98 results on '"Balazs AB"'
Search Results
2. Towards a New—and Broader—History of Hungary’s Troubled Peacemaking
- Author
-
Balázs Ablonczy and Gergely Romsics
- Subjects
Trianon, Momentum grant, World War I aftermath, population movements, border change, history, and public remembrance ,Archaeology ,CC1-960 ,Ethnology. Social and cultural anthropology ,GN301-674 ,History of Central Europe ,DAW1001-1051 - Abstract
This research report presents the endeavors and findings of the Trianon 100 Research Group, which was founded with the support of the Lendület [Momentum] program of the Hungarian Academy of Sciences. Based on a survey of over a dozen volumes published by the group and another ten collaborative volumes, as well as numerous essays and articles, the report argues for the possibility of using newer instruments in the historian’s toolbox to tackle controversial issues in modern history from new perspectives. The Trianon 100 Research Group focused in particular on the history of mentalities, the interactions among bodies and individuals at different levels of societal organization (local/regional/national/imperial), and an array of under-researched areas, such as the history of population movements after 1918. The essential aim was to suggest that it might be possible to (re-)write the history of post-World War I peacemaking from a less Westphalian perspective, informed in particular by historical political sociology and social history.
- Published
- 2023
- Full Text
- View/download PDF
3. Waters, Leslie. 2022. Borders on the Move: Territorial Changes and Ethnic Cleansing in the Hungarian-Slovak Borderlands, 1938-1948. Rochester, University of Rochester Press (Rochester Studies in East and Central Europe).
- Author
-
Balázs Ablonczy
- Subjects
Hungary ,DB901-999 ,Language and Literature - Published
- 2022
- Full Text
- View/download PDF
4. Broad immunogenicity to prior SARS-CoV-2 strains and JN.1 variant elicited by XBB.1.5 vaccination in nursing home residents.
- Author
-
Abul Y, Nugent C, Vishnepolskiy I, Wallace T, Dickerson E, Holland L, Esparza I, Winkis M, Wali KT, Chan PA, Baier RR, Recker A, Kaczynski M, Kamojjala S, Pralea A, Rice H, Osias O, Oyebanji OA, Olagunju O, Cao Y, Li CJ, Roederer A, Pfeifer WM, Bosch J, King CL, Nanda A, McNicoll L, Mujahid N, Raza S, Tyagi R, Wilson BM, White EM, Canaday DH, Gravenstein S, and Balazs AB
- Abstract
SARS-CoV-2 vaccination has reduced hospitalization and mortality for nursing home residents (NHRs) but emerging variants and waning immunity challenge vaccine effectiveness. This study assesses the immunogenicity of the most recent XBB.1.5 monovalent vaccine to variant strains among NHRs. Participants were subset of a longitudinal study of consented NHRs and Healthcare workers (HCWs) who have received serial blood draws to assess immunogenicity with each SARS-CoV-2 mRNA vaccine dose. We report data on participants who received the XBB.1.5 monovalent vaccine post-FDA approval in Fall 2023. NHRs were categorized by whether they had an interval SARS-CoV-2 infection between their first bivalent vaccine dose and their XBB.1.5 monovalent vaccination. The sample included 61 NHRs [median age 76 (IQR 68-86), 51% female] and 28 HCWs [median age 45 (IQR 31-58), 46% female). After XBB.1.5 vaccination, a robust geometric mean fold rise (GMFR) in XBB.1.5-specific neutralizing antibody titers was observed:17.3 (95% confidence interval [CI] 9.3, 32.4) and NHRs with interval infection and 11.3 (95% CI 5, 25.4) in those without and 13.6 (95% CI 8.4,22) in HCWs. For JN.1-specific titers, GMFRs were 14.9 (95% CI 7.9, 28) and 6.5 (95% CI 3.3, 13.1) in NHRs with and without interval infection, and 11.4 (95% CI 6.2, 20.9) in HCWs. NHRs with interval SARS-CoV-2 infection had higher titers across all analyzed strains analyzed. The XBB.1.5 vaccine significantly elevates Omicron-specific neutralizing antibody titers to XBB.1.5 and JN.1 strains in both NHRs and HCWs with more pronounced in those previously infected with SARS-CoV-2 since bivalent vaccination., (© 2024. This is a U.S. Government work and not under copyright protection in the US; foreign copyright protection may apply.)
- Published
- 2024
- Full Text
- View/download PDF
5. QuickFit: A High-Throughput RT-qPCR-Based Assay to Quantify Viral Growth and Fitness In Vitro.
- Author
-
Galvez NMS, Sheehan ML, Lin AZ, Cao Y, Lam EC, Jackson AM, and Balazs AB
- Subjects
- Humans, High-Throughput Screening Assays methods, HIV-1 genetics, HIV-1 physiology, HIV-1 growth & development, Real-Time Polymerase Chain Reaction methods, Genetic Fitness, HIV Infections virology, HIV Infections drug therapy, HIV genetics, HIV physiology, HIV growth & development, Cell Line, Virus Replication
- Abstract
Quantifying viral growth rates is key to understanding evolutionary dynamics and the potential for mutants to escape antiviral drugs. Defining evolutionary escape paths and their impact on viral fitness allows for the development of drugs that are resistant to escape. In the case of HIV, combination antiretroviral therapy can successfully prevent or treat infection, but it relies on strict adherence to prevent escape. Here, we present a method termed QuickFit that enables the quantification of viral fitness by employing large numbers of parallel viral cultures to measure growth rates accurately. QuickFit consistently recapitulated HIV growth measurements obtained by traditional approaches, but with significantly higher throughput and lower rates of error. This method represents a promising tool for rapid and consistent evaluation of viral fitness.
- Published
- 2024
- Full Text
- View/download PDF
6. A Novel Human Extravascular Monocyte Subset with Antiviral Functions Is Crucial for Resolving Lung Tissue Infection.
- Author
-
Kenney D, O'Connell AK, Tseng AE, Turcinovic J, Sheehan ML, Nitido AD, Montanaro P, Gertje HP, Ericsson M, Connor JH, Vrbanac V, Crossland NA, Harly C, Balazs AB, and Douam F
- Abstract
The recurring emergence of novel respiratory viruses has highlighted our poor understanding of the human immune mechanisms governing the resolution of lung infection in an immunologically naïve context. Using SARS-CoV-2 as a prototypical emerging respiratory virus, we leveraged mice co-engrafted with a genetically matched fetal lung xenograft (fLX) and a human immune system (BLT-L mice) to investigate such mechanisms. While BLT-L mice effectively resolve SARS-CoV-2 infection following acute viral replication in fLX, viral clearance is robustly abrogated through systemic depletion of CD4+, but not CD3+ or CD8+ cells, resulting in persistent infection. Leveraging single-cell transcriptomics to uncover the CD4-expressing subsets driving infection resolution, we identified a novel subset of lung extravascular inflammatory monocytes (ExiMO) with antiviral functions. ExiMO are the dominant CD163-expressing myeloid population emerging in fLX upon acute infection and derive from recruited circulating CD4+ monocytes. They are highly enriched in viral RNA and elicit a robust antiviral response before vanishing from tissues when infection resolves. Notably, systemic CD4+ cell depletion results in impaired recruitment of CD163+ cells into fLX and leads to a state of immune tolerance and chronic infection defined by the absence of ExiMO antiviral responses. Together, our study uncovers ExiMO as major sentinels driving SARS-CoV-2 infection resolution in human lung tissues without pre-existing immunity. This work expands our understanding of lung extravascular monocytes and unravels novel facets of the cellular determinants governing our vulnerability to viral respiratory pathogens., Competing Interests: The authors have declared that no conflict of interest exists.
- Published
- 2024
- Full Text
- View/download PDF
7. HIV broadly neutralizing antibody escapability drives the therapeutic efficacy of vectored immunotherapy.
- Author
-
Galvez NMS, Cao Y, Nitido AD, Deal CE, Boutros CL, MacDonald SW, Albrecht YES, Lam EC, Sheehan ML, Parsons D, Lin AZ, Deymier MJ, Brady JM, Moon B, Bullock CB, Tanno S, Pegu A, Chen X, Liu C, Koup RA, Mascola JR, Vrbanac VD, Lingwood D, and Balazs AB
- Abstract
Broadly neutralizing antibodies (bNAbs) have shown great promise for prevention and treatment of HIV infection. Breadth of bNAb neutralization, measured in vitro across panels of diverse viral isolates, is often used as a predictor of clinical potential. However, recent prevention studies demonstrate that the clinical efficacy of a broad and potent bNAb (VRC01) is undermined by neutralization resistance of circulating strains. Using HIV-infected humanized mice, we find that therapeutic efficacy of bNAbs delivered as Vectored ImmunoTherapy (VIT) is a function of both the fitness cost and resistance benefit of mutations that emerge during viral escape, which we term 'escapability'. Applying this mechanistic framework, we find that the sequence of the envelope V5-loop alters the resistance benefits of mutants that arise during escape, thereby impacting the therapeutic efficacy of VIT-mediated viral suppression. We also find that an emtricitabine-based antiretroviral drug regimen dramatically enhances the efficacy of VIT, by reducing the fitness of mutants along the escape path. Our findings demonstrate that bNAb escapability is a key determinant to consider in the rational design of antibody regimens with maximal efficacy and illustrates a tractable means of minimizing viral escape from existing bNAbs., Competing Interests: DECLARATION OF INTERESTS A.B.B. is a founder of Cure Systems LLC.
