; 45:511-522. [PMID: 38890026 DOI: 10.1016/j.it.2024.05.005] [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: 05/17/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024]
Abstract
The effect of COVID-19 on the high number of immunocompromised people living with HIV-1 (PLWH), particularly in Africa, remains a critical concern. Here, we identify key areas that still require further investigation, by examining COVID-19 vaccine effectiveness, and understanding antibody responses in SARS-CoV-2 infection and vaccination in comparison with people without HIV-1 (PWOH). We also assess the potential impact of pre-existing immunity against endemic human coronaviruses on SARS-CoV-2 responses in these individuals. Lastly, we discuss the consequences of persistent infection in PLWH (or other immunocompromised individuals), including prolonged shedding, increased viral diversity within the host, and the implications on SARS-CoV-2 evolution in Africa.
Collapse
Affiliation(s)
- Boitumelo M Motsoeneng
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Jinal N Bhiman
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Simone I Richardson
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa
| | - Penny L Moore
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases of the National Health Laboratory Services, Johannesburg, South Africa; Centre for the AIDS Program of Research in South Africa (CAPRISA), University of KwaZulu Natal, Durban, South Africa.
| |
Collapse
|
12
|
Jackson S, Marshall JL, Mawer A, Lopez-Ramon R, Harris SA, Satti I, Hughes E, Preston-Jones H, Cabrera Puig I, Longet S, Tipton T, Laidlaw S, Doherty RP, Morrison H, Mitchell R, Tanner R, Ateere A, Stylianou E, Wu MS, Fredsgaard-Jones TPW, Breuer J, Rapeport G, Ferreira VM, Gleeson F, Pollard AJ, Carroll M, Catchpole A, Chiu C, McShane H. Safety, tolerability, viral kinetics, and immune correlates of protection in healthy, seropositive UK adults inoculated with SARS-CoV-2: a single-centre, open-label, phase 1 controlled human infection study. THE LANCET. MICROBE 2024; 5:655-668. [PMID: 38703782 PMCID: PMC7616636 DOI: 10.1016/s2666-5247(24)00025-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 05/06/2024]
Abstract
BACKGROUND A SARS-CoV-2 controlled human infection model (CHIM) has been successfully established in seronegative individuals using a dose of 1×101 50% tissue culture infectious dose (TCID50) pre-alpha SARS-CoV-2 virus. Given the increasing prevalence of seropositivity to SARS-CoV-2, a CHIM that could be used for vaccine development will need to induce infection in those with pre-existing immunity. Our aim was to find a dose of pre-alpha SARS-CoV-2 virus that induced infection in previously infected individuals. METHODS Healthy, UK volunteers aged 18-30 years, with proven (quantitative RT-PCR or lateral flow antigen test) previous SARS-CoV-2 infection (with or without vaccination) were inoculated intranasally in a stepwise dose escalation CHIM with either 1×101, 1×102, 1×10³, 1×104, or 1×105 TCID50 SARS-CoV-2/human/GBR/484861/2020, the same virus used in the seronegative CHIM. Post-inoculation, volunteers were quarantined in functionally negative pressure rooms (Oxford, UK) for 14 days and until 12-hourly combined oropharyngeal-nasal swabs were negative for viable virus by focus-forming assay. Outpatient follow-up continued for 12 months post-enrolment, with additional visits for those who developed community-acquired SARS-CoV-2 infection. The primary objective was to identify a safe, well tolerated dose that induced infection (defined as two consecutive SARS-CoV-2 positive PCRs starting 24 h after inoculation) in 50% of seropositive volunteers. This study is registered with ClinicalTrials.gov (NCT04864548); enrolment and follow-up to 12 months post-enrolment are complete. FINDINGS Recruitment commenced on May 6, 2021, with the last volunteer enrolled into the dose escalation cohort on Nov 24, 2022. 36 volunteers were enrolled, with four to eight volunteers inoculated in each dosing group from 1×101 to 1×105 TCID50 SARS-CoV-2. All volunteers have completed quarantine, with follow-up to 12 months complete. Despite dose escalation to 1×105 TCID50, we were unable to induce sustained infection in any volunteers. Five (14%) of 36 volunteers were considered to have transient infection, based on the kinetic of their PCR-positive swabs. Transiently infected volunteers had significantly lower baseline mucosal and systemic SARS-CoV-2-specific antibody titres and significantly lower peripheral IFNγ responses against a CD8+ T-cell SARS-CoV-2 peptide pool than uninfected volunteers. 14 (39%) of 36 volunteers subsequently developed breakthrough infection with the omicron variant after discharge from quarantine. Most adverse events reported by volunteers in quarantine were mild, with fatigue (16 [44%]) and stuffy nose (16 [44%]) being the most common. There were no serious adverse events. INTERPRETATION Our study demonstrates potent protective immunity induced by homologous vaccination and homologous or heterologous previous SARS-CoV-2 infection. The community breakthrough infections seen with the omicron variant supports the use of newer variants to establish a model with sufficient rate of infection for use in vaccine and therapeutic development. FUNDING Wellcome Trust and Department for Health and Social Care.
Collapse
Affiliation(s)
- Susan Jackson
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Julia L Marshall
- Department of Paediatrics, University of Oxford, Oxford, UK; The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Andrew Mawer
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Iman Satti
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Eileen Hughes
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Stephanie Longet
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tom Tipton
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Stephen Laidlaw
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Hazel Morrison
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Rachel Tanner
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Alberta Ateere
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Meng-San Wu
- Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Judith Breuer
- Institute of Child Health, University College London, London, UK
| | - Garth Rapeport
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Fergus Gleeson
- Oxford Radiology Research Unit, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Miles Carroll
- The Wellcome Centre for Human Genetics and Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Helen McShane
- Department of Paediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Sánchez-Simarro Á, Fernández-Soto D, Grau B, Albert E, Giménez E, Avilés-Alía AI, Gozalbo-Rovira R, Rusu L, Olea B, Geller R, Reyburn HT, Navarro D. Functional antibody responses targeting the Spike protein of SARS-CoV-2 Omicron XBB.1.5 in elderly nursing home residents following Wuhan-Hu-1-based mRNA booster vaccination. Sci Rep 2024; 14:11896. [PMID: 38789475 PMCID: PMC11126592 DOI: 10.1038/s41598-024-62874-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/22/2024] [Indexed: 05/26/2024] Open
Abstract
The immune effector mechanisms involved in protecting against severe COVID-19 infection in elderly nursing home residents following vaccination or natural infection are not well understood. Here, we measured SARS-CoV-2 Spike (S)-directed functional antibody responses, including neutralizing antibodies (NtAb) and antibody Fc-mediated NK cell activity (degranulation and IFNγ production), against the Wuhan-Hu-1, BA.4/5 (for NtAb), and Omicron XBB.1.5 variants in elderly nursing home residents (n = 39; median age, 91 years) before and following a third (pre- and post-3D) and a fourth (pre- and post-4D) mRNA COVID-19 vaccine dose. Both 3D and 4D boosted NtAb levels against both (sub)variants. Likewise, 3D and 4D increased the ability of sera to trigger both LAMP1- and IFNγ-producing NK cells, in particular against XBB.1.5. In contrast to NtAb titres, the frequencies of LAMP1- and IFNγ-producing NK cells activated by antibodies binding to Wuhan-Hu-1 and Omicron XBB.1.5 S were comparable at all testing times. Stronger functional antibody responses were observed in vaccine-experienced participants compared to vaccine-naïve at some testing times. These findings can contribute to identifying a reliable correlate of protection in elderly nursing home residents against severe COVID-19 and inform future vaccine strategies in this population group.
Collapse
Grants
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- FIS, PI21/00563 Instituto de Salud Carlos III
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- 202020E079 y CSIC-COVID19-028 Fundación General CSIC
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
- PID2020-115506RB-I00 (HTR) Ministerio de Ciencia e Innovación
Collapse
Affiliation(s)
- Ángela Sánchez-Simarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Av. Blasco Ibáñez 17, 46010, Valencia, Spain
| | - Daniel Fernández-Soto
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - Brayan Grau
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, 46980, Valencia, Spain
| | - Eliseo Albert
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Av. Blasco Ibáñez 17, 46010, Valencia, Spain
| | - Estela Giménez
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Av. Blasco Ibáñez 17, 46010, Valencia, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Isabel Avilés-Alía
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, 46980, Valencia, Spain
| | | | - Luciana Rusu
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, 46980, Valencia, Spain
| | - Beatriz Olea
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Av. Blasco Ibáñez 17, 46010, Valencia, Spain
| | - Ron Geller
- Institute for Integrative Systems Biology (I2SysBio), Universitat de Valencia-CSIC, 46980, Valencia, Spain
| | - Hugh T Reyburn
- Department of Immunology and Oncology, National Centre for Biotechnology, CNB-CSIC, Madrid, Spain
| | - David Navarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Av. Blasco Ibáñez 17, 46010, Valencia, Spain.
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain.
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain.
| |
Collapse
|
14
|
Klingler J, Kowdle S, Bandres JC, Emami-Gorizi R, Alvarez RA, Rao PG, Amanat F, Gleason C, Kleiner G, Simon V, Edelstein A, Perandones C, Upadhyay C, Lee B, Hioe CE. Heterologous Ad26/Ad5 adenovirus-vectored vaccines elicited SARS-CoV-2-specific antibody responses with potent Fc activities. Front Immunol 2024; 15:1382619. [PMID: 38779671 PMCID: PMC11109367 DOI: 10.3389/fimmu.2024.1382619] [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: 02/05/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Introduction Antibodies against the SARS-CoV-2 spike protein are a critical immune determinant for protection against the virus. While virus neutralization is a key function of spike-specific antibodies, antibodies also mediate Fc-dependent activities that can play a role in protection or pathogenesis. Methods This study characterized serum antibody responses elicited after two doses of heterologous adenovirus-vectored (Ad26/ Ad5) vaccines. Results Vaccine-induced antibody binding titers and Fc-mediated functions decreased over six months, while neutralization titers remained stable. Comparison of antibody isotypes elicited after Ad26/Ad5 vs. LNP-mRNA vaccination and after infection showed that anti-spike IgG1 were dominant and produced to high levels in all groups. The Ad26/Ad5 vaccines also induced IgG4 but not IgG2 and IgG3, whereas the LNP-mRNA vaccines elicited a full Ig spectrum (IgM, IgG1-4, IgA1-2). Convalescent COVID-19 patients had mainly IgM and IgA1 alongside IgG1. Despite these differences, the neutralization potencies against early variants were similar. However, both vaccine groups had antibodies with greater Fc potencies of binding complement and Fcg receptors than the COVID-19 group. The Ad26/Ad5 group also displayed a greater potency of RBD-specific antibody-mediated cellular phagocytosis. Discussion Antibodies with distinctive quality were induced by different vaccines and infection. The data imply the utility of different vaccine platforms to elicit antibody responses with fine-tuned Fc activities.
Collapse
Affiliation(s)
- Jéromine Klingler
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Bronx, NY, United States
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | | | - Raymond A. Alvarez
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Priyanka G. Rao
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Charles Gleason
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Giulio Kleiner
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Viviana Simon
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Alexis Edelstein
- Administración Nacional de Laboratorios e Institutos de Salud (ANLIS) Dr. Carlos G. Malbrán, Buenos Aires, Argentina
| | - Claudia Perandones
- Administración Nacional de Laboratorios e Institutos de Salud (ANLIS) Dr. Carlos G. Malbrán, Buenos Aires, Argentina
| | - Chitra Upadhyay
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Catarina E. Hioe
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- James J. Peters VA Medical Center, Bronx, NY, United States
| |
Collapse
|
15
|
Gao R, Feng C, Sheng Z, Li F, Wang D. Research progress in Fc-effector functions against SARS-CoV-2. J Med Virol 2024; 96:e29638. [PMID: 38682662 DOI: 10.1002/jmv.29638] [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: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/18/2024] [Indexed: 05/01/2024]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has caused more than 676 million cases in the global human population with approximately 7 million deaths and vaccination has been proved as the most effective countermeasure in reducing clinical complications and mortality rate of SARS-CoV-2 infection in people. However, the protective elements and correlation of protection induced by vaccination are still not completely understood. Various antibodies with multiple protective mechanisms can be induced simultaneously by vaccination in vivo, thereby complicating the identification and characterization of individual correlate of protection. Recently, an increasing body of observations suggests that antibody-induced Fc-effector functions play a crucial role in combating SARS-CoV-2 infections, including neutralizing antibodies-escaping variants. Here, we review the recent progress in understanding the impact of Fc-effector functions in broadly disarming SARS-CoV-2 infectivity and discuss various efforts in harnessing this conserved antibody function to develop an effective SARS-CoV-2 vaccine that can protect humans against infections by SARS-CoV-2 virus and its variants of concern.