- Published
- 2024
- Full Text
- View/download PDF
8. Durability of immunity and clinical protection in nursing home residents following bivalent SARS-CoV-2 vaccination.
- Author
-
Gravenstein S, DeVone F, Oyebanji OA, Abul Y, Cao Y, Chan PA, Halladay CW, Rudolph JL, Nugent C, Bosch J, King CL, Wilson BM, Balazs AB, White EM, Canaday DH, and McConeghy KW
- Subjects
- Humans, Female, Male, Aged, Aged, 80 and over, Antibodies, Neutralizing immunology, Antibodies, Neutralizing blood, Immunization, Secondary, Vaccine Efficacy, Spike Glycoprotein, Coronavirus immunology, Nursing Homes, COVID-19 Vaccines immunology, COVID-19 Vaccines administration & dosage, COVID-19 prevention & control, COVID-19 immunology, SARS-CoV-2 immunology, Antibodies, Viral immunology, Antibodies, Viral blood, Vaccination
- Abstract
Background: Bivalent SARS-CoV-2 vaccines were developed to counter increasing susceptibility to emerging SARS-CoV-2 variants. We evaluated the durability of immunity and protection following first bivalent vaccination among nursing home residents., Methods: We evaluated anti-spike and neutralization titers from blood in 653 community nursing home residents before and after each monovalent booster, and a bivalent vaccine. Concurrent clinical outcomes were evaluated using electronic health record data from a separate cohort of 3783 residents of Veterans Affairs (VA) nursing homes who had received at least the primary series monovalent vaccination. Using target trial emulation, we compared VA residents who did and did not receive the bivalent vaccine to measure vaccine effectiveness against infection, hospitalization, and death., Findings: In the community cohort, Omicron BA.5 neutralization activity rose after each monovalent and bivalent booster vaccination regardless of prior infection history. Titers declined over time but six months post-bivalent vaccination, BA.5 neutralization persisted at detectable levels in 75% of infection-naive and 98% of prior-infected individuals. In the VA nursing home cohort, bivalent vaccine added effectiveness to monovalent booster vaccination by 18.5% for infection (95% confidence interval (CI) -5.6, 34.0%), and 29.2% for hospitalization or death (95% CI -14.2, 56.2%) over five months., Interpretation: The level of protection declined after bivalent vaccination over a 6 month period and may open a window of added vulnerability before the next updated vaccine becomes available, suggesting a subset of nursing home residents may benefit from an additional vaccination booster., Funding: CDC, NIH, VHA., Competing Interests: Declaration of interests Stefan Gravenstein (S. G.) and David H. Canaday (D. H. C.) are recipients of investigator-initiated grants to their universities from Pfizer to study pneumococcal vaccines, Moderna to study respiratory infections, and Sanofi Pasteur and Seqirus to study influenza vaccines, and S.G. from Genentech on influenza antivirals. S. G. also receives consulting fees from GlaxoSmithKline, Icosavax, Janssen, Merck, Moderna, Novavax, Pfizer, Reviral, Sanofi, Seqirus, and Vaxart, and has received fees for speaking for Janssen, Pfizer, Moderna, GlaxoSmithKline, Sanofi, and Seqirus. KWM Investigator initiated research support from Seqirus pharmaceuticals, Sanofi-Pasteur, Genentech and Pfizer., (Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
9. mRNA-based monkeypox virus vaccine prevents disease in non-human primates.
- Author
-
Nitido AN and Balazs AB
- Subjects
- Animals, Monkeypox virus immunology, Primates, mRNA Vaccines immunology, Smallpox Vaccine immunology, Viral Vaccines immunology
- Abstract
The mpox outbreak in 2022 launched a vaccination campaign employing an existing vaccine with moderate protection, highlighting the lack of scalable Orthopoxvirus vaccines with optimal protection. In this issue of Cell, Zuiani et al. report pre-clinical findings of an mRNA-based mpox vaccine, paving the way for Phase I/II clinical trials., Competing Interests: Declaration of interests A.B.B. is a founder of Cure Systems LLC., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
10. Ongoing evolution of SARS-CoV-2 drives escape from mRNA vaccine-induced humoral immunity.
- Author
-
Roederer AL, Cao Y, Denis KS, Sheehan ML, Li CJ, Lam EC, Gregory DJ, Poznansky MC, Iafrate AJ, Canaday DH, Gravenstein S, Garcia-Beltran WF, and Balazs AB
- Abstract
Since the COVID-19 pandemic began in 2020, viral sequencing has documented 131 individual mutations in the viral spike protein across 48 named variants. To determine the ability of vaccine-mediated humoral immunity to keep pace with continued SARS-CoV-2 evolution, we assessed the neutralization potency of sera from 76 vaccine recipients collected after 2 to 6 immunizations against a comprehensive panel of mutations observed during the pandemic. Remarkably, while many individual mutations that emerged between 2020 and 2022 exhibit escape from sera following primary vaccination, few escape boosted sera. However, progressive loss of neutralization was observed across newer variants, irrespective of vaccine doses. Importantly, an updated XBB.1.5 booster significantly increased titers against newer variants but not JN.1. These findings demonstrate that seasonal boosters improve titers against contemporaneous strains, but novel variants continue to evade updated mRNA vaccines, demonstrating the need for novel approaches to adequately control SARS-CoV-2 transmission., Competing Interests: DECLARATIONS OF INTEREST A.B.B. is a founder of Cure Systems LLC.
- Published
- 2024
- Full Text
- View/download PDF
11. Heterologous Sarbecovirus Receptor Binding Domains as Scaffolds for SARS-CoV-2 Receptor Binding Motif Presentation.
- Author
-
Hauser BM, Sangesland M, Lam EC, St Denis KJ, Sheehan ML, Vu ML, Cheng AH, Sordilla S, Lamson DT, Almawi AW, Balazs AB, Lingwood D, and Schmidt AG
- Subjects
- Humans, SARS-CoV-2, Antibodies, Neutralizing, Severe acute respiratory syndrome-related coronavirus, COVID-19
- Abstract
Structure-guided rational immunogen design can generate optimized immunogens that elicit a desired humoral response. Design strategies often center on targeting conserved sites on viral glycoproteins that will ultimately confer potent neutralization. For SARS-CoV-2 (SARS-2), the surface-exposed spike glycoprotein includes a broadly conserved portion, the receptor binding motif (RBM), that is required to engage the host cellular receptor, ACE2. Expanding humoral responses to this site may result in a more potent neutralizing antibody response against diverse sarbecoviruses. Here, we used a "resurfacing" approach and iterative design cycles to graft the SARS-2 RBM onto heterologous sarbecovirus scaffolds. The scaffolds were selected to vary the antigenic distance relative to SARS-2 to potentially focus responses to RBM. Multimerized versions of these immunogens elicited broad neutralization against sarbecoviruses in the context of preexisting SARS-2 immunity. These validated engineering approaches can help inform future immunogen design efforts for sarbecoviruses and are generally applicable to other viruses.
- Published
- 2024
- Full Text
- View/download PDF
12. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds.
- Author
-
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Case JB, Sanapala S, Lam EC, Denis KJS, Boucau J, Barczak AK, Balazs AB, Diamond MS, Schmidt AG, Lingwood D, and Bathe M
- Subjects
- Humans, Animals, Mice, Antibodies, Blocking, Antibodies, Neutralizing, DNA, Antibodies, Viral, Antibody Formation, Vaccines, Virus-Like Particle genetics, Spike Glycoprotein, Coronavirus
- Abstract
Protein-based virus-like particles (P-VLPs) are commonly used to spatially organize antigens and enhance humoral immunity through multivalent antigen display. However, P-VLPs are thymus-dependent antigens that are themselves immunogenic and can induce B cell responses that may neutralize the platform. Here, we investigate thymus-independent DNA origami as an alternative material for multivalent antigen display using the receptor binding domain (RBD) of the SARS-CoV-2 spike protein, the primary target of neutralizing antibody responses. Sequential immunization of mice with DNA-based VLPs (DNA-VLPs) elicits protective neutralizing antibodies to SARS-CoV-2 in a manner that depends on the valency of the antigen displayed and on T cell help. Importantly, the immune sera do not contain boosted, class-switched antibodies against the DNA scaffold, in contrast to P-VLPs that elicit strong B cell memory against both the target antigen and the scaffold. Thus, DNA-VLPs enhance target antigen immunogenicity without generating scaffold-directed immunity and thereby offer an important alternative material for particulate vaccine design., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
13. Neutralization and binding antibody response to second bivalent COVID-19 vaccination in nursing home residents.
- Author
-
Oyebanji OA, Abul Y, Wilson BM, Bosch J, Didion EM, Paxitzis AN, Sundheimer N, Ragavapuram V, Wilk D, Keresztesy D, Aung H, Cao Y, King CL, Balazs AB, White EM, Gravenstein S, and Canaday DH
- Subjects
- Humans, Antibody Formation, Nursing Homes, Vaccination, COVID-19 Vaccines, COVID-19 prevention & control
- Published
- 2023
- Full Text
- View/download PDF
14. De novo hematopoiesis from the fetal lung.
- Author
-
Yeung AK, Villacorta-Martin C, Lindstrom-Vautrin J, Belkina AC, Vanuytsel K, Dowrey TW, Ysasi AB, Bawa P, Wang F, Vrbanac V, Mostoslavsky G, Balazs AB, and Murphy GJ
- Subjects
- Animals, Mice, Humans, Hematopoietic Stem Cells metabolism, Cell Differentiation, Endothelium, Hematopoiesis, Hemangioblasts metabolism
- Abstract
Hemogenic endothelial cells (HECs) are specialized cells that undergo endothelial-to-hematopoietic transition (EHT) to give rise to the earliest precursors of hematopoietic progenitors that will eventually sustain hematopoiesis throughout the lifetime of an organism. Although HECs are thought to be primarily limited to the aorta-gonad-mesonephros (AGM) during early development, EHT has been described in various other hematopoietic organs and embryonic vessels. Though not defined as a hematopoietic organ, the lung houses many resident hematopoietic cells, aids in platelet biogenesis, and is a reservoir for hematopoietic stem and progenitor cells (HSPCs). However, lung HECs have never been described. Here, we demonstrate that the fetal lung is a potential source of HECs that have the functional capacity to undergo EHT to produce de novo HSPCs and their resultant progeny. Explant cultures of murine and human fetal lungs display adherent endothelial cells transitioning into floating hematopoietic cells, accompanied by the gradual loss of an endothelial signature. Flow cytometric and functional assessment of fetal-lung explants showed the production of multipotent HSPCs that expressed the EHT and pre-HSPC markers EPCR, CD41, CD43, and CD44. scRNA-seq and small molecule modulation demonstrated that fetal lung HECs rely on canonical signaling pathways to undergo EHT, including TGFβ/BMP, Notch, and YAP. Collectively, these data support the possibility that post-AGM development, functional HECs are present in the fetal lung, establishing this location as a potential extramedullary site of de novo hematopoiesis., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
15. Rheumatoid arthritis and older age are associated with lower humoral and cellular immune response to primary series COVID-19 mRNA vaccine.
- Author
-
Dudley HM, O'Mara M, Auma A, Gong J, Ross Y, Gurevich N, Carbone S, Reihs A, Nguyen Y, McComsey GA, Cao Y, Balazs AB, Gordesky L, Payne M, Singer N, Kostadinova L, Wilson B, Zidar DA, King CL, Canaday DH, Shive CL, Mattar MM, and Anthony DD
- Subjects
- Humans, Aged, COVID-19 Vaccines, Leukocytes, Mononuclear, Interleukin-2, Antibodies, Neutralizing, Immunity, Cellular, Immunoglobulin G, COVID-19 prevention & control, Arthritis, Rheumatoid
- Abstract
Objective: People with autoimmune disease have worse COVID-19 infection-related outcomes, lower antibody responses to COVID-19 vaccine, and higher rates of breakthrough infection. Immunosuppressive medications used to treat rheumatoid arthritis (RA) are associated with lower COVID-19 vaccine responses, though independent contributions of comorbidities, T-cell immunity, and age are less clear. We sought to test the hypothesis that RA, immunosuppressive medications used to treat RA, and older age, contribute to reduced B and T cell response to COVID-19 vaccine., Methods: We evaluated serum samples, taken the day of 1st vaccine dose, the day of 2nd dose, 2-6 weeks after 2nd dose, 7-12 weeks after 2nd dose, 13-24 weeks after 2nd dose, and 2-6 weeks after the 3rd dose, for anti-spike IgG and neutralizing antibody levels to Wuhan and Omicron BA.1 and peripheral blood mononuclear cells (PBMC) for spike-specific IFN-γ and IL-2 production by ELISPOT assay in 46 RA and 101 non-autoimmune control participants before and after the primary series COVID-19 mRNA vaccination., Results: RA participants had lower spike-specific IgG and Wuhan-strain neutralizing antibody levels 2-6 weeks compared to controls after the second dose of primary vaccine series. Neutralizing antibody levels against Omicron BA.1 were low in both groups. IFN-γ production correlated with Wuhan neutralizing antibody levels, while older age negatively correlated with spike-specific IL-2, IFN-γ and IgG. Lower antibody levels were associated with older age, RA status, and medication usage, while lower T cell responses were associated primarily with older age., Conclusions: These data indicate lower COVID-19 mRNA vaccine-induced antibody levels in persons with RA compared to individuals without RA, likely partially attributable to immune suppressive medications. At the same time, older age is associated with lower antibody and cellular immune response to COVID-19 vaccines., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
16. Heterologous sarbecovirus receptor binding domains as scaffolds for SARS-CoV-2 receptor binding motif presentation.
- Author
-
Hauser BM, Sangesland M, Lam EC, Denis KJS, Sheehan ML, Vu ML, Cheng AH, Balazs AB, Lingwood D, and Schmidt AG
- Abstract
Structure-guided rational immunogen design can generate optimized immunogens that elicit a desired humoral response. Design strategies often center upon targeting conserved sites on viral glycoproteins that will ultimately confer potent neutralization. For SARS-CoV-2 (SARS-2), the surface-exposed spike glycoprotein includes a broadly conserved portion, the receptor binding motif (RBM), that is required to engage the host cellular receptor, ACE2. Expanding humoral responses to this site may result in a more potently neutralizing antibody response against diverse sarbecoviruses. Here, we used a "resurfacing" approach and iterative design cycles to graft the SARS-2 RBM onto heterologous sarbecovirus scaffolds. The scaffolds were selected to vary the antigenic distance relative to SARS-2 to potentially focus responses to RBM. Multimerized versions of these immunogens elicited broad neutralization against sarbecoviruses in the context of preexisting SARS-2 immunity. These validated engineering approaches can help inform future immunogen design efforts for sarbecoviruses and are generally applicable to other viruses., Competing Interests: Declaration of interests: The authors have no competing interests to declare.
- Published
- 2023
- Full Text
- View/download PDF
17. Delivery platforms for broadly neutralizing antibodies.
- Author
-
Joshi LR, Gálvez NMS, Ghosh S, Weiner DB, and Balazs AB
- Subjects
- Animals, Humans, Broadly Neutralizing Antibodies, HIV Antibodies, Antibodies, Neutralizing, Pandemics, Antibodies, Monoclonal genetics, HIV Infections prevention & control, HIV-1 genetics, COVID-19 therapy
- Abstract
Purpose of Review: Passive administration of broadly neutralizing antibodies (bNAbs) is being evaluated as a therapeutic approach to prevent or treat HIV infections. However, a number of challenges face the widespread implementation of passive transfer for HIV. To reduce the need of recurrent administrations of bNAbs, gene-based delivery approaches have been developed which overcome the limitations of passive transfer., Recent Findings: The use of DNA and mRNA for the delivery of bNAbs has made significant progress. DNA-encoded monoclonal antibodies (DMAbs) have shown great promise in animal models of disease and the underlying DNA-based technology is now being tested in vaccine trials for a variety of indications. The COVID-19 pandemic greatly accelerated the development of mRNA-based technology to induce protective immunity. These advances are now being successfully applied to the delivery of monoclonal antibodies using mRNA in animal models. Delivery of bNAbs using viral vectors, primarily adeno-associated virus (AAV), has shown great promise in preclinical animal models and more recently in human studies. Most recently, advances in genome editing techniques have led to engineering of monoclonal antibody expression from B cells. These efforts aim to turn B cells into a source of evolving antibodies that can improve through repeated exposure to the respective antigen., Summary: The use of these different platforms for antibody delivery has been demonstrated across a wide range of animal models and disease indications, including HIV. Although each approach has unique strengths and weaknesses, additional advances in efficiency of gene delivery and reduced immunogenicity will be necessary to drive widespread implementation of these technologies. Considering the mounting clinical evidence of the potential of bNAbs for HIV treatment and prevention, overcoming the remaining technical challenges for gene-based bNAb delivery represents a relatively straightforward path towards practical interventions against HIV infection., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
18. Enhancing antibody responses by multivalent antigen display on thymus-independent DNA origami scaffolds.
- Author
-
Wamhoff EC, Ronsard L, Feldman J, Knappe GA, Hauser BM, Romanov A, Lam E, Denis KS, Boucau J, Barczak AK, Balazs AB, Schmidt A, Lingwood D, and Bathe M
- Abstract
Multivalent antigen display is a well-established principle to enhance humoral immunity. Protein-based virus-like particles (VLPs) are commonly used to spatially organize antigens. However, protein-based VLPs are limited in their ability to control valency on fixed scaffold geometries and are thymus-dependent antigens that elicit neutralizing B cell memory themselves, which can distract immune responses. Here, we investigated DNA origami as an alternative material for multivalent antigen display in vivo, applied to the receptor binding domain (RBD) of SARS-CoV2 that is the primary antigenic target of neutralizing antibody responses. Icosahedral DNA-VLPs elicited neutralizing antibodies to SARS-CoV-2 in a valency-dependent manner following sequential immunization in mice, quantified by pseudo- and live-virus neutralization assays. Further, induction of B cell memory against the RBD required T cell help, but the immune sera did not contain boosted, class-switched antibodies against the DNA scaffold. This contrasted with protein-based VLP display of the RBD that elicited B cell memory against both the target antigen and the scaffold. Thus, DNA-based VLPs enhance target antigen immunogenicity without generating off-target, scaffold-directed immune memory, thereby offering a potentially important alternative material for particulate vaccine design., Competing Interests: Competing interests The Massachusetts Institute of Technology has filed a patent (US application number 16/752,394) covering the use of DNA origami as a vaccine platform on behalf of the co-inventors (E.-C. W. and M. B.).