Collapse
Affiliation(s)
- Rongyuan Gao
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Chenchen Feng
- Department of Biology and Microbiology, South Dakota State University, Brookings, South Dakota, USA
| | - Zizhang Sheng
- Zuckerman Mind Brian Behavior Institute, Columbia University, New York, New York, USA
| | - Feng Li
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| | - Dan Wang
- Maxwell H. Gluck Equine Research Center, Department of Veterinary Science, University of Kentucky, Lexington, Kentucky, USA
| |
Collapse
|
16
|
Izadi A, Karami Y, Bratanis E, Wrighton S, Khakzad H, Nyblom M, Olofsson B, Happonen L, Tang D, Sundwall M, Godzwon M, Chao Y, Toledo AG, Schmidt T, Ohlin M, Nilges M, Malmström J, Bahnan W, Shannon O, Malmström L, Nordenfelt P. The hinge-engineered IgG1-IgG3 hybrid subclass IgGh 47 potently enhances Fc-mediated function of anti-streptococcal and SARS-CoV-2 antibodies. Nat Commun 2024; 15:3600. [PMID: 38678029 PMCID: PMC11055898 DOI: 10.1038/s41467-024-47928-8] [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: 06/30/2023] [Accepted: 04/15/2024] [Indexed: 04/29/2024] Open
Abstract
Streptococcus pyogenes can cause invasive disease with high mortality despite adequate antibiotic treatments. To address this unmet need, we have previously generated an opsonic IgG1 monoclonal antibody, Ab25, targeting the bacterial M protein. Here, we engineer the IgG2-4 subclasses of Ab25. Despite having reduced binding, the IgG3 version promotes stronger phagocytosis of bacteria. Using atomic simulations, we show that IgG3's Fc tail has extensive movement in 3D space due to its extended hinge region, possibly facilitating interactions with immune cells. We replaced the hinge of IgG1 with four different IgG3-hinge segment subclasses, IgGhxx. Hinge-engineering does not diminish binding as with IgG3 but enhances opsonic function, where a 47 amino acid hinge is comparable to IgG3 in function. IgGh47 shows improved protection against S. pyogenes in a systemic infection mouse model, suggesting that IgGh47 has promise as a preclinical therapeutic candidate. Importantly, the enhanced opsonic function of IgGh47 is generalizable to diverse S. pyogenes strains from clinical isolates. We generated IgGh47 versions of anti-SARS-CoV-2 mAbs to broaden the biological applicability, and these also exhibit strongly enhanced opsonic function compared to the IgG1 subclass. The improved function of the IgGh47 subclass in two distant biological systems provides new insights into antibody function.
Collapse
Affiliation(s)
- Arman Izadi
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Yasaman Karami
- Université de Lorraine, CNRS, Inria, LORIA, F-54000, Nancy, France
- Institut Pasteur, Université Paris cite, CNRS UMR3528, Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, F-75015, Paris, France
| | - Eleni Bratanis
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Sebastian Wrighton
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Hamed Khakzad
- Université de Lorraine, CNRS, Inria, LORIA, F-54000, Nancy, France
| | - Maria Nyblom
- Department of Biology & Lund Protein Production Platform (LP3), Lund University, Lund, Sweden
| | - Berit Olofsson
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Lotta Happonen
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Di Tang
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Martin Sundwall
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Magdalena Godzwon
- Department of Immunotechnology and SciLifeLab Drug Discovery and Development Platform, Lund University, Lund, Sweden
| | - Yashuan Chao
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Alejandro Gomez Toledo
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Tobias Schmidt
- Department of Clinical Sciences Lund, Division of Pediatrics, Faculty of Medicine, Lund University, Lund, Sweden
| | - Mats Ohlin
- Department of Immunotechnology and SciLifeLab Drug Discovery and Development Platform, Lund University, Lund, Sweden
| | - Michael Nilges
- Institut Pasteur, Université Paris cite, CNRS UMR3528, Structural Bioinformatics Unit, Department of Structural Biology and Chemistry, F-75015, Paris, France
| | - Johan Malmström
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Wael Bahnan
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Oonagh Shannon
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
- Section for Oral Biology and Pathology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Lars Malmström
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden
| | - Pontus Nordenfelt
- Department of Clinical Sciences Lund, Infection Medicine, Faculty of Medicine, Lund University, Lund, Sweden.
- Department of Laboratory Medicine, Clinical Microbiology, Skåne University Hospital Lund, Lund University, Lund, Sweden.
| |
Collapse
|
17
|
Tong X, Wang Q, Jung W, Chicz TM, Blanc R, Parker LJ, Barouch DH, McNamara RP. Compartment-Specific Antibody Correlates of Protection to SARS-CoV-2 Omicron in Macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582951. [PMID: 38464001 PMCID: PMC10925337 DOI: 10.1101/2024.03.01.582951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Antibodies represent a primary mediator of protection against respiratory viruses such as SARS-CoV-2. Serum neutralizing antibodies (NAbs) are often considered a primary correlate of protection. However, detailed antibody profiles including characterization of antibody functions in different anatomic compartments are not well understood. Here we show that antibody correlates of protection against SARS-CoV-2 challenge are different in systemic versus mucosal compartments in rhesus macaques. In serum, neutralizing antibodies were the strongest correlate of protection and were linked to Spike-specific binding antibodies and other extra-neutralizing antibody functions that create a larger protective network. In contrast, in bronchiolar lavage (BAL), antibody-dependent cellular phagocytosis (ADCP) proved the strongest correlate of protection rather than NAbs. Within BAL, ADCP was linked to mucosal Spike-specific IgG, IgA/secretory IgA, and Fcγ-receptor binding antibodies. Our results support a model in which antibodies with different functions mediate protection at different anatomic sites. The correlation of ADCP and other Fc functional antibody responses with protection in BAL suggests that these antibody responses may be critical for protection against SARS-CoV-2 Omicron challenge in mucosa.
Collapse
Affiliation(s)
- Xin Tong
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Qixin Wang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Wonyeong Jung
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Taras M. Chicz
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Ross Blanc
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Lily J. Parker
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Dan H. Barouch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ryan P. McNamara
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| |
Collapse
|
18
|
Woodruff MC, Faliti CE, Sanz I. Systems biology of B cells in COVID-19. Semin Immunol 2024; 72:101875. [PMID: 38489999 DOI: 10.1016/j.smim.2024.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/17/2024]
Abstract
The integration of multi-'omic datasets into complex systems-wide assessments has become a mainstay in immunologic investigation. This focus on high-dimensional data collection and analysis was on full display in the investigation of COVID-19, the respiratory illness resulting from infection by the novel coronavirus SARS-CoV-2. Particularly in the area of B cell biology, tremendous efforts in both cellular and serologic investigation have resulted in an increasingly detailed mapping of the coordinated effector, memory, and antibody secreting cell responses that underpin the development of humoral immunity in response to primary viral infection. Further, the rapid development and deployment of effective vaccines has allowed for the assessment of developing memory responses across a wide variety of immune contexts, including in patients with compromised immune function. The result has been a period of rapid gains in the understanding of B cell biology unrestricted to the study of COVID-19. Here, we outline the systems-level technologies that have been routinely implemented in these investigations throughout the pandemic, and discuss how their use has led to clear and applicable gains in pursuance of the amelioration of human infectious disease and beyond.
Collapse
Affiliation(s)
- Matthew C Woodruff
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Caterina E Faliti
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA.
| | - Ignacio Sanz
- Department of Medicine, Division of Rheumatology, Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA; Emory Autoimmunity Center of Excellence, Emory University, Atlanta, GA, USA
| |
Collapse
|
19
|
Lasrado N, Collier ARY, Miller J, Hachmann NP, Liu J, Anand T, A. Bondzie E, Fisher JL, Mazurek CR, Patio RC, Rodrigues SL, Rowe M, Surve N, Ty DM, Wu C, Chicz TM, Tong X, Korber B, McNamara RP, Barouch DH. Waning immunity and IgG4 responses following bivalent mRNA boosting. SCIENCE ADVANCES 2024; 10:eadj9945. [PMID: 38394195 PMCID: PMC10889350 DOI: 10.1126/sciadv.adj9945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Messenger RNA (mRNA) vaccines were highly effective against the ancestral SARS-CoV-2 strain, but the efficacy of bivalent mRNA boosters against XBB variants was substantially lower. Here, we show limited durability of neutralizing antibody (NAb) responses against XBB variants and isotype switching to immunoglobulin G4 (IgG4) responses following bivalent mRNA boosting. Bivalent mRNA boosting elicited modest XBB.1-, XBB.1.5-, and XBB.1.16-specific NAbs that waned rapidly within 3 months. In contrast, bivalent mRNA boosting induced more robust and sustained NAbs against the ancestral WA1/2020 strain, suggesting immune imprinting. Following bivalent mRNA boosting, serum antibody responses were primarily IgG2 and IgG4 responses with poor Fc functional activity. In contrast, a third monovalent mRNA immunization boosted all isotypes including IgG1 and IgG3 with robust Fc functional activity. These data show substantial immune imprinting for the ancestral spike and isotype switching to IgG4 responses following bivalent mRNA boosting, with important implications for future booster designs and boosting strategies.
Collapse
Affiliation(s)
- Ninaad Lasrado
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ai-ris Y. Collier
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Miller
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nicole P. Hachmann
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jinyan Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Trisha Anand
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Esther A. Bondzie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jana L. Fisher
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Camille R. Mazurek
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert C. Patio
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Marjorie Rowe
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nehalee Surve
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Darren M. Ty
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cindy Wu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Taras M. Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Xin Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Bette Korber
- Los Alamos National Laboratory and New Mexico Consortium, Los Alamos, NM, USA
| | | | - Dan H. Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
20
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging Maternal and Cord Antibody Functions From SARS-CoV-2 Infection and Vaccination in Pregnancy. J Infect Dis 2024; 229:462-472. [PMID: 37815524 PMCID: PMC10873180 DOI: 10.1093/infdis/jiad421] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/27/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023] Open
Abstract
Maternal immunity impacts the infant, but how is unclear. To understand the implications of the immune exposures of vaccination and infection in pregnancy for neonatal immunity, we evaluated antibody functions in paired peripheral maternal and cord blood. We compared those who in pregnancy received mRNA coronavirus disease 2019 (COVID-19) vaccine, were infected by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and the combination. We found that vaccination enriched a subset of neutralizing activities and Fc effector functions that was driven by IgG1 and was minimally impacted by antibody glycosylation in maternal blood. In paired cord blood, maternal vaccination also enhanced IgG1. However, Fc effector functions compared to neutralizing activities were preferentially transferred. Moreover, changes in IgG posttranslational glycosylation contributed more to cord than peripheral maternal blood antibody functional potency. These differences were enhanced with the combination of vaccination and infection as compared to either alone. Thus, Fc effector functions and antibody glycosylation highlight underexplored maternal opportunities to safeguard newborns.