- Published
- 2023
- Full Text
- View/download PDF
19. Brief Report: Declining Rates of SARS-CoV-2 Vaccine Uptake Among Patients With Thoracic Malignancies.
- Author
-
Meador CB, Naranbhai V, Hambelton G, Rivera J, Nabel CS, Lewinsohn R, Sakhi M, Balazs AB, Iafrate AJ, and Gainor JF
- Subjects
- Humans, COVID-19 Vaccines, SARS-CoV-2, COVID-19 epidemiology, COVID-19 prevention & control, Lung Neoplasms, Thoracic Neoplasms
- Published
- 2023
- Full Text
- View/download PDF
20. Generation and characterization of infectious molecular clones of transmitted/founder HIV-1 subtype C viruses.
- Author
-
Luthuli B, Gounder K, Deymier MJ, Dong KL, Balazs AB, Mann JK, and Ndung'u T
- Subjects
- Humans, Male, Female, Persistent Infection, Clone Cells, HIV-1, HIV Infections
- Abstract
The genetic diversity of HIV impedes vaccine development. Identifying the viral properties of transmitted/founder (T/F) variants may provide a common vaccine target. To study the biological nature of T/F viruses, we constructed full-length clones from women detected during Fiebig stage I acute HIV-1 infection (AHI) from heterosexual male-to-female (MTF) transmission; and clones after one year of infection using In-Fusion-based cloning. Eighteen full-length T/F clones were generated from 9 women and six chronic infection clones were from 2 individuals. All clones but one were non-recombinant subtype C. Three of the 5 T/F clones and 3 chronic clones tested replicated efficiently in PBMCs and utilised CCR5 coreceptor for cell entry. Transmitted/founder and chronic infection clones displayed heterogenous in vitro replicative capacity and resistance to type I interferon. T/F viruses had shorter Env glycoproteins and fewer N-linked glycosylation sites in Env. Our findings suggest MTF transmission may select viruses with compact envelopes., Competing Interests: Declaration of competing interest The authors declare that none of the authors have any conflicts of interest regarding this study., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Second monovalent SARS-CoV-2 mRNA booster restores Omicron-specific neutralizing activity in both nursing home residents and health care workers.
- Author
-
Nugent C, Abul Y, White EM, Shehadeh F, Kaczynski M, Oscar Felix L, Ganesan N, Oyebanji OA, Vishnepolskiy I, Didion EM, Paxitzis A, Sheehan ML, Chan PA, Pfeifer WM, Dickerson E, Kamojjala S, Wilson BM, Mylonakis E, King CL, Balazs AB, Canaday DH, and Gravenstein S
- Subjects
- Humans, COVID-19 Vaccines, SARS-CoV-2 genetics, Health Personnel, RNA, Messenger, Nursing Homes, Antibodies, Neutralizing, Antibodies, Viral, COVID-19 prevention & control
- Abstract
We examined whether the second monovalent SARS-CoV-2 mRNA booster increased antibody levels and their neutralizing activity to Omicron variants in nursing home residents (NH) residents and healthcare workers (HCW). We sampled 376 NH residents and 63 HCW after primary mRNA vaccination, first and second boosters, for antibody response and pseudovirus neutralization assay against SARS-CoV-2 wild-type (WT) (Wuhan-Hu-1) strain, Omicron BA.1 and BA.5 variants. Antibody levels and neutralizing activity progressively increased with each booster but subsequently waned over 3-6 months. NH residents, both those without and with prior infection, had a robust geometric mean fold rise (GMFR) of 8.1 (95% CI 4.4, 14.8) and 7.8 (95% CI 4.8, 12.9) respectively in Omicron-BA.1 subvariant specific neutralizing antibody levels following the second booster vaccination (p < 0.001). These results support the ongoing efforts to ensure that both NH residents and HCW are up-to-date on recommended SARS-CoV-2 vaccine booster doses., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: S. G. and D. H. C. are recipients of investigator-initiated grants to their universities from Pfizer to study pneumococcal vaccines and Sanofi Pasteur and Seqirus to study influenza vaccines, and S.G. from Genentech on influenza antivirals. S. G. also does consulting for Seqirus, Sanofi, Merck, Vaxart, Novavax, Moderna and Janssen; has served on the speaker’s bureaus for Seqirus and Sanofi; and reports personal fees from Pfizer and data and safety monitoring board (DSMB) fees from Longevoron and SciClone., (Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
22. Durability of immunity and clinical protection in nursing home residents following bivalent SARS-CoV-2 vaccination.
- Author
-
Gravenstein S, DeVone F, Oyebanji OA, Abul Y, Cao Y, Chan PA, Halladay CW, McConeghy KW, Nugent C, Bosch J, King CL, Wilson BM, Balazs AB, White EM, and Canaday DH
- Abstract
Background: Vaccines have substantially mitigated the disproportional impact of SARS-CoV-2 on the high morbidity and mortality experienced by nursing home residents. However, variation in vaccine efficacy, immune senescence and waning immunity all undermine vaccine effectiveness over time. The introduction of the bivalent vaccine in September 2022 aimed to counter this increasing susceptibility and consequences of breakthrough infection, however data on the durability and protection of the vaccine are limited. We evaluated the durability of immunity and protection after the first bivalent vaccination to SARS-CoV-2 in nursing home residents., Methods: For the immunologic evaluation, community nursing home volunteers agreed to serial blood sampling before, at two weeks, three and six months after each vaccination for antibodies to spike protein and pseudovirus neutralization activity over time. Concurrent clinical outcomes were evaluated by reviewing electronic health record data from residents living in Veterans Administration managed nursing home units. Residents without recent infection but prior vaccination to SARS-CoV-2 were followed over time beginning with administration of the newly available bivalent vaccine using a target trial emulation (TTE) approach; TTE compared time to breakthrough infection, hospitalization and death between those who did and did not receive the bivalent vaccine., Results: We evaluated antibodies in 650 nursing home residents; 452 had data available following a first monovalent booster, 257 following a second monovalent booster and 321 following a bivalent vaccine. We found a rise in BA.5 neutralization activity from the first and second monovalent boosters through the bivalent vaccination regardless of prior SARS-CoV-2 history. Titers declined at three and six months after the bivalent vaccination but generally exceeded those at three months compared to either prior boost. BA.5 neutralization titers six months after the bivalent vaccination were diminished but had detectable levels in 80% of infection-naive and 100% of prior infected individuals. TTE evaluated 5903 unique subjects, of whom 2235 received the bivalent boost. TTE demonstrated 39% or greater reduction in risk of infection, hospitalization or death at four months following the bivalent boost., Conclusion: Immunologic results mirrored those of the TTE and suggest bivalent vaccination added substantial protection for up to six months after bivalent vaccination with notable exceptions. However, the level of protection declined over this period, and by six months may open a window of added vulnerability to infection before the next updated vaccine becomes available. We strongly agree with the CDC recommendation that those who have not received a bivalent vaccination receive that now and these results support a second bivalent booster for those at greatest risk which includes many nursing home residents.
- Published
- 2023
- Full Text
- View/download PDF
23. Diagnostic TR-FRET assays for detection of antibodies in patient samples.
- Author
-
Yue H, Nowak RP, Overwijn D, Payne NC, Fischinger S, Atyeo C, Lam EC, St Denis K, Brais LK, Konishi Y, Sklavenitis-Pistofidis R, Baden LR, Nilles EJ, Karlson EW, Yu XG, Li JZ, Woolley AE, Ghobrial IM, Meyerhardt JA, Balazs AB, Alter G, Mazitschek R, and Fischer ES
- Subjects
- Humans, Fluorescence Resonance Energy Transfer, Antibodies, Viral, Nucleocapsid, COVID-19 Testing, SARS-CoV-2, COVID-19 diagnosis
- Abstract
Serological assays are important diagnostic tools for surveying exposure to the pathogen, monitoring immune response post vaccination, and managing spread of the infectious agent among the population. Current serological laboratory assays are often limited because they require the use of specialized laboratory technology and/or work with a limited number of sample types. Here, we evaluate an alternative by developing time-resolved Förster resonance energy transfer (TR-FRET) homogeneous assays that exhibited exceptional versatility, scalability, and sensitivity and outperformed or matched currently used strategies in terms of sensitivity, specificity, and precision. We validated the performance of the assays measuring total immunoglobulin G (IgG) levels; antibodies against severe acute respiratory syndrome coronavirus (SARS-CoV) or Middle Eastern respiratory syndrome (MERS)-CoV spike (S) protein; and SARS-CoV-2 S and nucleocapsid (N) proteins and applied it to several large sample sets and real-world applications. We further established a TR-FRET-based ACE2-S competition assay to assess the neutralization propensity of the antibodies. Overall, these TR-FRET-based serological assays can be rapidly extended to other antigens and are compatible with commonly used plate readers., Competing Interests: E.S.F. is an equity holder and scientific advisor for Neomorph, Inc. (board member), Civetta Therapeutics, Proximity Therapeutics, Lighthorse Therapeutics, Avilar Therapeutics, and Photys Therapeutics and is a consultant to Novartis, Sanofi, AbbVie, Pfizer, Astellas, EcoR1 Capital, and Deerfield. The Fischer lab receives or has received research funding from Novartis, Ajax, Deerfield, and Astellas not related to this work. R.M. is a scientific advisory board (SAB) member and equity holder of Regenacy Pharmaceuticals, ERX Pharmaceuticals, and Frequency Therapeutics. H.Y., R.P.N., D.O., N.C.P., R.M., and E.S.F. are inventors on patent applications related to this work., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
24. Association of Cytomegalovirus Serostatus With Severe Acute Respiratory Syndrome Coronavirus 2 Vaccine Responsiveness in Nursing Home Residents and Healthcare Workers.
- Author
-
Freeman ML, Oyebanji OA, Moisi D, Payne M, Sheehan ML, Balazs AB, Bosch J, King CL, Gravenstein S, Lederman MM, and Canaday DH
- Abstract
Background: Latent cytomegalovirus (CMV) infection is immunomodulatory and could affect mRNA vaccine responsiveness. We sought to determine the association of CMV serostatus and prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection with antibody (Ab) titers after primary and booster BNT162b2 mRNA vaccinations in healthcare workers (HCWs) and nursing home (NH) residents., Methods: Nursing home residents ( N = 143) and HCWs ( N = 107) were vaccinated and serological responses monitored by serum neutralization activity against Wuhan and Omicron (BA.1) strain spike proteins, and by bead-multiplex immunoglobulin G immunoassay to Wuhan spike protein and its receptor-binding domain (RBD). Cytomegalovirus serology and levels of inflammatory biomarkers were also measured., Results: Severe acute respiratory syndrome coronavirus 2-naive CMV seropositive (CMV
+ ) HCWs had significantly reduced Wuhan-neutralizing Ab ( P = .013), anti-spike ( P = .017), and anti-RBD ( P = .011) responses 2 weeks after primary vaccination series compared with responses among CMV seronegative (CMV- ) HCWs, adjusting for age, sex, and race. Among NH residents without prior SARS-CoV-2 infection, Wuhan-neutralizing Ab titers were similar 2 weeks after primary series but were reduced 6 months later ( P = .012) between CMV+ and CMV- subjects. Wuhan-neutralizing Ab titers from CMV+ NH residents who had prior SARS-CoV-2 infection consistently trended lower than titers from SARS-CoV-2 experienced CMV- donors. These impaired Ab responses in CMV+ versus CMV- individuals were not observed after booster vaccination or with prior SARS-CoV-2 infection., Conclusions: Latent CMV infection adversely affects vaccine-induced responsiveness to SARS-CoV-2 spike protein, a neoantigen not previously encountered, in both HCWs and NH residents. Multiple antigenic challenges may be required for optimal mRNA vaccine immunogenicity in CMV+ adults., Competing Interests: Potential conflicts of interest. All authors: No reported conflicts of interest., (© The Author(s) 2023. Published by Oxford University Press on behalf of Infectious Diseases Society of America.)- Published
- 2023
- Full Text
- View/download PDF
25. SARS-CoV-2 Antibody Responses to the Ancestral SARS-CoV-2 Strain and Omicron BA.1 and BA.4/BA.5 Variants in Nursing Home Residents After Receipt of Bivalent COVID-19 Vaccine - Ohio and Rhode Island, September-November 2022.
- Author
-
Canaday DH, Oyebanji OA, White EM, Bosch J, Nugent C, Vishnepolskiy I, Abul Y, Didion EM, Paxitzis A, Sundheimer N, Ragavapuram V, Wilk D, Keresztesy D, Cao Y, St Denis K, McConeghy KW, McDonald LC, Jernigan JA, Mylonakis E, Wilson BM, King CL, Balazs AB, and Gravenstein S
- Subjects
- Adult, Humans, SARS-CoV-2, COVID-19 Vaccines, Vaccines, Combined, Rhode Island, Antibody Formation, Ohio, Antibodies, Viral, Nursing Homes, Antibodies, Neutralizing, COVID-19 epidemiology, COVID-19 prevention & control
- Abstract
Introduction of monovalent COVID-19 mRNA vaccines in late 2020 helped to mitigate disproportionate COVID-19-related morbidity and mortality in U.S. nursing homes (1); however, reduced effectiveness of monovalent vaccines during the period of Omicron variant predominance led to recommendations for booster doses with bivalent COVID-19 mRNA vaccines that include an Omicron BA.4/BA.5 spike protein component to broaden immune response and improve vaccine effectiveness against circulating Omicron variants (2). Recent studies suggest that bivalent booster doses provide substantial additional protection against SARS-CoV-2 infection and severe COVID-19-associated disease among immunocompetent adults who previously received only monovalent vaccines (3).* The immunologic response after receipt of bivalent boosters among nursing home residents, who often mount poor immunologic responses to vaccines, remains unknown. Serial testing of anti-spike protein antibody binding and neutralizing antibody titers in serum collected from 233 long-stay nursing home residents from the time of their primary vaccination series and including any subsequent booster doses, including the bivalent vaccine, was performed. The bivalent COVID-19 mRNA vaccine substantially increased anti-spike and neutralizing antibody titers against Omicron sublineages, including BA.1 and BA.4/BA.5, irrespective of previous SARS-CoV-2 infection or previous receipt of 1 or 2 booster doses. These data, in combination with evidence of low uptake of bivalent booster vaccination among residents and staff members in nursing homes (4), support the recommendation that nursing home residents and staff members receive a bivalent COVID-19 booster dose to reduce associated morbidity and mortality (2)., Competing Interests: All authors have completed and submitted the International Committee of Medical Journal Editors form for disclosure of potential conflicts of interest. Stefan Gravenstein and David H. Canaday are recipients of support from the U.S. Department of Veterans Affairs and investigator-initiated grants to their universities from the National Institute of Allergy and Infectious Diseases (NIAID) to study influenza vaccine and COVID-19 in the nursing home, Pfizer to study pneumococcal vaccines, and from Sanofi Pasteur and Seqirus to study influenza vaccines. Stefan Gravenstein also performs consulting work for Janssen, Merck, Moderna, Novavax, Pfizer, Sanofi, Seqirus, and Vaxart; has served on the speaker’s bureaus for Seqirus and Sanofi; and was paid to chair data safety monitoring boards from Longeveron and SciClone. David H. Canaday has performed consulting work for Seqirus. Elizabeth M. White reports support from the National Institute on Aging, and membership on the Society for Post-acute and Long-term Care Medicine Workforce Development Committee and on the John Hartford Foundation Moving Forward Coalition Workforce Committee. Jürgen Bosch is the cofounder and Chief Executive Officer of InterRayBio, LLC. Yi Cao, Kerri St. Denis, and Alejandro B. Balazs report support from the Ragon Institute of Massachusetts General Hospital, the Massachusetts Institute of Technology, and Harvard University for equipment used in the current study. Kevin W. McConeghy reports grant support from Sanofi-Pasteur, Sequirus Pharmaceuticals, Genentech, and Janssen, unrelated to the current work. Eleftherios Mylonakis reports institutional support from the Biomedical Advanced Research and Development Authority, NIAID, the National Institute of General Medical Sciences, National Institutes of Health, Leidos Biomedical Research, Inc., Regeneron, Pfizer, Chemic lags/KODA therapeutics, Cidara, the National Cancer Institute, and SciClone Pharmaceuticals, and receipt of consulting fees from Basilea Pharmaceutica International, Ltd. Christopher L. King reports National Cancer Institute support for Early Drivers of Humoral Immunity to SARS-CoV-2 Infections. No other potential conflicts of interest were disclosed.
- Published
- 2023
- Full Text
- View/download PDF
26. COVID-19 booster dose induces robust antibody response in pregnant, lactating, and nonpregnant women.
- Author
-
Atyeo C, Shook LL, Nziza N, Deriso EA, Muir C, Baez AM, Lima RS, Demidkin S, Brigida S, De Guzman RM, Burns MD, Balazs AB, Fasano A, Yonker LM, Gray KJ, Alter G, and Edlow AG
- Subjects
- Infant, Newborn, Pregnancy, Female, Humans, BNT162 Vaccine, COVID-19 Vaccines, Lactation, SARS-CoV-2, Immunoglobulin G, Antibodies, Viral, Antibody Formation, COVID-19
- Abstract
Background: Although emerging data during the SARS-CoV-2 pandemic have demonstrated robust messenger RNA vaccine-induced immunogenicity across populations, including pregnant and lactating individuals, the rapid waning of vaccine-induced immunity and the emergence of variants of concern motivated the use of messenger RNA vaccine booster doses. Whether all populations, including pregnant and lactating individuals, will mount a comparable response to a booster dose is not known., Objective: This study aimed to profile the humoral immune response to a COVID-19 messenger RNA booster dose in a cohort of pregnant, lactating, and nonpregnant age-matched women., Study Design: This study characterized the antibody response against ancestral Spike and Omicron in a cohort of 31 pregnant, 12 lactating, and 20 nonpregnant age-matched controls who received a BNT162b2 or messenger RNA-1273 booster dose after primary COVID-19 vaccination. In addition, this study examined the vaccine-induced antibody profiles of 15 maternal-to-cord dyads at delivery., Results: Receiving a booster dose during pregnancy resulted in increased immunoglobulin G1 levels against Omicron Spike (postprimary vaccination vs postbooster dose; P=.03). Pregnant and lactating individuals exhibited equivalent Spike-specific total immunoglobulin G1, immunoglobulin M, and immunoglobulin A levels and neutralizing titers against Omicron compared with nonpregnant women. Subtle differences in Fc receptor binding and antibody subclass profiles were observed in the immune response to a booster dose in pregnant vs nonpregnant individuals. The analysis of maternal and cord antibody profiles at delivery demonstrated equivalent total Spike-specific immunoglobulin G1 in maternal and cord blood, yet higher Spike-specific FcγR3a-binding antibodies in the cord relative to maternal blood (P=.002), consistent with the preferential transfer of highly functional immunoglobulin. Spike-specific immunoglobulin G1 levels in the cord were positively correlated with the time elapsed since receiving the booster dose (Spearman R, .574; P=.035)., Conclusion: Study data suggested that receiving a booster dose during pregnancy induces a robust Spike-specific humoral immune response, including against Omicron. If boosting occurs in the third trimester of pregnancy, higher Spike-specific cord immunoglobulin G1 levels are achieved with greater time elapsed between receiving the booster and delivery. Receiving a booster dose has the potential to augment maternal and neonatal immunity., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