Collapse
Affiliation(s)
- Emily H Adhikari
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health, Dallas Texas, USA
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ye Jin Kang
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ann R McDonald
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jessica E Pruszynski
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Timothy A Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Savannah K McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Fikadu G Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, Oregon, USA
| | - Lenette L Lu
- Parkland Health, Dallas Texas, USA
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Zimmerman O, Altman Doss AM, Ying B, Liang CY, Mackin SR, Davis-Adams HG, Adams LJ, VanBlargan LA, Chen RE, Scheaffer SM, Desai P, Raju S, Mantia TL, O’Shaughnessy CC, Monroy JM, Wedner HJ, Rigell CJ, Kau AL, Dy TB, Ren Z, Turner JS, O’Halloran JA, Presti RM, Kendall PL, Fremont DH, Ellebedy AH, Diamond MS. Immunoglobulin replacement products protect against SARS-CoV-2 infection in vivo despite poor neutralizing activity. JCI Insight 2024; 9:e176359. [PMID: 38175703 PMCID: PMC10967375 DOI: 10.1172/jci.insight.176359] [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: 10/03/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024] Open
Abstract
Immunoglobulin (IG) replacement products are used routinely in patients with immune deficiency and other immune dysregulation disorders who have poor responses to vaccination and require passive immunity conferred by commercial antibody products. The binding, neutralizing, and protective activity of intravenously administered IG against SARS-CoV-2 emerging variants remains unknown. Here, we tested 198 different IG products manufactured from December 2019 to August 2022. We show that prepandemic IG had no appreciable cross-reactivity or neutralizing activity against SARS-CoV-2. Anti-spike antibody titers and neutralizing activity against SARS-CoV-2 WA1/2020 D614G increased gradually after the pandemic started and reached levels comparable to vaccinated healthy donors 18 months after the diagnosis of the first COVID-19 case in the United States in January 2020. The average time between production to infusion of IG products was 8 months, which resulted in poor neutralization of the variant strain circulating at the time of infusion. Despite limited neutralizing activity, IG prophylaxis with clinically relevant dosing protected susceptible K18-hACE2-transgenic mice against clinical disease, lung infection, and lung inflammation caused by the XBB.1.5 Omicron variant. Moreover, following IG prophylaxis, levels of XBB.1.5 infection in the lung were higher in FcγR-KO mice than in WT mice. Thus, IG replacement products with poor neutralizing activity against evolving SARS-CoV-2 variants likely confer protection to patients with immune deficiency disorders through Fc effector function mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Andrew L. Kau
- Department of Medicine, and
- Department of Molecular Microbiology
- Center for Women’s Infectious Disease Research
| | | | | | | | | | - Rachel M. Presti
- Department of Medicine, and
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, and
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| | | | | | - Ali H. Ellebedy
- Department of Pathology and Immunology
- Department of Molecular Microbiology
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, and
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Michael S. Diamond
- Department of Medicine, and
- Department of Pathology and Immunology
- Department of Molecular Microbiology
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, and
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
22
|
Reinig S, Shih SR. Non-neutralizing functions in anti-SARS-CoV-2 IgG antibodies. Biomed J 2024; 47:100666. [PMID: 37778697 PMCID: PMC10825350 DOI: 10.1016/j.bj.2023.100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 08/31/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023] Open
Abstract
Most individuals infected with or vaccinated against COVID-19 develop antigenic neutralizing immunoglobulin G (IgG) antibodies against the SARS-CoV-2 spike protein. Although neutralizing antibodies are biomarkers of the adaptive immune response, their mere presence is insufficient to explain the protection afforded against the disease or its pathology. IgG exhibits other secondary effector functions that activate innate immune components, including complement, natural killer cells, and macrophages. The affinity for effector cells depends on the isotypes and glycosylation of IgG antibodies. The anti-spike IgG titer should be sufficient to provide significant Fc-mediated effects in severe COVID-19, mRNA, and protein subunit vaccinations. In combination with aberrant effector cells, pro-inflammatory afucosylated IgG1 and IgG3 may be detrimental in severe COVID-19. The antibody response of mRNA vaccines leads to higher fucosylation and a less inflammatory IgG profile, with a long-term shift to IgG4, which is correlated with protection from disease.
Collapse
Affiliation(s)
- Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| |
Collapse
|
23
|
Lapuente D, Winkler TH, Tenbusch M. B-cell and antibody responses to SARS-CoV-2: infection, vaccination, and hybrid immunity. Cell Mol Immunol 2024; 21:144-158. [PMID: 37945737 PMCID: PMC10805925 DOI: 10.1038/s41423-023-01095-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 prompted scientific, medical, and biotech communities to investigate infection- and vaccine-induced immune responses in the context of this pathogen. B-cell and antibody responses are at the center of these investigations, as neutralizing antibodies (nAbs) are an important correlate of protection (COP) from infection and the primary target of SARS-CoV-2 vaccine modalities. In addition to absolute levels, nAb longevity, neutralization breadth, immunoglobulin isotype and subtype composition, and presence at mucosal sites have become important topics for scientists and health policy makers. The recent pandemic was and still is a unique setting in which to study de novo and memory B-cell (MBC) and antibody responses in the dynamic interplay of infection- and vaccine-induced immunity. It also provided an opportunity to explore new vaccine platforms, such as mRNA or adenoviral vector vaccines, in unprecedented cohort sizes. Combined with the technological advances of recent years, this situation has provided detailed mechanistic insights into the development of B-cell and antibody responses but also revealed some unexpected findings. In this review, we summarize the key findings of the last 2.5 years regarding infection- and vaccine-induced B-cell immunity, which we believe are of significant value not only in the context of SARS-CoV-2 but also for future vaccination approaches in endemic and pandemic settings.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany.
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany
| |
Collapse
|
24
|
Nziza N, Deng Y, Wood L, Dhanoa N, Dulit-Greenberg N, Chen T, Kane AS, Swank Z, Davis JP, Demokritou M, Chitnis AP, Fasano A, Edlow AG, Jain N, Horwitz BH, McNamara RP, Walt DR, Lauffenburger DA, Julg B, Shreffler WG, Alter G, Yonker LM. Humoral profiles of toddlers and young children following SARS-CoV-2 mRNA vaccination. Nat Commun 2024; 15:905. [PMID: 38291080 PMCID: PMC10827750 DOI: 10.1038/s41467-024-45181-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
Although young children generally experience mild symptoms following infection with SARS-CoV-2, severe acute and long-term complications can occur. SARS-CoV-2 mRNA vaccines elicit robust immunoglobulin profiles in children ages 5 years and older, and in adults, corresponding with substantial protection against hospitalizations and severe disease. Whether similar immune responses and humoral protection can be observed in vaccinated infants and young children, who have a developing and vulnerable immune system, remains poorly understood. To study the impact of mRNA vaccination on the humoral immunity of infant, we use a system serology approach to comprehensively profile antibody responses in a cohort of children ages 6 months to 5 years who were vaccinated with the mRNA-1273 COVID-19 vaccine (25 μg). Responses are compared with vaccinated adults (100 μg), in addition to naturally infected toddlers and young children. Despite their lower vaccine dose, vaccinated toddlers elicit a functional antibody response as strong as adults, with higher antibody-dependent phagocytosis compared to adults, without report of side effects. Moreover, mRNA vaccination is associated with a higher IgG3-dependent humoral profile against SARS-CoV-2 compared to natural infection, supporting that mRNA vaccination is effective at eliciting a robust antibody response in toddlers and young children.
Collapse
Affiliation(s)
- Nadège Nziza
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yixiang Deng
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lianna Wood
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Boston Children's Hospital, Department of Pediatric Gastroenterology, Boston, MA, USA
| | - Navneet Dhanoa
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
| | | | - Tina Chen
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Abigail S Kane
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Zoe Swank
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Jameson P Davis
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Melina Demokritou
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
| | - Anagha P Chitnis
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
| | - Alessio Fasano
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Andrea G Edlow
- Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Boston, MA, USA
- Massachusetts General Hospital, Vincent Center for Reproductive Biology, Boston, MA, USA
| | - Nitya Jain
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Bruce H Horwitz
- Harvard Medical School, Boston, MA, USA
- Boston Children's Hospital, Department of Emergency Medicine, Boston, MA, USA
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - David R Walt
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Boris Julg
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Wayne G Shreffler
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Lael M Yonker
- Massachusetts General Hospital, Department of Pediatrics, Boston, MA, USA.
- Massachusetts General Hospital, Mucosal Immunology and Biology Research Center, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Lenart K, Arcoverde Cerveira R, Hellgren F, Ols S, Sheward DJ, Kim C, Cagigi A, Gagne M, Davis B, Germosen D, Roy V, Alter G, Letscher H, Van Wassenhove J, Gros W, Gallouët AS, Le Grand R, Kleanthous H, Guebre-Xabier M, Murrell B, Patel N, Glenn G, Smith G, Loré K. Three immunizations with Novavax's protein vaccines increase antibody breadth and provide durable protection from SARS-CoV-2. NPJ Vaccines 2024; 9:17. [PMID: 38245545 PMCID: PMC10799869 DOI: 10.1038/s41541-024-00806-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/08/2023] [Indexed: 01/22/2024] Open
Abstract
The immune responses to Novavax's licensed NVX-CoV2373 nanoparticle Spike protein vaccine against SARS-CoV-2 remain incompletely understood. Here, we show in rhesus macaques that immunization with Matrix-MTM adjuvanted vaccines predominantly elicits immune events in local tissues with little spillover to the periphery. A third dose of an updated vaccine based on the Gamma (P.1) variant 7 months after two immunizations with licensed NVX-CoV2373 resulted in significant enhancement of anti-spike antibody titers and antibody breadth including neutralization of forward drift Omicron variants. The third immunization expanded the Spike-specific memory B cell pool, induced significant somatic hypermutation, and increased serum antibody avidity, indicating considerable affinity maturation. Seven months after immunization, vaccinated animals controlled infection by either WA-1 or P.1 strain, mediated by rapid anamnestic antibody and T cell responses in the lungs. In conclusion, a third immunization with an adjuvanted, low-dose recombinant protein vaccine significantly improved the quality of B cell responses, enhanced antibody breadth, and provided durable protection against SARS-CoV-2 challenge.
Collapse
Affiliation(s)
- Klara Lenart
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrika Hellgren
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel J Sheward
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Changil Kim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Alberto Cagigi
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden
- Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Matthew Gagne
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Brandon Davis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Vicky Roy
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Hélène Letscher
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Jérôme Van Wassenhove
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Wesley Gros
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Anne-Sophie Gallouët
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, Paris, France
| | - Harry Kleanthous
- Bill & Melinda Gates Foundation, Seattle, WA, USA
- SK Biosciences, Boston, MA, USA
| | | | - Ben Murrell
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | - Karin Loré
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet, Stockholm, Sweden.
- Karolinska University Hospital, Stockholm, Sweden.
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
26
|
Cruz Cisneros MC, Anderson EJ, Hampton BK, Parotti B, Sarkar S, Taft-Benz S, Bell TA, Blanchard M, Dillard JA, Dinnon KH, Hock P, Leist SR, Madden EA, Shaw GD, West A, Baric RS, Baxter VK, Pardo-Manuel de Villena F, Heise MT, Ferris MT. Host Genetic Variation Impacts SARS-CoV-2 Vaccination Response in the Diversity Outbred Mouse Population. Vaccines (Basel) 2024; 12:103. [PMID: 38276675 PMCID: PMC10821422 DOI: 10.3390/vaccines12010103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
The COVID-19 pandemic led to the rapid and worldwide development of highly effective vaccines against SARS-CoV-2. However, there is significant individual-to-individual variation in vaccine efficacy due to factors including viral variants, host age, immune status, environmental and host genetic factors. Understanding those determinants driving this variation may inform the development of more broadly protective vaccine strategies. While host genetic factors are known to impact vaccine efficacy for respiratory pathogens such as influenza and tuberculosis, the impact of host genetic variation on vaccine efficacy against COVID-19 is not well understood. To model the impact of host genetic variation on SARS-CoV-2 vaccine efficacy, while controlling for the impact of non-genetic factors, we used the Diversity Outbred (DO) mouse model. We found that DO mice immunized against SARS-CoV-2 exhibited high levels of variation in vaccine-induced neutralizing antibody responses. While the majority of the vaccinated mice were protected from virus-induced disease, similar to human populations, we observed vaccine breakthrough in a subset of mice. Importantly, we found that this variation in neutralizing antibody, virus-induced disease, and viral titer is heritable, indicating that the DO serves as a useful model system for studying the contribution of genetic variation of both vaccines and disease outcomes.
Collapse
Affiliation(s)
- Marta C. Cruz Cisneros
- Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA; (M.C.C.C.); (B.K.H.)
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Elizabeth J. Anderson
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (E.J.A.); (V.K.B.)
| | - Brea K. Hampton
- Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA; (M.C.C.C.); (B.K.H.)
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Breantié Parotti
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Sharon Taft-Benz
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Timothy A. Bell
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Matthew Blanchard
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Jacob A. Dillard
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA; (J.A.D.); (E.A.M.); (R.S.B.)
| | - Kenneth H. Dinnon
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA; (J.A.D.); (E.A.M.); (R.S.B.)
| | - Pablo Hock
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Sarah R. Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.R.L.)
| | - Emily A. Madden
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA; (J.A.D.); (E.A.M.); (R.S.B.)
| | - Ginger D. Shaw
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.R.L.)
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA; (J.A.D.); (E.A.M.); (R.S.B.)
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (S.R.L.)
| | - Victoria K. Baxter
- Division of Comparative Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; (E.J.A.); (V.K.B.)
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
- Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | - Fernando Pardo-Manuel de Villena
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Mark T. Heise
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC 27599, USA; (J.A.D.); (E.A.M.); (R.S.B.)
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA; (B.P.); (S.S.); (S.T.-B.); (T.A.B.); (M.B.); (P.H.); (G.D.S.); (F.P.-M.d.V.); (M.T.H.)
| |
Collapse
|
27
|
Jung W, Yuan D, Kellman B, Gonzalez IGDS, Clemens R, Milan EP, Sprinz E, Cerbino Neto J, Smolenov I, Alter G, McNamara RP, Costa Clemens SA. Boosting with adjuvanted SCB-2019 elicits superior Fcγ-receptor engagement driven by IgG3 to SARS-CoV-2 spike. NPJ Vaccines 2024; 9:7. [PMID: 38182593 PMCID: PMC10770118 DOI: 10.1038/s41541-023-00791-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 12/08/2023] [Indexed: 01/07/2024] Open
Abstract
With the continued emergence of variants of concern, the global threat of COVID-19 persists, particularly in low- and middle-income countries with limited vaccine access. Protein-based vaccines, such as SCB-2019, can be produced on a large scale at a low cost while antigen design and adjuvant use can modulate efficacy and safety. While effective humoral immunity against SARS-CoV-2 variants has been shown to depend on both neutralization and Fc-mediated immunity, data on the effectiveness of protein-based vaccines with enhanced Fc-mediated immunity is limited. Here, we assess the humoral profile, including antibody isotypes, subclasses, and Fc receptor binding generated by a boosting with a recombinant trimer-tag protein vaccine SCB-2019. Individuals who were primed with 2 doses of the ChAdOx1 vaccine were equally divided into 4 groups and boosted with following formulations: Group 1: 9 μg SCB-2019 and Alhydrogel; Group 2: 9 μg SCB-2019, CpG 1018, and Alhydrogel; Group 3: 30 μg SCB-2019, CpG 1018, and Alhydrogel; Group 4: ChAdOx1. Group 3 showed enhanced antibody FcγR binding against wild-type and variants compared to Groups 1 and 2, showing a dose-dependent enhancement of immunity conferred by the SCB-2019 vaccine. Moreover, from day 15 after vaccination, Group 3 exhibited higher IgG3 and FcγR binding across variants of concerns, including Omicron and its subvariants, compared to the ChAdOx1-boosted individuals. Overall, this highlights the potential of SCB-2019 as a cost-efficient boosting regimen effective across variants of concerns.
Collapse
Affiliation(s)
- Wonyeong Jung
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | | | - Ralf Clemens
- International Vaccine Institute, Seoul, Republic of Korea
| | - Eveline Pipolo Milan
- Centro de Estudos e Pesquisa em Moléstias Infecciosas Ltda. (CEPCLIN), Natal, Brazil
| | - Eduardo Sprinz
- Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - José Cerbino Neto
- D'Or Institute for Research and Education (IDOR), Rio de Janeiro, Brazil
| | | | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| | - Sue Ann Costa Clemens
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
- Siena University, Siena, Italy
| |
Collapse
|
28
|
Capuano C, De Federicis D, Ciuti D, Turriziani O, Angeloni A, Anastasi E, Giannini G, Belardinilli F, Molfetta R, Alvaro D, Palmieri G, Galandrini R. Impact of SARS-CoV-2 vaccination on FcγRIIIA/CD16 dynamics in Natural Killer cells: relevance for antibody-dependent functions. Front Immunol 2023; 14:1285203. [PMID: 38045702 PMCID: PMC10693335 DOI: 10.3389/fimmu.2023.1285203] [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: 08/29/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Natural Killer (NK) cells contribute to the protective effects of vaccine-induced antibodies thanks to the low affinity receptor for IgG, FcγRIIIA/CD16, whose aggregation leads to the killing of infected cells and IFNγ release, through which they potentiate adaptive immune responses. Methods Forty-seven healthy young individuals undergoing either homologous (ChAdOx1-S/ChAdOx1-S) or heterologous (ChAdOx1-S/BNT162B2) SARS-CoV-2 vaccination settings were recruited. Peripheral blood samples were collected immediately prior to vaccination and 8 weeks after the booster dose. The phenotypic and functional profile of NK cells was evaluated by flow cytometry at both time points. Serum samples were tested to evaluate circulating anti-Spike IgG levels and cytomegalovirus serostatus. CD16 F158V polymorphism was assessed by sequencing analysis. Results The downregulation of CD16 and the selective impairment of antibody-dependent cytotoxicity and IFNγ production in CD56dim NK population, persisting 8 weeks after boosting, were observed in heterologous, but not in homologous SARS-CoV-2 vaccination scheme. While the magnitude of CD16-dependent functions of the global CD56dim pool correlated with receptor levels before and after vaccination, the responsivity of NKG2C+ subset, that displays amplified size and functionality in HCMV+ individuals, resulted intrinsically insensitive to CD16 levels. Individual CD16 responsiveness was also affected by CD16F158V polymorphism; F/F low affinity individuals, characterized by reduced CD16 levels and functions independently of vaccination, did not show post-vaccinal functional impairment with respect to intermediate and high affinity ones, despite a comparable CD16 downregulation. Further, CD16 high affinity ligation conditions by means of afucosylated mAb overcame vaccine-induced and genotype-dependent functional defects. Finally, the preservation of CD16 expression directly correlated with anti-Spike IgG titer, hinting that the individual magnitude of receptor-dependent functions may contribute to the amplification of the vaccinal response. Conclusion This study demonstrates a durable downmodulation of CD16 levels and Ab-dependent NK functions after SARS-CoV-2 heterologous vaccination, and highlights the impact of genetic and environmental host-related factors in modulating NK cell susceptibility to post-vaccinal Fc-dependent functional impairment.
Collapse
Affiliation(s)
- Cristina Capuano
- Departmental Faculty of Medicine and Surgery, UniCamillus-Saint Camillus International University of Health and Medical Sciences, Rome, Italy
| | - Davide De Federicis
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniel Ciuti
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Emanuela Anastasi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Rosa Molfetta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Domenico Alvaro
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Gabriella Palmieri
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
29
|
Routhu NK, Stampfer SD, Lai L, Akhtar A, Tong X, Yuan D, Chicz TM, McNamara RP, Jakkala K, Davis-Gardner ME, St Pierre EL, Smith B, Green KM, Golden N, Picou B, Jean SM, Wood J, Cohen J, Moore IN, Patel N, Guebre-Xabier M, Smith G, Glenn G, Kozlowski PA, Alter G, Ahmed R, Suthar MS, Amara RR. Efficacy of mRNA-1273 and Novavax ancestral or BA.1 spike booster vaccines against SARS-CoV-2 BA.5 infection in nonhuman primates. Sci Immunol 2023; 8:eadg7015. [PMID: 37191508 PMCID: PMC10451060 DOI: 10.1126/sciimmunol.adg7015] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Omicron SARS-CoV-2 variants escape vaccine-induced neutralizing antibodies and cause nearly all current COVID-19 cases. Here, we compared the efficacy of three booster vaccines against Omicron BA.5 challenge in rhesus macaques: mRNA-1273, the Novavax ancestral spike protein vaccine (NVX-CoV2373), or Omicron BA.1 spike protein version (NVX-CoV2515). All three booster vaccines induced a strong BA.1 cross-reactive binding antibody and changed immunoglobulin G (Ig) dominance from IgG1 to IgG4 in the serum. All three booster vaccines also induced strong and comparable neutralizing antibody responses against multiple variants of concern, including BA.5 and BQ.1.1, along with long-lived plasma cells in the bone marrow. The ratio of BA.1 to WA-1 spike-specific antibody-secreting cells in the blood was higher in NVX-CoV2515 animals compared with NVX-CoV2373 animals, suggesting a better recall of BA.1-specific memory B cells by the BA.1 spike-specific vaccine compared with the ancestral spike-specific vaccine. Further, all three booster vaccines induced low levels of spike-specific CD4 but not CD8 T cell responses in the blood. After challenge with SARS-CoV-2 BA.5 variant, all three vaccines showed strong protection in the lungs and controlled virus replication in the nasopharynx. In addition, both Novavax vaccines blunted viral replication in nasopharynx at day 2. The protection against SARS-CoV-2 BA.5 infection in the upper respiratory airways correlated with binding, neutralizing, and ADNP activities of the serum antibody. These data have important implications for COVID-19 vaccine development, because vaccines that lower nasopharyngeal virus may help to reduce transmission.
Collapse
Affiliation(s)
- Nanda Kishore Routhu
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Samuel David Stampfer
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lilin Lai
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Akil Akhtar
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Xin Tong
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Dansu Yuan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Taras M. Chicz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Ryan P. McNamara
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Kishor Jakkala
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Meredith E. Davis-Gardner
- Emory Vaccine Center, Division of Microbiology and Immunology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA; Department of Pediatrics, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | - Brandon Smith
- Tulane National Primate Research Center, Covington, LA, USA
| | | | - Nadia Golden
- Tulane National Primate Research Center, Covington, LA, USA
| | - Breanna Picou
- Tulane National Primate Research Center, Covington, LA, USA
| | - Sherrie M. Jean
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Jennifer Wood
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Joyce Cohen
- Division of Animal Resources, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Psychiatry and Behavioral Sciences, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Ian N. Moore
- Division of Pathology, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Nita Patel
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | | | - Gale Smith
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | - Greg Glenn
- Novavax, Inc., 21 Firstfield Road, Gaithersburg, MD 20878, USA
| | - Pamela A. Kozlowski
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | - Mehul S. Suthar
- Department of Pediatrics, Division of Infectious Diseases Vaccine Center, Emory Vaccine Center, Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329
| | - Rama Rao Amara
- Division of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
- Department of Microbiology and Immunology, Emory School of Medicine, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
30
|
Chen D, Li X, Hao X, Qiu Y, Song Y, Sun H, Liu Y, Du J, Zhang Y, Xiao F, Song C, Yan Y, Song R, Wang X, Zhao X, Jin R. Reduced neutralization and Fc effector function to Omicron subvariants in sera from SARS-CoV-1 survivors after two doses of CoronaVac plus one dose subunit vaccine. J Med Virol 2023; 95:e29136. [PMID: 37804496 DOI: 10.1002/jmv.29136] [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: 06/13/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron harbors more than 30 mutations of the spike protein and exhibits substantial immune evasion. Although previous study indicated that BNT162b2 messenger RNA vaccine induces potent cross-clade pan-sarbecovirus neutralizing antibodies in survivors of the infection by SARS-CoV-1, the neutralization activity and Fc-mediated effector functions of these cross-reactive antibodies elicited in SARS-CoV-1 survivors to Omicron subvariants still remain largely unknown. In this study, the neutralization activity and Fc-mediated effector functions of antibodies boosted by a third dose vaccination were characterized in SARS-CoV-1 convalescents and healthy individuals. Potent cross-clade broadly neutralizing antibodies were observed in SARS-CoV-1 survivors who received a three-dose vaccination regimen consisting of two priming doses of CoronaVac followed by one booster dose of the protein subunit vaccine ZF2001. However, the induced antibodies exhibited both reduced neutralization and impaired Fc effector functions targeting multiple Omicron subvariants. Importantly, the data also support the notion that immune imprints resulted from SARS-CoV-1 infection may exacerbate the impairment of neutralization activity and Fc-mediated effector functions to Omicron subvariants and provided invaluable information to vaccination strategy in future.
Collapse
Affiliation(s)
- Danying Chen
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Xinglin Li
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Xiaohua Hao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yaruo Qiu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- Peking University Ditan Teaching Hospital, Beijing, China
| | - Yanjun Song
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Hui Sun
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Yongmei Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Juan Du
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Yuanyuan Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Fan Xiao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Chuan Song
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Yonghong Yan
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Rui Song
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Xuesen Zhao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Ronghua Jin
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Infectious Diseases, Beijing, China
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| |
Collapse
|
31
|
Selva KJ, Ramanathan P, Haycroft ER, Reynaldi A, Cromer D, Tan CW, Wang LF, Wines BD, Hogarth PM, Downie LE, Davis SK, Purcell RA, Kent HE, Juno JA, Wheatley AK, Davenport MP, Kent SJ, Chung AW. Preexisting immunity restricts mucosal antibody recognition of SARS-CoV-2 and Fc profiles during breakthrough infections. JCI Insight 2023; 8:e172470. [PMID: 37737263 PMCID: PMC10561726 DOI: 10.1172/jci.insight.172470] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/15/2023] [Indexed: 09/23/2023] Open
Abstract
Understanding mucosal antibody responses from SARS-CoV-2 infection and/or vaccination is crucial to develop strategies for longer term immunity, especially against emerging viral variants. We profiled serial paired mucosal and plasma antibodies from COVID-19 vaccinated only vaccinees (vaccinated, uninfected), COVID-19-recovered vaccinees (recovered, vaccinated), and individuals with breakthrough Delta or Omicron BA.2 infections (vaccinated, infected). Saliva from COVID-19-recovered vaccinees displayed improved antibody-neutralizing activity, Fcγ receptor (FcγR) engagement, and IgA levels compared with COVID-19-uninfected vaccinees. Furthermore, repeated mRNA vaccination boosted SARS-CoV-2-specific IgG2 and IgG4 responses in both mucosa biofluids (saliva and tears) and plasma; however, these rises only negatively correlated with FcγR engagement in plasma. IgG and FcγR engagement, but not IgA, responses to breakthrough COVID-19 variants were dampened and narrowed by increased preexisting vaccine-induced immunity against the ancestral strain. Salivary antibodies delayed initiation following breakthrough COVID-19 infection, especially Omicron BA.2, but rose rapidly thereafter. Importantly, salivary antibody FcγR engagements were enhanced following breakthrough infections. Our data highlight how preexisting immunity shapes mucosal SARS-CoV-2-specific antibody responses and has implications for long-term protection from COVID-19.