27. Cross-reactive SARS-CoV-2 epitope targeted across donors informs immunogen design.
- Author
-
Hauser BM, Feldman J, Sangesland M, Ronsard L, St Denis KJ, Sheehan ML, Cao Y, Boucau J, Windsor IW, Cheng AH, Vu ML, Cardoso MR, Kannegieter T, Balazs AB, Lingwood D, Garcia-Beltran WF, and Schmidt AG
- Subjects
- Animals, Humans, Mice, Epitopes genetics, COVID-19 Vaccines, Antibodies, SARS-CoV-2 genetics, COVID-19 prevention & control
- Abstract
The emergence of the antigenically distinct and highly transmissible Omicron variant highlights the possibility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immune escape due to viral evolution. This continued evolution, along with the possible introduction of new sarbecoviruses from zoonotic reservoirs, may evade host immunity elicited by current SARS-CoV-2 vaccines. Identifying cross-reactive antibodies and defining their epitope(s) can provide templates for rational immunogen design strategies for next-generation vaccines. Here, we characterize the receptor-binding-domain-directed, cross-reactive humoral repertoire across 10 human vaccinated donors. We identify cross-reactive antibodies from diverse gene rearrangements targeting two conserved receptor-binding domain epitopes. An engineered immunogen enriches antibody responses to one of these conserved epitopes in mice with pre-existing SARS-CoV-2 immunity; elicited responses neutralize SARS-CoV-2, variants, and related sarbecoviruses. These data show how immune focusing to a conserved epitope targeted by human cross-reactive antibodies may guide pan-sarbecovirus vaccine development, providing a template for identifying such epitopes and translating to immunogen design., Competing Interests: Declaration of interests B.M.H. and A.G.S. have filed a provisional patent for the described immunogens., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
28. Maternally transferred mAbs protect neonatal mice from HSV-induced mortality and morbidity.
- Author
-
Backes IM, Byrd BK, Slein MD, Patel CD, Taylor SA, Garland CR, MacDonald SW, Balazs AB, Davis SC, Ackerman ME, and Leib DA
- Subjects
- Animals, Animals, Newborn, Antibodies, Monoclonal therapeutic use, Antibodies, Viral, Antiviral Agents, Glycoproteins, Humans, Mice, Morbidity, Pregnancy Complications, Infectious, Herpes Simplex
- Abstract
Neonatal herpes simplex virus (nHSV) infections often result in significant mortality and neurological morbidity despite antiviral drug therapy. Maternally transferred herpes simplex virus (HSV)-specific antibodies reduce the risk of clinically overt nHSV, but this observation has not been translationally applied. Using a neonatal mouse model, we tested the hypothesis that passive transfer of HSV-specific human mAbs can prevent mortality and morbidity associated with nHSV. The mAbs were expressed in vivo via vectored immunoprophylaxis or recombinantly. Through these maternally derived routes or through direct administration to pups, diverse mAbs to HSV glycoprotein D protected against neonatal HSV-1 and HSV-2 infection. Using in vivo bioluminescent imaging, both pre- and post-exposure mAb treatment significantly reduced viral load in mouse pups. Together these studies support the notion that HSV-specific mAb-based therapies could prevent or improve HSV infection outcomes in neonates., (© 2022 Backes et al.)
- Published
- 2022
- Full Text
- View/download PDF
29. BNT162b2 induces robust cross-variant SARS-CoV-2 immunity in children.
- Author
-
Bartsch YC, Chen JW, Kang J, Burns MD, St Denis KJ, Sheehan ML, Davis JP, Edlow AG, Balazs AB, Yonker LM, and Alter G
- Abstract
Currently available mRNA vaccines are extremely safe and effective to prevent severe SARS-CoV-2 infections. However, the emergence of variants of concerns (VOCs) has highlighted the importance of high population-based vaccine rates to effectively suppress viral transmission and breakthrough infections. While initially left out from vaccine efforts, children have become one of the most affected age groups and are key targets to stop community and household spread. Antibodies are central for vaccine-induced protection and emerging data points to the importance of additional Fc effector functions like opsononophagocytosis or cytotoxicity, particularly in the context of VOCs that escape neutralizing antibodies. Here, we observed delayed induction and reduced magnitude of vaccine-induced antibody titers in children 5-11 years receiving two doses of the age-recommended 10 μg dose of the Pfizer SARS-CoV-2 BNT162b2 vaccine compared to adolescents (12-15 years) or adults receiving the 30 μg dose. Conversely, children mounted equivalent or more robust neutralization and opsonophagocytic functions at peak immunogenicity, pointing to a qualitatively more robust humoral functional response in children. Moreover, broad cross-VOC responses were observed across children, with enhanced IgM and parallel IgG cross-reactivity to VOCs in children compared to adults. Collectively, these data argue that despite the lower magnitude of the BNT162b2-induced antibody response in children, vaccine-induced immunity in children target VOCs broadly and exhibit enhanced functionality that may contribute to the attenuation of disease., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
30. SARS-CoV-2 mRNA vaccination elicits robust antibody responses in children.
- Author
-
Bartsch YC, St Denis KJ, Kaplonek P, Kang J, Lam EC, Burns MD, Farkas EJ, Davis JP, Boribong BP, Edlow AG, Fasano A, Shreffler WG, Zavadska D, Johnson M, Goldblatt D, Balazs AB, Yonker LM, and Alter G
- Subjects
- Child, Humans, 2019-nCoV Vaccine mRNA-1273, Antibodies, Viral, Receptors, Fc, SARS-CoV-2, Vaccination, Antibody Formation, COVID-19 prevention & control, COVID-19 Vaccines immunology
- Abstract
Although children have been largely spared from coronavirus disease 2019 (COVID-19), the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) with increased transmissibility, combined with fluctuating mask mandates and school reopenings, has led to increased infections and disease among children. Thus, there is an urgent need to roll out COVID-19 vaccines to children of all ages. However, whether children respond equivalently to adults to mRNA vaccines and whether dosing will elicit optimal immunity remain unclear. Here, we aimed to deeply profile the vaccine-induced humoral immune response in 6- to 11-year-old children receiving either a pediatric (50 μg) or adult (100 μg) dose of the mRNA-1273 vaccine and to compare these responses to vaccinated adults, infected children, and children who experienced multisystem inflammatory syndrome in children (MIS-C). Children elicited an IgG-dominant vaccine-induced immune response, surpassing adults at a matched 100-μg dose but more variable immunity at a 50-μg dose. Irrespective of titer, children generated antibodies with enhanced Fc receptor binding capacity. Moreover, like adults, children generated cross-VOC humoral immunity, marked by a decline of omicron-specific receptor binding domain, but robustly preserved omicron spike protein binding. Fc receptor binding capabilities were also preserved in a dose-dependent manner. These data indicate that both the 50- and 100-μg doses of mRNA vaccination in children elicit robust cross-VOC antibody responses and that 100-μg doses in children result in highly preserved omicron-specific functional humoral immunity.
- Published
- 2022
- Full Text
- View/download PDF
31. Significant Reduction in Vaccine-Induced Antibody Levels and Neutralization Activity Among Healthcare Workers and Nursing Home Residents 6 Months Following Coronavirus Disease 2019 BNT162b2 mRNA Vaccination.
- Author
-
Canaday DH, Oyebanji OA, Keresztesy D, Payne M, Wilk D, Carias L, Aung H, St Denis K, Lam EC, Rowley CF, Berry SD, Cameron CM, Cameron MJ, Wilson BM, Balazs AB, King CL, and Gravenstein S
- Subjects
- Antibodies, Neutralizing, Antibodies, Viral, BNT162 Vaccine, Health Personnel, Humans, Nursing Homes, RNA, Messenger, Vaccination, COVID-19 prevention & control, Influenza Vaccines
- Abstract
Antibody decline occurred from 2 weeks to 6 months post-BNT162b2 mRNA vaccination in nursing home (NH) residents and healthcare workers. Antispike, receptor-binding domain, and neutralization levels dropped >81% irrespective of prior infection. Notably, 69% of infection-naive NH residents had neutralizing antibodies at or below the assay's limit of detection., (© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America.)
- Published
- 2022
- Full Text
- View/download PDF
32. Humoral responses to the SARS-CoV-2 spike and receptor binding domain in context of pre-existing immunity confer broad sarbecovirus neutralization.