Collapse
Affiliation(s)
- Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Pradhipa Ramanathan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Ebene R. Haycroft
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Singhealth Duke-NUS Global Health Institute, Singapore
| | - Bruce D. Wines
- Immune Therapies Laboratory, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - P. Mark Hogarth
- Immune Therapies Laboratory, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Laura E. Downie
- Department of Optometry and Vision Sciences, University of Melbourne, Carlton, Victoria, Australia
| | - Samantha K. Davis
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Helen E. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Kensington, New South Wales, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Sexual Health Centre and Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
32
|
Quirk GE, Schoenle MV, Peyton KL, Uhrlaub JL, Lau B, Burgess JL, Ellingson K, Beitel S, Romine J, Lutrick K, Fowlkes A, Britton A, Tyner HL, Caban-Martinez AJ, Naleway A, Gaglani M, Yoon S, Edwards L, Olsho L, Dake M, LaFleur BJ, Nikolich JŽ, Sprissler R, Worobey M, Bhattacharya D. Determinants of de novo B cell responses to drifted epitopes in post-vaccination SARS-CoV-2 infections. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.09.12.23295384. [PMID: 37745498 PMCID: PMC10516057 DOI: 10.1101/2023.09.12.23295384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Vaccine-induced immunity may impact subsequent de novo responses to drifted epitopes in SARS-CoV-2 variants, but this has been difficult to quantify due to the challenges in recruiting unvaccinated control groups whose first exposure to SARS-CoV-2 is a primary infection. Through local, statewide, and national SARS-CoV-2 testing programs, we were able to recruit cohorts of individuals who had recovered from either primary or post-vaccination infections by either the Delta or Omicron BA.1 variants. Regardless of variant, we observed greater Spike-specific and neutralizing antibody responses in post-vaccination infections than in those who were infected without prior vaccination. Through analysis of variant-specific memory B cells as markers of de novo responses, we observed that Delta and Omicron BA.1 infections led to a marked shift in immunodominance in which some drifted epitopes elicited minimal responses, even in primary infections. Prior immunity through vaccination had a small negative impact on these de novo responses, but this did not correlate with cross-reactive memory B cells, arguing against competitive inhibition of naïve B cells. We conclude that dampened de novo B cell responses against drifted epitopes are mostly a function of altered immunodominance hierarchies that are apparent even in primary infections, with a more modest contribution from pre-existing immunity, perhaps due to accelerated antigen clearance.
Collapse
Affiliation(s)
- Grace E Quirk
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
| | - Marta V Schoenle
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Kameron L Peyton
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Jennifer L Uhrlaub
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Branden Lau
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Jefferey L Burgess
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - Katherine Ellingson
- Department of Epidemiology and Biostatistics, Zuckerman College of Public Health, University of Arizona, Tucson
| | - Shawn Beitel
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - James Romine
- Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Arizona, USA
| | - Karen Lutrick
- College of Medicine-Tucson, University of Arizona, Tucson, Arizona, USA
| | - Ashley Fowlkes
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA
| | - Amadea Britton
- National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA
| | - Harmony L Tyner
- St. Luke's Regional Health Care System, Duluth, Minnesota, USA
| | | | - Allison Naleway
- Kaiser Permanente Northwest Center for Health Research, Portland, Oregon, USA
| | - Manjusha Gaglani
- Baylor Scott & White Health and Texas A&M University College of Medicine, Temple, Texas, USA
| | - Sarang Yoon
- Rocky Mountain Center for Occupational and Environmental Health, Department of Family and Preventive Medicine, University of Utah Health, Salt Lake City, Utah, USA
| | | | | | - Michael Dake
- Office of the Senior Vice-President for Health Sciences, University of Arizona, Tucson, AZ, USA
| | | | - Janko Ž Nikolich
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- University of Arizona Center on Aging, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Ryan Sprissler
- University of Arizona Genomics Core and the Arizona Research Labs, University of Arizona Genetics Core, University of Arizona, Tucson, AZ, USA
| | - Michael Worobey
- Department of Ecology and Evolutionary Biology, University of Arizona, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
| | - Deepta Bhattacharya
- Department of Immunobiology, University of Arizona College of Medicine, Tucson, AZ, USA
- BIO5 Institute, University of Arizona, Tucson, AZ, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
33
|
Li B, Jiang AY, Raji I, Atyeo C, Raimondo TM, Gordon AGR, Rhym LH, Samad T, MacIsaac C, Witten J, Mughal H, Chicz TM, Xu Y, McNamara RP, Bhatia S, Alter G, Langer R, Anderson DG. Enhancing the immunogenicity of lipid-nanoparticle mRNA vaccines by adjuvanting the ionizable lipid and the mRNA. Nat Biomed Eng 2023:10.1038/s41551-023-01082-6. [PMID: 37679571 DOI: 10.1038/s41551-023-01082-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 07/27/2023] [Indexed: 09/09/2023]
Abstract
To elicit optimal immune responses, messenger RNA vaccines require intracellular delivery of the mRNA and the careful use of adjuvants. Here we report a multiply adjuvanted mRNA vaccine consisting of lipid nanoparticles encapsulating an mRNA-encoded antigen, optimized for efficient mRNA delivery and for the enhanced activation of innate and adaptive responses. We optimized the vaccine by screening a library of 480 biodegradable ionizable lipids with headgroups adjuvanted with cyclic amines and by adjuvanting the mRNA-encoded antigen by fusing it with a natural adjuvant derived from the C3 complement protein. In mice, intramuscular or intranasal administration of nanoparticles with the lead ionizable lipid and with mRNA encoding for the fusion protein (either the spike protein or the receptor-binding domain of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)) increased the titres of antibodies against SARS-CoV-2 tenfold with respect to the vaccine encoding for the unadjuvanted antigen. Multiply adjuvanted mRNA vaccines may improve the efficacy, safety and ease of administration of mRNA-based immunization.
Collapse
Affiliation(s)
- Bowen Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Allen Yujie Jiang
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Idris Raji
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Theresa M Raimondo
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Akiva G R Gordon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Luke H Rhym
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tahoura Samad
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob Witten
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Haseeb Mughal
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Taras M Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Yue Xu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Sangeeta Bhatia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Wyss Institute at Harvard, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Harvard-MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
34
|
Hederman AP, Natarajan H, Heyndrickx L, Ariën KK, Wiener JA, Wright PF, Bloch EM, Tobian AAR, Redd AD, Blankson JN, Rottenstreich A, Zarbiv G, Wolf D, Goetghebuer T, Marchant A, Ackerman ME. SARS-CoV-2 vaccination elicits broad and potent antibody effector functions to variants of concern in vulnerable populations. Nat Commun 2023; 14:5171. [PMID: 37620337 PMCID: PMC10449910 DOI: 10.1038/s41467-023-40960-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
SARS-CoV-2 variants have continuously emerged in the face of effective vaccines. Reduced neutralization against variants raises questions as to whether other antibody functions are similarly compromised, or if they might compensate for lost neutralization activity. Here, the breadth and potency of antibody recognition and effector function is surveyed following either infection or vaccination. Considering pregnant women as a model cohort with higher risk of severe illness and death, we observe similar binding and functional breadth for healthy and immunologically vulnerable populations, but considerably greater functional antibody breadth and potency across variants associated with vaccination. In contrast, greater antibody functional activity targeting the endemic coronavirus OC43 is noted among convalescent individuals, illustrating a dichotomy in recognition between close and distant human coronavirus strains associated with exposure history. This analysis of antibody functions suggests the differential potential for antibody effector functions to contribute to protecting vaccinated and convalescent subjects as novel variants continue to evolve.
Collapse
Affiliation(s)
| | - Harini Natarajan
- Department of Immunology and Microbiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Leo Heyndrickx
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joshua A Wiener
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joel N Blankson
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amihai Rottenstreich
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gila Zarbiv
- Clinical Virology Unit, Hadassah University Medical Center, Jerusalem, Israel
| | - Dana Wolf
- Clinical Virology Unit, Hadassah University Medical Center, Jerusalem, Israel
| | - Tessa Goetghebuer
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
- Pediatric Department, CHU St Pierre, Brussels, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
- Department of Immunology and Microbiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
35
|
Azarias Da Silva M, Nioche P, Soudaramourty C, Bull-Maurer A, Tiouajni M, Kong D, Zghidi-Abouzid O, Picard M, Mendes-Frias A, Santa-Cruz A, Carvalho A, Capela C, Pedrosa J, Castro AG, Loubet P, Sotto A, Muller L, Lefrant JY, Roger C, Claret PG, Duvnjak S, Tran TA, Tokunaga K, Silvestre R, Corbeau P, Mammano F, Estaquier J. Repetitive mRNA vaccination is required to improve the quality of broad-spectrum anti-SARS-CoV-2 antibodies in the absence of CXCL13. SCIENCE ADVANCES 2023; 9:eadg2122. [PMID: 37540749 PMCID: PMC10403221 DOI: 10.1126/sciadv.adg2122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 07/05/2023] [Indexed: 08/06/2023]
Abstract
Since the initial spread of severe acute respiratory syndrome coronavirus 2 infection, several viral variants have emerged and represent a major challenge for immune control, particularly in the context of vaccination. We evaluated the quantity, quality, and persistence of immunoglobulin G (IgG) and IgA in individuals who received two or three doses of messenger RNA (mRNA) vaccines, compared with previously infected vaccinated individuals. We show that three doses of mRNA vaccine were required to match the humoral responses of preinfected vaccinees. Given the importance of antibody-dependent cell-mediated immunity against viral infections, we also measured the capacity of IgG to recognize spike variants expressed on the cell surface and found that cross-reactivity was also strongly improved by repeated vaccination. Last, we report low levels of CXCL13, a surrogate marker of germinal center activation and formation, in vaccinees both after two and three doses compared with preinfected individuals, providing a potential explanation for the short duration and low quality of Ig induced.
Collapse
Affiliation(s)
| | - Pierre Nioche
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | | | | | - Mounira Tiouajni
- INSERM-U1124, Université Paris Cité, Paris, France
- Structural and Molecular Analysis Platform, BioMedTech Facilities INSERM US36-CNRS UMS2009, Université Paris Cité, Paris, France
| | - Dechuan Kong
- Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, Japan
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | | | | | - Ana Mendes-Frias
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - André Santa-Cruz
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Alexandre Carvalho
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Carlos Capela
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
- Department of Internal Medicine, Hospital of Braga, Braga, Portugal
| | - Jorge Pedrosa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - António Gil Castro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Paul Loubet
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Albert Sotto
- Service des Maladies Infectieuses et Tropicales, CHU de Nîmes, Nîmes, France
| | - Laurent Muller
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Claire Roger
- Service de Réanimation Chirugicale, CHU de Nîmes, Nîmes, France
| | | | - Sandra Duvnjak
- Service de Gérontologie et Prévention du Vieillissement, CHU de Nîmes, Nîmes, France
| | - Tu-Anh Tran
- Service de Pédiatrie, CHU de Nîmes, Nîmes, France
| | - Kenzo Tokunaga
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ricardo Silvestre
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Pierre Corbeau
- Institut de Génétique Humaine, UMR9002 CNRS-Université de Montpellier, Montpellier, France
- Laboratoire d’Immunologie, CHU de Nîmes, Nîmes, France
| | - Fabrizio Mammano
- INSERM-U1124, Université Paris Cité, Paris, France
- Université de Tours, INSERM, UMR1259 MAVIVH, Tours, France
| | - Jérôme Estaquier
- INSERM-U1124, Université Paris Cité, Paris, France
- CHU de Québec-Université Laval Research Center, Québec City, Québec, Canada
| |
Collapse
|
36
|
Kaplonek P, Cizmeci D, Kwatra G, Izu A, Lee JSL, Bertera HL, Fischinger S, Mann C, Amanat F, Wang W, Koen AL, Fairlie L, Cutland CL, Ahmed K, Dheda K, Barnabas SL, Bhorat QE, Briner C, Krammer F, Saphire EO, Gilbert SC, Lambe T, Pollard AJ, Nunes M, Wuhrer M, Lauffenburger DA, Madhi SA, Alter G. ChAdOx1 nCoV-19 (AZD1222) vaccine-induced Fc receptor binding tracks with differential susceptibility to COVID-19. Nat Immunol 2023; 24:1161-1172. [PMID: 37322179 PMCID: PMC10307634 DOI: 10.1038/s41590-023-01513-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 04/12/2023] [Indexed: 06/17/2023]
Abstract
Despite the success of COVID-19 vaccines, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern have emerged that can cause breakthrough infections. Although protection against severe disease has been largely preserved, the immunological mediators of protection in humans remain undefined. We performed a substudy on the ChAdOx1 nCoV-19 (AZD1222) vaccinees enrolled in a South African clinical trial. At peak immunogenicity, before infection, no differences were observed in immunoglobulin (Ig)G1-binding antibody titers; however, the vaccine induced different Fc-receptor-binding antibodies across groups. Vaccinees who resisted COVID-19 exclusively mounted FcγR3B-binding antibodies. In contrast, enhanced IgA and IgG3, linked to enriched FcγR2B binding, was observed in individuals who experienced breakthrough. Antibodies unable to bind to FcγR3B led to immune complex clearance and resulted in inflammatory cascades. Differential antibody binding to FcγR3B was linked to Fc-glycosylation differences in SARS-CoV-2-specific antibodies. These data potentially point to specific FcγR3B-mediated antibody functional profiles as critical markers of immunity against COVID-19.