- Author
-
Hauser BM, Sangesland M, Lam EC, Feldman J, Balazs AB, Lingwood D, and Schmidt AG
- Subjects
- Antibodies, Viral, Epitopes, Humans, Spike Glycoprotein, Coronavirus, COVID-19 prevention & control, SARS-CoV-2
- Abstract
Since the emergence of SARS-CoV-2 (SARS-2), multiple vaccine candidates were developed and studied both preclinically and clinically. Nearly all are based on the SARS-2 spike glycoprotein or its receptor binding domain (RBD). Studies of these vaccine candidates have largely been in a SARS-2 naïve context. However, pre-existing immunity to SARS-2 acquired through infection or vaccination continues to increase. Evaluating future vaccine candidates in context of this pre-existing immunity is necessary to understand how immune responses are subsequently influenced. Here, we evaluated the serum and IgG
+ B cell responses to the SARS-2 RBD in context of pre-existing immunity elicited by the full SARS-2 spike, and we compared this to boosting with the full SARS-2 spike. Boosting with the SARS-2 RBD resulted in increased reactivity to RBD epitopes, but both immunization regimens resulted in similarly broad neutralization across diverse sarbecoviruses. These findings may inform comparison among SARS-2 RBD-based vaccine candidates to currently approved spike-based candidates., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Hauser, Sangesland, Lam, Feldman, Balazs, Lingwood and Schmidt.)- Published
- 2022
- Full Text
- View/download PDF
33. Antibody-mediated prevention of vaginal HIV transmission is dictated by IgG subclass in humanized mice.
- Author
-
Brady JM, Phelps M, MacDonald SW, Lam EC, Nitido A, Parsons D, Boutros CL, Deal CE, Garcia-Beltran WF, Tanno S, Natarajan H, Ackerman ME, Vrbanac VD, and Balazs AB
- Subjects
- Animals, Antibodies, Neutralizing, Broadly Neutralizing Antibodies, Female, HIV Antibodies, Mice, Vagina, HIV Infections prevention & control, Immunoglobulin G
- Abstract
HIV broadly neutralizing antibodies (bNAbs) are capable of both blocking viral entry and driving innate immune responses against HIV-infected cells through their Fc region. Vaccination or productive infection results in a polyclonal mixture of class-switched immunoglobulin G (IgG) antibodies composed of four subclasses, each encoding distinct Fc regions that differentially engage innate immune functions. Despite evidence that innate immunity contributes to protection, the relative contribution of individual IgG subclasses is unknown. Here, we used vectored immunoprophylaxis in humanized mice to interrogate the efficacy of individual IgG subclasses during prevention of vaginal HIV transmission by VRC07, a potent CD4-binding site-directed bNAb. We find that VRC07 IgG2, which lacks Fc-mediated functionality, exhibited substantially reduced protection in vivo relative to other subclasses. Low concentrations of highly functional VRC07 IgG1 yielded substantial protection against vaginal challenge, suggesting that interventions capable of eliciting modest titers of functional IgG subclasses may provide meaningful benefit against infection.
- Published
- 2022
- Full Text
- View/download PDF
34. Maternal immune response and placental antibody transfer after COVID-19 vaccination across trimester and platforms.
- Author
-
Atyeo CG, Shook LL, Brigida S, De Guzman RM, Demidkin S, Muir C, Akinwunmi B, Baez AM, Sheehan ML, McSweeney E, Burns MD, Nayak R, Kumar MK, Patel CD, Fialkowski A, Cvrk D, Goldfarb IT, Yonker LM, Fasano A, Balazs AB, Elovitz MA, Gray KJ, Alter G, and Edlow AG
- Subjects
- Ad26COVS1, Antibodies, Viral, BNT162 Vaccine, COVID-19 Vaccines, Female, Humans, Immunity, Infant, Newborn, Placenta, Pregnancy, SARS-CoV-2, United States, Vaccination methods, COVID-19 prevention & control, Pregnancy Complications, Infectious prevention & control
- Abstract
The availability of three COVID-19 vaccines in the United States provides an unprecedented opportunity to examine how vaccine platforms and timing of vaccination in pregnancy impact maternal and neonatal immunity. Here, we characterize the antibody profile after Ad26.COV2.S, mRNA-1273 or BNT162b2 vaccination in 158 pregnant individuals and evaluate transplacental antibody transfer by profiling maternal and umbilical cord blood in 175 maternal-neonatal dyads. These analyses reveal lower vaccine-induced functions and Fc receptor-binding after Ad26.COV2.S compared to mRNA vaccination and subtle advantages in titer and function with mRNA-1273 versus BN162b2. mRNA vaccines have higher titers and functions against SARS-CoV-2 variants of concern. First and third trimester vaccination results in enhanced maternal antibody-dependent NK-cell activation, cellular and neutrophil phagocytosis, and complement deposition relative to second trimester. Higher transplacental transfer ratios following first and second trimester vaccination may reflect placental compensation for waning maternal titers. These results provide novel insight into the impact of platform and trimester of vaccination on maternal humoral immune response and transplacental antibody transfer., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
35. COVID-19 vaccine booster dose needed to achieve Omicron-specific neutralisation in nursing home residents.
- Author
-
Canaday DH, Oyebanji OA, White E, Keresztesy D, Payne M, Wilk D, Carias L, Aung H, St Denis K, Sheehan ML, Berry SD, Cameron CM, Cameron MJ, Wilson BM, Balazs AB, King CL, and Gravenstein S
- Subjects
- Aged, Antibodies, Viral, BNT162 Vaccine, Humans, Immunization, Secondary, Middle Aged, Nursing Homes, SARS-CoV-2, Vaccines, Synthetic, mRNA Vaccines, COVID-19 prevention & control, COVID-19 Vaccines
- Abstract
Background: Nursing home (NH) residents have borne a disproportionate share of SARS-CoV-2 morbidity and mortality. Vaccines have limited hospitalisation and death from earlier variants in this vulnerable population. With the rise of Omicron and future variants, it is vital to sustain and broaden vaccine-induced protection. We examined the effect of boosting with BNT162b2 mRNA vaccine on humoral immunity and Omicron-specific neutralising activity among NH residents and healthcare workers (HCWs)., Methods: We longitudinally enrolled 85 NH residents (median age 77) and 48 HCWs (median age 51), and sampled them after the initial vaccination series; and just before and 2 weeks after booster vaccination. Anti-spike, anti-receptor binding domain (RBD) and neutralisation titres to the original Wuhan strain and neutralisation to the Omicron strain were obtained., Findings: Booster vaccination significantly increased vaccine-specific anti-spike, anti-RBD, and neutralisation levels above the pre-booster levels in NH residents and HCWs, both in those with and without prior SARS-CoV-2 infection. Omicron-specific neutralisation activity was low after the initial 2 dose series with only 28% of NH residents' and 28% HCWs' titres above the assay's lower limit of detection. Omicron neutralising activity following the booster lifted 86% of NH residents and 93% of HCWs to the detectable range., Interpretation: With boosting, the vast majority of HCWs and NH residents developed detectable Omicron-specific neutralising activity. These data provide immunologic evidence that strongly supports booster vaccination to broaden neutralising activity and counter waning immunity in the hope it will better protect this vulnerable, high-risk population against the Omicron variant., Funding: NIH AI129709-03S1, U01 CA260539-01, CDC 200-2016-91773, and VA BX005507-01., (Published by Elsevier B.V.)
- Published
- 2022
- Full Text
- View/download PDF
36. Safety and tolerability of AAV8 delivery of a broadly neutralizing antibody in adults living with HIV: a phase 1, dose-escalation trial.
- Author
-
Casazza JP, Cale EM, Narpala S, Yamshchikov GV, Coates EE, Hendel CS, Novik L, Holman LA, Widge AT, Apte P, Gordon I, Gaudinski MR, Conan-Cibotti M, Lin BC, Nason MC, Trofymenko O, Telscher S, Plummer SH, Wycuff D, Adams WC, Pandey JP, McDermott A, Roederer M, Sukienik AN, O'Dell S, Gall JG, Flach B, Terry TL, Choe M, Shi W, Chen X, Kaltovich F, Saunders KO, Stein JA, Doria-Rose NA, Schwartz RM, Balazs AB, Baltimore D, Nabel GJ, Koup RA, Graham BS, Ledgerwood JE, and Mascola JR
- Subjects
- Adult, Antibodies, Neutralizing, Broadly Neutralizing Antibodies, Dependovirus genetics, HIV Antibodies, Humans, HIV Infections drug therapy, HIV-1
- Abstract
Adeno-associated viral vector-mediated transfer of DNA coding for broadly neutralizing anti-HIV antibodies (bnAbs) offers an alternative to attempting to induce protection by vaccination or by repeated infusions of bnAbs. In this study, we administered a recombinant bicistronic adeno-associated virus (AAV8) vector coding for both the light and heavy chains of the potent broadly neutralizing HIV-1 antibody VRC07 (AAV8-VRC07) to eight adults living with HIV. All participants remained on effective anti-retroviral therapy (viral load (VL) <50 copies per milliliter) throughout this phase 1, dose-escalation clinical trial ( NCT03374202 ). AAV8-VRC07 was given at doses of 5 × 10
10 , 5 × 1011 and 2.5 × 1012 vector genomes per kilogram by intramuscular (IM) injection. Primary endpoints of this study were to assess the safety and tolerability of AAV8-VRC07; to determine the pharmacokinetics and immunogenicity of in vivo VRC07 production; and to describe the immune response directed against AAV8-VRC07 vector and its products. Secondary endpoints were to assess the clinical effects of AAV8-VRC07 on CD4 T cell count and VL and to assess the persistence of VRC07 produced in participants. In this cohort, IM injection of AAV8-VRC07 was safe and well tolerated. No clinically significant change in CD4 T cell count or VL occurred during the 1-3 years of follow-up reported here. In participants who received AAV8-VRC07, concentrations of VRC07 were increased 6 weeks (P = 0.008) and 52 weeks (P = 0.016) after IM injection of the product. All eight individuals produced measurable amounts of serum VRC07, with maximal VRC07 concentrations >1 µg ml-1 in three individuals. In four individuals, VRC07 serum concentrations remained stable near maximal concentration for up to 3 years of follow-up. In exploratory analyses, neutralizing activity of in vivo produced VRC07 was similar to that of in vitro produced VRC07. Three of eight participants showed a non-idiotypic anti-drug antibody (ADA) response directed against the Fab portion of VRC07. This ADA response appeared to decrease the production of serum VRC07 in two of these three participants. These data represent a proof of concept that adeno-associated viral vectors can durably produce biologically active, difficult-to-induce bnAbs in vivo, which could add valuable new tools to the fight against infectious diseases., (© 2022. This is a U.S. government work and not under copyright protection in the U.S.; foreign copyright protection may apply.)- Published
- 2022
- Full Text
- View/download PDF
37. Humanized mice reveal a macrophage-enriched gene signature defining human lung tissue protection during SARS-CoV-2 infection.