Collapse
Affiliation(s)
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Gaurav Kwatra
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Alane Izu
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Harry L Bertera
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Colin Mann
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Wenjun Wang
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Anthonet L Koen
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Johannesburg, South Africa
| | - Lee Fairlie
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Wits Reproductive Health and HIV Institute, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Clare L Cutland
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | - Keertan Dheda
- Division of Pulmonology, Groote Schuur Hospital and the University of Cape Town, Cape Town, South Africa
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, London, UK
| | - Shaun L Barnabas
- Family Centre for Research With Ubuntu, Department of Paediatrics, University of Stellenbosch, Cape Town, South Africa
| | | | - Carmen Briner
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erica Ollman Saphire
- Center for Infectious Disease and Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Sarah C Gilbert
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, and the NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Marta Nunes
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Manfred Wuhrer
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
- Department of Science and Innovation/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases Unit, University of the Witwatersrand, Johannesburg, South Africa.
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
37
|
Masson LC, Servian CDP, Jardim VH, Dos Anjos D, Dorta ML, Batalha-Carvalho JV, Moro AM, Romão PRT, Souza M, Fiaccadori FS, Fonseca SG. Heterologous Booster with BNT162b2 Induced High Specific Antibody Levels in CoronaVac Vaccinees. Vaccines (Basel) 2023; 11:1183. [PMID: 37514999 PMCID: PMC10383528 DOI: 10.3390/vaccines11071183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Immune responses after COVID-19 vaccination should be evaluated in different populations around the world. This study compared antibody responses induced by ChAdOx1 nCoV-19, CoronaVac, and BNT162b2 vaccines. Blood samples from vaccinees were collected pre- and post-vaccinations with the second and third doses. The study enrolled 78 vaccinees, of whom 62.8% were women, with the following median ages: 26 years-ChAdOx1 nCoV-19; 40 years-CoronaVac; 30 years-BNT162b2. Serum samples were quantified for anti-RBD IgG and anti-RBD IgA and anti-spike IgG by ELISA. After two vaccine doses, BNT162b2 vaccinees produced higher levels of anti-RBD IgA and IgG, and anti-spike IgG compared to ChAdOx1 nCoV-19 and CoronaVac vaccinees. The third dose booster with BNT162b2 induced higher levels of anti-RBD IgA and IgG, and anti-spike IgG in CoronaVac vaccinees. Individuals who reported a SARS-CoV-2 infection before or during the study had higher anti-RBD IgA and IgG production. In conclusion, two doses of the studied vaccines induced detectable levels of anti-RBD IgA and IgG and anti-spike IgG in vaccinees. The heterologous booster with BNT162b2 increased anti-RBD IgA and IgG and anti-spike IgG levels in CoronaVac vaccinees and anti-RBD IgA levels in ChAdOx1 nCoV-19 vaccinees. Furthermore, SARS-CoV-2 infection induced higher anti-RBD IgA and IgG levels in CoronaVac vaccinees.
Collapse
Affiliation(s)
- Letícia Carrijo Masson
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Carolina do Prado Servian
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Vitor Hugo Jardim
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Déborah Dos Anjos
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Miriam Leandro Dorta
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | | | - Ana Maria Moro
- Laboratório de Biofármacos, Instituto Butantan, São Paulo 05503-900, SP, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| | - Pedro Roosevelt Torres Romão
- Laboratório de Imunologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| | - Menira Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Fabiola Souza Fiaccadori
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Simone Gonçalves Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| |
Collapse
|
38
|
Purcell RA, Theisen RM, Arnold KB, Chung AW, Selva KJ. Polyfunctional antibodies: a path towards precision vaccines for vulnerable populations. Front Immunol 2023; 14:1183727. [PMID: 37600816 PMCID: PMC10433199 DOI: 10.3389/fimmu.2023.1183727] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/30/2023] [Indexed: 08/22/2023] Open
Abstract
Vaccine efficacy determined within the controlled environment of a clinical trial is usually substantially greater than real-world vaccine effectiveness. Typically, this results from reduced protection of immunologically vulnerable populations, such as children, elderly individuals and people with chronic comorbidities. Consequently, these high-risk groups are frequently recommended tailored immunisation schedules to boost responses. In addition, diverse groups of healthy adults may also be variably protected by the same vaccine regimen. Current population-based vaccination strategies that consider basic clinical parameters offer a glimpse into what may be achievable if more nuanced aspects of the immune response are considered in vaccine design. To date, vaccine development has been largely empirical. However, next-generation approaches require more rational strategies. We foresee a generation of precision vaccines that consider the mechanistic basis of vaccine response variations associated with both immunogenetic and baseline health differences. Recent efforts have highlighted the importance of balanced and diverse extra-neutralising antibody functions for vaccine-induced protection. However, in immunologically vulnerable populations, significant modulation of polyfunctional antibody responses that mediate both neutralisation and effector functions has been observed. Here, we review the current understanding of key genetic and inflammatory modulators of antibody polyfunctionality that affect vaccination outcomes and consider how this knowledge may be harnessed to tailor vaccine design for improved public health.
Collapse
Affiliation(s)
- Ruth A. Purcell
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Robert M. Theisen
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Kelly B. Arnold
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States
| | - Amy W. Chung
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
39
|
Morrison H, Jackson S, McShane H. Controlled human infection models in COVID-19 and tuberculosis: current progress and future challenges. Front Immunol 2023; 14:1211388. [PMID: 37304270 PMCID: PMC10248465 DOI: 10.3389/fimmu.2023.1211388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Controlled Human Infection Models (CHIMs) involve deliberately exposing healthy human volunteers to a known pathogen, to allow the detailed study of disease processes and evaluate methods of treatment and prevention, including next generation vaccines. CHIMs are in development for both tuberculosis (TB) and Covid-19, but challenges remain in their ongoing optimisation and refinement. It would be unethical to deliberately infect humans with virulent Mycobacteria tuberculosis (M.tb), however surrogate models involving other mycobacteria, M.tb Purified Protein Derivative or genetically modified forms of M.tb either exist or are under development. These utilise varying routes of administration, including via aerosol, per bronchoscope or intradermal injection, each with their own advantages and disadvantages. Intranasal CHIMs with SARS-CoV-2 were developed against the backdrop of the evolving Covid-19 pandemic and are currently being utilised to both assess viral kinetics, interrogate the local and systemic immunological responses post exposure, and identify immune correlates of protection. In future it is hoped they can be used to assess new treatments and vaccines. The changing face of the pandemic, including the emergence of new virus variants and increasing levels of vaccination and natural immunity within populations, has provided a unique and complex environment within which to develop a SARS-CoV-2 CHIM. This article will discuss current progress and potential future developments in CHIMs for these two globally significant pathogens.
Collapse
|
40
|
Dewald F, Pirkl M, Paluschinski M, Kühn J, Elsner C, Schulte B, Knüfer J, Ahmadov E, Schlotz M, Oral G, Bernhard M, Michael M, Luxenburger M, Andrée M, Hennies MT, Hafezi W, Müller MM, Kümpers P, Risse J, Kill C, Manegold RK, von Frantzki U, Richter E, Emmert D, Monzon-Posadas WO, Gräff I, Kogej M, Büning A, Baum M, Teipel F, Mochtarzadeh B, Wolff M, Gruell H, Di Cristanziano V, Burst V, Streeck H, Dittmer U, Ludwig S, Timm J, Klein F. Impaired humoral immunity to BQ.1.1 in convalescent and vaccinated patients. Nat Commun 2023; 14:2835. [PMID: 37208323 DOI: 10.1038/s41467-023-38127-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 04/18/2023] [Indexed: 05/21/2023] Open
Abstract
Determining SARS-CoV-2 immunity is critical to assess COVID-19 risk and the need for prevention and mitigation strategies. We measured SARS-CoV-2 Spike/Nucleocapsid seroprevalence and serum neutralizing activity against Wu01, BA.4/5 and BQ.1.1 in a convenience sample of 1,411 patients receiving medical treatment in the emergency departments of five university hospitals in North Rhine-Westphalia, Germany, in August/September 2022. 62% reported underlying medical conditions and 67.7% were vaccinated according to German COVID-19 vaccination recommendations (13.9% fully vaccinated, 54.3% one booster, 23.4% two boosters). We detected Spike-IgG in 95.6%, Nucleocapsid-IgG in 24.0%, and neutralization against Wu01, BA.4/5 and BQ.1.1 in 94.4%, 85.0%, and 73.8% of participants, respectively. Neutralization against BA.4/5 and BQ.1.1 was 5.6- and 23.4-fold lower compared to Wu01. Accuracy of S-IgG detection for determination of neutralizing activity against BQ.1.1 was reduced substantially. We explored previous vaccinations and infections as correlates of BQ.1.1 neutralization using multivariable and Bayesian network analyses. Given a rather moderate adherence to COVID-19 vaccination recommendations, this analysis highlights the need to improve vaccine-uptake to reduce the COVID-19 risk of immune evasive variants. The study was registered as clinical trial (DRKS00029414).
Collapse
Affiliation(s)
- Felix Dewald
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Martin Pirkl
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Martha Paluschinski
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Joachim Kühn
- Institute of Virology, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Carina Elsner
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
| | - Bianca Schulte
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, 38124, Braunschweig, Germany
| | - Jacqueline Knüfer
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Elvin Ahmadov
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Maike Schlotz
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Göksu Oral
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Michael Bernhard
- Emergency Department, Medical Faculty and University Hospital of Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Mark Michael
- Emergency Department, Medical Faculty and University Hospital of Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Maura Luxenburger
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Marcel Andrée
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Marc Tim Hennies
- Institute of Virology, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Wali Hafezi
- Institute of Virology, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Marlin Maybrit Müller
- Institute of Virology, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Philipp Kümpers
- Division of General Internal and Emergency Medicine, Nephrology, Hypertension and Rheumatology, Department of Medicine D, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Joachim Risse
- Center of Emergency Medicine, University Hospital Essen, 45147, Essen, Germany
| | - Clemens Kill
- Center of Emergency Medicine, University Hospital Essen, 45147, Essen, Germany
| | | | - Ute von Frantzki
- Center of Emergency Medicine, University Hospital Essen, 45147, Essen, Germany
| | - Enrico Richter
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, 38124, Braunschweig, Germany
| | - Dorian Emmert
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | | | - Ingo Gräff
- Emergency Department, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Monika Kogej
- Emergency Department, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Antonia Büning
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Maximilian Baum
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
| | - Finn Teipel
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Babak Mochtarzadeh
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Martin Wolff
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Henning Gruell
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Veronica Di Cristanziano
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Volker Burst
- Department II of Internal Medicine: Nephrology, Rheumatology, Diabetes and General Internal Medicine, Faculty of Medicine and University Hospital Cologne University of Cologne, 50931, Cologne, Germany
- Emergency Department, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Hendrik Streeck
- Institute of Virology, University Hospital Bonn, University of Bonn, 53127, Bonn, Germany
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, 38124, Braunschweig, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University Duisburg-Essen, 45141, Essen, Germany
| | - Stephan Ludwig
- Institute of Virology, Faculty of Medicine and University Hospital Muenster, University of Muenster, 48149, Muenster, Germany
| | - Jörg Timm
- Institute of Virology, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Florian Klein
- Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
- German Center for Infection Research (DZIF), Partner site Bonn-Cologne, 38124, Braunschweig, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
41
|
Kaplonek P, Deng Y, Shih-Lu Lee J, Zar HJ, Zavadska D, Johnson M, Lauffenburger DA, Goldblatt D, Alter G. Hybrid immunity expands the functional humoral footprint of both mRNA and vector-based SARS-CoV-2 vaccines. Cell Rep Med 2023; 4:101048. [PMID: 37182520 PMCID: PMC10126214 DOI: 10.1016/j.xcrm.2023.101048] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 12/13/2022] [Accepted: 04/20/2023] [Indexed: 05/16/2023]
Abstract
Despite the successes of current coronavirus disease 2019 (COVID-19) vaccines, waning immunity, the emergence of variants of concern, and breakthrough infections among vaccinees have begun to highlight opportunities to improve vaccine platforms. Real-world vaccine efficacy studies have highlighted the reduced risk of breakthrough infections and diseases among individuals infected and vaccinated, referred to as hybrid immunity. Thus, we sought to define whether hybrid immunity shapes the humoral immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) following Pfizer/BNT162b2, Moderna mRNA-1273, ChadOx1/AZD1222, and Ad26.COV2.S vaccination. Each vaccine exhibits a unique functional humoral profile in vaccination only or hybrid immunity. However, hybrid immunity shows a unique augmentation of S2-domain-specific functional immunity that was poorly induced for the vaccination only. These data highlight the importance of natural infection in breaking the immunodominance away from the evolutionarily unstable S1 domain and potentially affording enhanced cross-variant protection by targeting the more highly conserved S2 domain of SARS-CoV-2.
Collapse
Affiliation(s)
- Paulina Kaplonek
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA
| | - Yixiang Deng
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Heather J Zar
- Department of Pediatrics and Child Health, Red Cross War Memorial Children's Hospital, University of Cape Town, Cape Town, South Africa; SA MRC Unit on Child and Adolescent Health, University of Cape Town, Cape Town, South Africa
| | - Dace Zavadska
- Children's Clinical University Hospital, Riga, Latvia
| | - Marina Johnson
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK
| | - Douglas A Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David Goldblatt
- Great Ormond Street Institute of Child Health Biomedical Research Centre, University College London, London, UK.
| | - Galit Alter
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
42
|
Tawinprai K, Jungsomsri P, Pinijnai O, Tavonvunchai F, Lievjaroen A, Suwannaroj P, Siripongboonsitti T, Porntharukchareon T, Sornsamdang G, Ungtrakul T. Immunogenicity and reactogenicity of heterologous prime-boost vaccination with inactivated COVID-19 and ChAdOx1 nCoV-19 (AZD1222) vaccines, a quasi-experimental study. Hum Vaccin Immunother 2023:2206360. [PMID: 37140889 DOI: 10.1080/21645515.2023.2206360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
The global supply of COVID-19 vaccines has been limited, and concerns have arisen about vaccine supply chain disruptions in developing countries. Heterologous prime-boost vaccination, which involves using different vaccines for the first and second doses, has been proposed to enhance the immune response. We aimed to compare the immunogenicity and safety of a heterologous prime-boost vaccination using an inactivated COVID-19 vaccine and AZD1222 vaccine with that of a homologous vaccination using AZD1222. This pilot involved 164 healthy volunteers without prior SARS-CoV-2 infection aged 18 years or older assigned to receive either the heterologous or homologous vaccination. The results showed that the heterologous approach was safe and well-tolerated, although the reactogenicity of the heterologous approach was higher. At 4 weeks after receiving the booster dose, the heterologous approach elicited a non-inferior immune response compared to the homologous approach in neutralizing antibody and cell-mediated immune response. The percentage of inhibition was 83.88 (79.72-88.03) in the heterologous and 79.88 (75.50-84.25) in the homologous group, a mean difference of 4.60 (-1.67-10.88). The geometric mean of interferon-gamma was 1072.53 mIU/mL (799.29-1439.18) in the heterologous group and 867.67 mIU/mL (671.94-1120.40) in the homologous group, a GMR of 1.24 (0.82-1.85). However, the binding antibody test of the heterologous group was inferior to the homologous group. Our findings suggest that the use of heterologous prime-boost vaccination with different types of COVID-19 vaccines is a viable strategy, especially in settings where vaccine supply is limited or where vaccine distribution is challenging.
Collapse
Affiliation(s)
- Kriangkrai Tawinprai
- Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Pawornrath Jungsomsri
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Onnicha Pinijnai
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Fahsiri Tavonvunchai
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Anchisa Lievjaroen
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Paphada Suwannaroj
- Department of General Practice and Family Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Taweegrit Siripongboonsitti
- Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Thachanun Porntharukchareon
- Department of Medicine, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Gaidganok Sornsamdang
- Central Laboratory Center, Chulabhorn Hospital, Chulabhorn Royal Academy, Bangkok, Thailand
| | - Teerapat Ungtrakul
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, Thailand
| |
Collapse
|
43
|
Adhikari EH, Lu P, Kang YJ, McDonald AR, Pruszynski JE, Bates TA, McBride SK, Trank-Greene M, Tafesse FG, Lu LL. Diverging maternal and infant cord antibody functions from SARS-CoV-2 infection and vaccination in pregnancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538955. [PMID: 37205338 PMCID: PMC10187183 DOI: 10.1101/2023.05.01.538955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Immunization in pregnancy is a critical tool that can be leveraged to protect the infant with an immature immune system but how vaccine-induced antibodies transfer to the placenta and protect the maternal-fetal dyad remains unclear. Here, we compare matched maternal-infant cord blood from individuals who in pregnancy received mRNA COVID-19 vaccine, were infected by SARS-CoV-2, or had the combination of these two immune exposures. We find that some but not all antibody neutralizing activities and Fc effector functions are enriched with vaccination compared to infection. Preferential transport to the fetus of Fc functions and not neutralization is observed. Immunization compared to infection enriches IgG1-mediated antibody functions with changes in antibody post-translational sialylation and fucosylation that impact fetal more than maternal antibody functional potency. Thus, vaccine enhanced antibody functional magnitude, potency and breadth in the fetus are driven more by antibody glycosylation and Fc effector functions compared to maternal responses, highlighting prenatal opportunities to safeguard newborns as SARS-CoV-2 becomes endemic.
Collapse
Affiliation(s)
- Emily H. Adhikari
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
- Parkland Health, Dallas TX
| | - Pei Lu
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ye jin Kang
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Ann R. McDonald
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
| | - Jessica E. Pruszynski
- Division of Maternal-Fetal Medicine and Department of Obstetrics and Gynecology, UTSW Medical Center, Dallas, TX
| | - Timothy A. Bates
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Savannah K. McBride
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Mila Trank-Greene
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Fikadu G. Tafesse
- Department of Microbiology and Immunology, Oregon Health and Science University, Portland, OR
| | - Lenette L. Lu
- Parkland Health, Dallas TX
- Division of Infectious Diseases and Geographic Medicine and Department of Internal Medicine, UTSW Medical Center, Dallas, TX
- Department of Immunology, UTSW Medical Center, Dallas, TX
| |
Collapse
|
44
|
Adams LE, Leist SR, Dinnon KH, West A, Gully KL, Anderson EJ, Loome JF, Madden EA, Powers JM, Schäfer A, Sarkar S, Castillo IN, Maron JS, McNamara RP, Bertera HL, Zweigert MR, Higgins JS, Hampton BK, Premkumar L, Alter G, Montgomery SA, Baxter VK, Heise MT, Baric RS. Fc-mediated pan-sarbecovirus protection after alphavirus vector vaccination. Cell Rep 2023; 42:112326. [PMID: 37000623 PMCID: PMC10063157 DOI: 10.1016/j.celrep.2023.112326] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/21/2022] [Accepted: 03/17/2023] [Indexed: 04/01/2023] Open
Abstract
Group 2B β-coronaviruses (sarbecoviruses) have caused regional and global epidemics in modern history. Here, we evaluate the mechanisms of cross-sarbecovirus protective immunity, currently less clear yet important for pan-sarbecovirus vaccine development, using a panel of alphavirus-vectored vaccines covering bat to human strains. While vaccination does not prevent virus replication, it protects against lethal heterologous disease outcomes in both severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and clade 2 bat sarbecovirus challenge models. The spike vaccines tested primarily elicit a highly S1-specific homologous neutralizing antibody response with no detectable cross-virus neutralization. Rather, non-neutralizing antibody functions, mechanistically linked to FcgR4 and spike S2, mediate cross-protection in wild-type mice. Protection is lost in FcR knockout mice, further supporting a model for non-neutralizing, protective antibodies. These data highlight the importance of FcR-mediated cross-protective immune responses in universal pan-sarbecovirus vaccine designs.
Collapse
Affiliation(s)
- Lily E Adams
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah R Leist
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kenneth H Dinnon
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ande West
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kendra L Gully
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth J Anderson
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer F Loome
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Emily A Madden
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sanjay Sarkar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Izabella N Castillo
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jenny S Maron
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, USA
| | - Ryan P McNamara
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, USA
| | - Harry L Bertera
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, USA
| | - Mark R Zweigert
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jaclyn S Higgins
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brea K Hampton
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard University, Cambridge, MA, USA
| | - Stephanie A Montgomery
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Dallas Tissue Research, Dallas, TX, USA
| | - Victoria K Baxter
- Division of Comparative Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark T Heise
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Ralph S Baric
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Rapidly Emerging Antiviral Drug Discovery Initiative, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
45
|
Mackin SR, Desai P, Whitener BM, Karl CE, Liu M, Baric RS, Edwards DK, Chicz TM, McNamara RP, Alter G, Diamond MS. Fc-γR-dependent antibody effector functions are required for vaccine-mediated protection against antigen-shifted variants of SARS-CoV-2. Nat Microbiol 2023; 8:569-580. [PMID: 37012355 PMCID: PMC10797606 DOI: 10.1038/s41564-023-01359-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/10/2023] [Indexed: 04/05/2023]
Abstract
Emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants with antigenic changes in the spike protein are neutralized less efficiently by serum antibodies elicited by legacy vaccines against the ancestral Wuhan-1 virus. Nonetheless, these vaccines, including mRNA-1273 and BNT162b2, retained their ability to protect against severe disease and death, suggesting that other aspects of immunity control infection in the lung. Vaccine-elicited antibodies can bind Fc gamma receptors (FcγRs) and mediate effector functions against SARS-CoV-2 variants, and this property correlates with improved clinical coronavirus disease 2019 outcome. However, a causal relationship between Fc effector functions and vaccine-mediated protection against infection has not been established. Here, using passive and active immunization approaches in wild-type and FcγR-knockout mice, we determined the requirement for Fc effector functions to control SARS-CoV-2 infection. The antiviral activity of passively transferred immune serum was lost against multiple SARS-CoV-2 strains in mice lacking expression of activating FcγRs, especially murine FcγR III (CD16), or depleted of alveolar macrophages. After immunization with the pre-clinical mRNA-1273 vaccine, control of Omicron BA.5 infection in the respiratory tract also was lost in mice lacking FcγR III. Our passive and active immunization studies in mice suggest that Fc-FcγR engagement and alveolar macrophages are required for vaccine-induced antibody-mediated protection against infection by antigenically changed SARS-CoV-2 variants, including Omicron strains.
Collapse
Affiliation(s)
- Samantha R Mackin
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bradley M Whitener
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Courtney E Karl
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Meizi Liu
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ralph S Baric
- Department of Epidemiology, University of North Carolina, Chapel Hill, NC, USA
| | | | - Taras M Chicz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | | | - Galit Alter
- Moderna, Inc., Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
46
|
Cotter CA, Americo JL, Earl PL, Moss B. Protection from SARS-CoV-2 Variants by MVAs expressing matched or mismatched S administered intranasally to mice. NPJ Vaccines 2023; 8:47. [PMID: 36973267 PMCID: PMC10040904 DOI: 10.1038/s41541-023-00645-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
SARS-CoV-2 vaccines prevent severe disease but are less efficient in averting infection and transmission of variant strains, making it imperative to explore ways of enhancing protection. Use of inbred mice expressing the human SARS-CoV-2 receptor facilitates such investigations. We employed recombinant MVAs (rMVAs) expressing modified S of several SARS-CoV-2 strains and compared their ability to neutralize variants, bind S proteins and protect K18-hACE2 mice against SARS-CoV-2 challenge when administered intramuscularly or intranasally. The rMVAs expressing Wuhan, Beta and Delta S induced substantial cross neutralizing activities to each other but very low neutralization of Omicron; while rMVA expressing Omicon S induced neutralizing antibody predominanly to Omicron. In mice primed and boosted with rMVA expressing the Wuhan S, neutralizing antibodies to Wuhan increased after one immunization with rMVA expressing Omicron S due to original antigenic sin, but substantial neutralizing antibody to Omicron required a second immunization. Nevertheless, monovalent vaccines with S mismatched to the challenge virus still protected against severe disease and reduced the amounts of virus and subgenomic RNAs in the lungs and nasal turbinates, though not as well as vaccines with matched S. Passive transfer of Wuhan immune serum with Omicron S binding but undetectable neutralizing activity reduced infection of the l-ungs by Omicron suggesting additional effector functions. Notably, there was less infectious virus and viral subgenomic RNAs in the nasal turbinates and lungs when the rMVAs were administered intranasally rather than intramuscularly and this held true for vaccines that were matched or mismatched to the challenge strain of SARS-CoV-2.
Collapse
Affiliation(s)
- Catherine A Cotter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey L Americo
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Patricia L Earl
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bernard Moss
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Voysey M, Flaxman A, Aboagye J, Aley PK, Belij-Rammerstorfer S, Bibi S, Bittaye M, Cappuccini F, Charlton S, Clutterbuck EA, Davies S, Dold C, Edwards NJ, Ewer KJ, Faust SN, Folegatti PM, Fowler J, Gilbride C, Gilbert SC, Godfrey L, Hallis B, Humphries HE, Jenkin D, Kerridge S, Mujadidi YF, Plested E, Ramasamy MN, Robinson H, Sanders H, Snape MD, Song R, Thomas KM, Ulaszewska M, Woods D, Wright D, Pollard AJ, Lambe T. Persistence of the immune response after two doses of ChAdOx1 nCov-19 (AZD1222): 1 year of follow-up of two randomized controlled trials. Clin Exp Immunol 2023; 211:280-287. [PMID: 36729167 PMCID: PMC10038323 DOI: 10.1093/cei/uxad013] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/04/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
The trajectory of immune responses following the primary dose series determines the decline in vaccine effectiveness over time. Here we report on maintenance of immune responses during the year following a two-dose schedule of ChAdOx1 nCoV-19/AZD1222, in the absence of infection, and also explore the decay of antibody after infection. Total spike-specific IgG antibody titres were lower with two low doses of ChAdOx1 nCoV-19 vaccines (two low doses) (P = 0.0006) than with 2 standard doses (the approved dose) or low dose followed by standard dose vaccines regimens. Longer intervals between first and second doses resulted in higher antibody titres (P < 0.0001); however, there was no evidence that the trajectory of antibody decay differed by interval or by vaccine dose, and the decay of IgG antibody titres followed a similar trajectory after a third dose of ChAdOx1 nCoV-19. Trends in post-infection samples were similar with an initial rapid decay in responses but good persistence of measurable responses thereafter. Extrapolation of antibody data, following two doses of ChAdOx1 nCov-19, demonstrates a slow rate of antibody decay with modelling, suggesting that antibody titres are well maintained for at least 2 years. These data suggest a persistent immune response after two doses of ChAdOx1 nCov-19 which will likely have a positive impact against serious disease and hospitalization.
Collapse
Affiliation(s)
- Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Amy Flaxman
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Jeremy Aboagye
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Mustapha Bittaye
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Federica Cappuccini
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Elizabeth A Clutterbuck
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sophie Davies
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Nick J Edwards
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK
| | - Katie J Ewer
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust and Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Pedro M Folegatti
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jamie Fowler
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ciaran Gilbride
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah C Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK
| | - Leila Godfrey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | | | - Daniel Jenkin
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK
| | - Simon Kerridge
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Yama F Mujadidi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Emma Plested
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Maheshi N Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Hannah Robinson
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Helen Sanders
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Rinn Song
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Marta Ulaszewska
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK
| | - Danielle Woods
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel Wright
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| |
Collapse
|
48
|
Moore KA, Leighton T, Ostrowsky JT, Anderson CJ, Danila RN, Ulrich AK, Lackritz EM, Mehr AJ, Baric RS, Baylor NW, Gellin BG, Gordon JL, Krammer F, Perlman S, Rees HV, Saville M, Weller CL, Osterholm MT. A research and development (R&D) roadmap for broadly protective coronavirus vaccines: A pandemic preparedness strategy. Vaccine 2023; 41:2101-2112. [PMID: 36870874 PMCID: PMC9941884 DOI: 10.1016/j.vaccine.2023.02.032] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/23/2023]
Abstract
Broadly protective coronavirus vaccines are an important tool for protecting against future SARS-CoV-2 variants and could play a critical role in mitigating the impact of future outbreaks or pandemics caused by novel coronaviruses. The Coronavirus Vaccines Research and Development (R&D) Roadmap (CVR) is aimed at promoting the development of such vaccines. The CVR, funded by the Bill & Melinda Gates Foundation and The Rockefeller Foundation, was generated through a collaborative and iterative process, which was led by the Center for Infectious Disease Research and Policy (CIDRAP) at the University of Minnesota and involved 50 international subject matter experts and recognized leaders in the field. This report summarizes the major issues and areas of research outlined in the CVR and identifies high-priority milestones. The CVR covers a 6-year timeframe and is organized into five topic areas: virology, immunology, vaccinology, animal and human infection models, and policy and finance. Included in each topic area are key barriers, gaps, strategic goals, milestones, and additional R&D priorities. The roadmap includes 20 goals and 86 R&D milestones, 26 of which are ranked as high priority. By identifying key issues, and milestones for addressing them, the CVR provides a framework to guide funding and research campaigns that promote the development of broadly protective coronavirus vaccines.
Collapse
Affiliation(s)
- Kristine A Moore
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA; Center for Infectious Disease Research and Policy, C315 Mayo Memorial Building, MMC 263, 420 Delaware Street, SE, Minneapolis, Minnesota 55455, USA.
| | - Tabitha Leighton
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Julia T Ostrowsky
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cory J Anderson
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Angela K Ulrich
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Eve M Lackritz
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Angela J Mehr
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ralph S Baric
- University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | - Jennifer L Gordon
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Florian Krammer
- Department of Microbiology, Department of Pathology, Molecular and Cell-Based Medicine, and Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | | | - Helen V Rees
- Wits RHI, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Melanie Saville
- Coalition for Epidemic Preparedness Innovations, London, United Kingdom
| | | | - Michael T Osterholm
- Center for Infectious Disease Research and Policy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
49
|
Moore SC, Kronsteiner B, Longet S, Adele S, Deeks AS, Liu C, Dejnirattisai W, Reyes LS, Meardon N, Faustini S, Al-Taei S, Tipton T, Hering LM, Angyal A, Brown R, Nicols AR, Dobson SL, Supasa P, Tuekprakhon A, Cross A, Tyerman JK, Hornsby H, Grouneva I, Plowright M, Zhang P, Newman TAH, Nell JM, Abraham P, Ali M, Malone T, Neale I, Phillips E, Wilson JD, Murray SM, Zewdie M, Shields A, Horner EC, Booth LH, Stafford L, Bibi S, Wootton DG, Mentzer AJ, Conlon CP, Jeffery K, Matthews PC, Pollard AJ, Brown A, Rowland-Jones SL, Mongkolsapaya J, Payne RP, Dold C, Lambe T, Thaventhiran JED, Screaton G, Barnes E, Hopkins S, Hall V, Duncan CJA, Richter A, Carroll M, de Silva TI, Klenerman P, Dunachie S, Turtle L. Evolution of long-term vaccine-induced and hybrid immunity in healthcare workers after different COVID-19 vaccine regimens. MED 2023; 4:191-215.e9. [PMID: 36863347 PMCID: PMC9933851 DOI: 10.1016/j.medj.2023.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023]
Abstract
BACKGROUND Both infection and vaccination, alone or in combination, generate antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, the maintenance of such responses-and hence protection from disease-requires careful characterization. In a large prospective study of UK healthcare workers (HCWs) (Protective Immunity from T Cells in Healthcare Workers [PITCH], within the larger SARS-CoV-2 Immunity and Reinfection Evaluation [SIREN] study), we previously observed that prior infection strongly affected subsequent cellular and humoral immunity induced after long and short dosing intervals of BNT162b2 (Pfizer/BioNTech) vaccination. METHODS Here, we report longer follow-up of 684 HCWs in this cohort over 6-9 months following two doses of BNT162b2 or AZD1222 (Oxford/AstraZeneca) vaccination and up to 6 months following a subsequent mRNA booster vaccination. FINDINGS We make three observations: first, the dynamics of humoral and cellular responses differ; binding and neutralizing antibodies declined, whereas T and memory B cell responses were maintained after the second vaccine dose. Second, vaccine boosting restored immunoglobulin (Ig) G levels; broadened neutralizing activity against variants of concern, including Omicron BA.1, BA.2, and BA.5; and boosted T cell responses above the 6-month level after dose 2. Third, prior infection maintained its impact driving larger and broader T cell responses compared with never-infected people, a feature maintained until 6 months after the third dose. CONCLUSIONS Broadly cross-reactive T cell responses are well maintained over time-especially in those with combined vaccine and infection-induced immunity ("hybrid" immunity)-and may contribute to continued protection against severe disease. FUNDING Department for Health and Social Care, Medical Research Council.
Collapse
Affiliation(s)
- Shona C Moore
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stephanie Longet
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sandra Adele
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Alexandra S Deeks
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Chang Liu
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Wanwisa Dejnirattisai
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Division of Emerging Infectious Disease, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Laura Silva Reyes
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Naomi Meardon
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Sian Faustini
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Saly Al-Taei
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK
| | - Tom Tipton
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Luisa M Hering
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Adrienn Angyal
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Rebecca Brown
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Alexander R Nicols
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Susan L Dobson
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Aekkachai Tuekprakhon
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew Cross
- Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Jessica K Tyerman
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Hailey Hornsby
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Irina Grouneva
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Megan Plowright
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Peijun Zhang
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Thomas A H Newman
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Jeremy M Nell
- Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Priyanka Abraham
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Mohammad Ali
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Tom Malone
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Isabel Neale
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Eloise Phillips
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Joseph D Wilson
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Oxford University Medical School, Medical Sciences Division, University of Oxford, Oxford, UK
| | - Sam M Murray
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Martha Zewdie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Adrian Shields
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Emily C Horner
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lucy H Booth
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Lizzie Stafford
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Daniel G Wootton
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK; Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Christopher P Conlon
- Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Katie Jeffery
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Philippa C Matthews
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; The Francis Crick Institute, London, UK; Division of Infection and Immunity, University College London, London, UK; Department of Infectious Diseases, University College London Hospital NHS Foundation Trust, London, UK
| | - Andrew J Pollard
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Anthony Brown
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Sarah L Rowland-Jones
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Rebecca P Payne
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK
| | - Christina Dold
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | | | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Susan Hopkins
- UK Health Security Agency, London, UK; Faculty of Medicine, Department of Infectious Disease, Imperial College London, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Victoria Hall
- UK Health Security Agency, London, UK; NIHR Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, University of Oxford, Oxford, UK
| | - Christopher J A Duncan
- Translational and Clinical Research Institute, Immunity and Inflammation Theme, Newcastle University, Newcastle, UK; Department of Infection and Tropical Medicine, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Alex Richter
- Institute for Immunology and Immunotherapy, College of Medical and Dental Science, University of Birmingham, Birmingham, UK; University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Miles Carroll
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK; Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Thushan I de Silva
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK.
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford Centre for Global Health Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Lance Turtle
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK; Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
50
|
Addetia A, Piccoli L, Case JB, Park YJ, Beltramello M, Guarino B, Dang H, Pinto D, Scheaffer S, Sprouse K, Bassi J, Silacci-Fregni C, Muoio F, Dini M, Vincenzetti L, Acosta R, Johnson D, Subramanian S, Saliba C, Giurdanella M, Lombardo G, Leoni G, Culap K, McAlister C, Rajesh A, Dellota E, Zhou J, Farhat N, Bohan D, Noack J, Lempp FA, Cameroni E, Whitener B, Giannini O, Ceschi A, Ferrari P, Franzetti-Pellanda A, Biggiogero M, Garzoni C, Zappi S, Bernasconi L, Kim MJ, Schnell G, Czudnochowski N, Franko N, Logue JK, Yoshiyama C, Stewart C, Chu H, Schmid MA, Purcell LIA, Snell G, Lanzavecchia A, Diamond M, Corti D, Veesler D. Therapeutic and vaccine-induced cross-reactive antibodies with effector function against emerging Omicron variants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.523798. [PMID: 36711984 PMCID: PMC9882201 DOI: 10.1101/2023.01.17.523798] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Currently circulating SARS-CoV-2 variants acquired convergent mutations at receptor-binding domain (RBD) hot spots. Their impact on viral infection, transmission, and efficacy of vaccines and therapeutics remains poorly understood. Here, we demonstrate that recently emerged BQ.1.1. and XBB.1 variants bind ACE2 with high affinity and promote membrane fusion more efficiently than earlier Omicron variants. Structures of the BQ.1.1 and XBB.1 RBDs bound to human ACE2 and S309 Fab (sotrovimab parent) explain the altered ACE2 recognition and preserved antibody binding through conformational selection. We show that sotrovimab binds avidly to all Omicron variants, promotes Fc-dependent effector functions and protects mice challenged with BQ.1.1, the variant displaying the greatest loss of neutralization. Moreover, in several donors vaccine-elicited plasma antibodies cross-react with and trigger effector functions against Omicron variants despite reduced neutralizing activity. Cross-reactive RBD-directed human memory B cells remained dominant even after two exposures to Omicron spikes, underscoring persistent immune imprinting. Our findings suggest that this previously overlooked class of cross-reactive antibodies, exemplified by S309, may contribute to protection against disease caused by emerging variants through elicitation of effector functions.
Collapse
|