- Author
-
Kenney DJ, O'Connell AK, Turcinovic J, Montanaro P, Hekman RM, Tamura T, Berneshawi AR, Cafiero TR, Al Abdullatif S, Blum B, Goldstein SI, Heller BL, Gertje HP, Bullitt E, Trachtenberg AJ, Chavez E, Nono ET, Morrison C, Tseng AE, Sheikh A, Kurnick S, Grosz K, Bosmann M, Ericsson M, Huber BR, Saeed M, Balazs AB, Francis KP, Klose A, Paragas N, Campbell JD, Connor JH, Emili A, Crossland NA, Ploss A, and Douam F
- Subjects
- Animals, Disease Models, Animal, Humans, Immunity, Innate, Lung pathology, Macrophages, Mice, SARS-CoV-2, COVID-19 genetics
- Abstract
The human immunological mechanisms defining the clinical outcome of SARS-CoV-2 infection remain elusive. This knowledge gap is mostly driven by the lack of appropriate experimental platforms recapitulating human immune responses in a controlled human lung environment. Here, we report a mouse model (i.e., HNFL mice) co-engrafted with human fetal lung xenografts (fLX) and a myeloid-enhanced human immune system to identify cellular and molecular correlates of lung protection during SARS-CoV-2 infection. Unlike mice solely engrafted with human fLX, HNFL mice are protected against infection, severe inflammation, and histopathological phenotypes. Lung tissue protection from infection and severe histopathology associates with macrophage infiltration and differentiation and the upregulation of a macrophage-enriched signature composed of 11 specific genes mainly associated with the type I interferon signaling pathway. Our work highlights the HNFL model as a transformative platform to investigate, in controlled experimental settings, human myeloid immune mechanisms governing lung tissue protection during SARS-CoV-2 infection., Competing Interests: Declaration of interests K.P.F. is an employee of PerkinElmer, Inc., and a technology advisor of InVivo Analytics. N.P. and A.K. are shareholders of InVivo Analytics with issued patents., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
38. Comparative Immunogenicity and Effectiveness of mRNA-1273, BNT162b2, and Ad26.COV2.S COVID-19 Vaccines.
- Author
-
Naranbhai V, Garcia-Beltran WF, Chang CC, Berrios Mairena C, Thierauf JC, Kirkpatrick G, Onozato ML, Cheng J, St Denis KJ, Lam EC, Kaseke C, Tano-Menka R, Yang D, Pavlovic M, Yang W, Kui A, Miller TE, Astudillo MG, Cahill JE, Dighe AS, Gregory DJ, Poznansky MC, Gaiha GD, Balazs AB, and Iafrate AJ
- Subjects
- 2019-nCoV Vaccine mRNA-1273, Adult, BNT162 Vaccine, COVID-19 Vaccines, Humans, Immunogenicity, Vaccine, SARS-CoV-2 genetics, Vaccines, Synthetic, mRNA Vaccines, Ad26COVS1, COVID-19 prevention & control
- Abstract
Background: Understanding immunogenicity and effectiveness of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is critical to guide rational use., Methods: We compared the immunogenicity of mRNA-1273, BNT-162b2, and Ad26.COV2.S in healthy ambulatory adults. We performed an inverse-variance meta-analysis of population-level effectiveness from public health reports in > 40 million individuals., Results: A single dose of either mRNA vaccine yielded comparable antibody and neutralization titers to convalescent individuals. Ad26.COV2.S yielded lower antibody concentrations and frequently undetectable neutralization titers. Bulk and cytotoxic T-cell responses were higher in mRNA1273 and BNT162b2 than Ad26.COV2.S recipients. Regardless of vaccine, <50% of vaccinees demonstrated CD8+ T-cell responses. Antibody concentrations and neutralization titers increased comparably after the first dose of either vaccine, and further in recipients of a second dose. Prior infection was associated with high antibody concentrations and neutralization even after a single dose and regardless of vaccine. Neutralization of Beta, Gamma, and Delta strains were poorer regardless of vaccine. In meta-analysis, relative to mRNA1273 the effectiveness of BNT162b2 was lower against infection and hospitalization, and Ad26COV2.S was lower against infection, hospitalization, and death., Conclusions: Variation in the immunogenicity correlates with variable effectiveness of the 3 vaccines deployed in the United States., (© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America. All rights reserved. For permissions, e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
39. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all individuals.
- Author
-
Naranbhai V, Nathan A, Kaseke C, Berrios C, Khatri A, Choi S, Getz MA, Tano-Menka R, Ofoman O, Gayton A, Senjobe F, Zhao Z, St Denis KJ, Lam EC, Carrington M, Garcia-Beltran WF, Balazs AB, Walker BD, Iafrate AJ, and Gaiha GD
- Published
- 2022
- Full Text
- View/download PDF
40. Durability and Cross-Reactivity of SARS-CoV-2 mRNA Vaccine in Adolescent Children.
- Author
-
Burns MD, Boribong BP, Bartsch YC, Loiselle M, St Denis KJ, Sheehan ML, Chen JW, Davis JP, Lima R, Edlow AG, Fasano A, Balazs AB, Alter G, and Yonker LM
- Abstract
Emergent SARS-CoV-2 variants and waning humoral immunity in vaccinated individuals have resulted in increased infections and hospitalizations. Children are not spared from infection nor complications of COVID-19, and the recent recommendation for boosters in individuals ages 12 years or older calls for broader understanding of the adolescent immune profile after mRNA vaccination. We tested the durability and cross-reactivity of anti-SARS-CoV-2 serologic responses over a six-month time course in vaccinated adolescents against the SARS-CoV-2 D614G ("wild type") and Omicron antigens. Serum from 77 adolescents showed that anti-Spike antibodies wane significantly over six months. After completion of a two-vaccine series, cross-reactivity against Omicron-specific receptor-binding domain (RBD) was seen. Functional humoral activation against wild type and Omicron SARS-CoV-2 also declines over time in vaccinated adolescent children. Evidence of waning mRNA-induced vaccine immunity underscores vulnerabilities in long-term pediatric protection against SARS-CoV-2 infection, while cross-reactivity highlights the additional benefits of vaccination. Characterization of adolescent immune signatures post-vaccination will inform guidance on vaccine platforms and timelines, and ultimately optimize immunoprotection of children.
- Published
- 2022
- Full Text
- View/download PDF
41. Rationally designed immunogens enable immune focusing following SARS-CoV-2 spike imprinting.
- Author
-
Hauser BM, Sangesland M, St Denis KJ, Lam EC, Case JB, Windsor IW, Feldman J, Caradonna TM, Kannegieter T, Diamond MS, Balazs AB, Lingwood D, and Schmidt AG
- Subjects
- Animals, Mice, Mice, Inbred BALB C, SARS-CoV-2, Spike Glycoprotein, Coronavirus, COVID-19, Viral Envelope Proteins
- Abstract
Eliciting antibodies to surface-exposed viral glycoproteins can generate protective responses that control and prevent future infections. Targeting conserved sites may reduce the likelihood of viral escape and limit the spread of related viruses with pandemic potential. Here we leverage rational immunogen design to focus humoral responses on conserved epitopes. Using glycan engineering and epitope scaffolding in boosting immunogens, we focus murine serum antibody responses to conserved receptor binding motif (RBM) and receptor binding domain (RBD) epitopes following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike imprinting. Although all engineered immunogens elicit a robust SARS-CoV-2-neutralizing serum response, RBM-focusing immunogens exhibit increased potency against related sarbecoviruses, SARS-CoV, WIV1-CoV, RaTG13-CoV, and SHC014-CoV; structural characterization of representative antibodies defines a conserved epitope. RBM-focused sera confer protection against SARS-CoV-2 challenge. Thus, RBM focusing is a promising strategy to elicit breadth across emerging sarbecoviruses without compromising SARS-CoV-2 protection. These engineering strategies are adaptable to other viral glycoproteins for targeting conserved epitopes., Competing Interests: Declaration of interests B.M.H., T.M.C., and A.G.S. have filed a provisional patent for the described immunogens. M.S.D. is a consultant for Inbios, Vir Biotechnology, and Carnival Corporation and on the Scientific Advisory Boards of Moderna and Immunome. The Diamond laboratory has received unrelated funding support in sponsored research agreements from Vir Biotechnology, Moderna, and Emergent BioSolutions., (Copyright © 2022 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
42. Multi-modal profiling of human fetal liver hematopoietic stem cells reveals the molecular signature of engraftment.
- Author
-
Vanuytsel K, Villacorta-Martin C, Lindstrom-Vautrin J, Wang Z, Garcia-Beltran WF, Vrbanac V, Parsons D, Lam EC, Matte TM, Dowrey TW, Kumar SS, Li M, Wang F, Yeung AK, Mostoslavsky G, Dries R, Campbell JD, Belkina AC, Balazs AB, and Murphy GJ
- Subjects
- Hematopoietic Stem Cells metabolism, Humans, Liver, Hematopoietic Stem Cell Transplantation
- Abstract
The human hematopoietic stem cell harbors remarkable regenerative potential that can be harnessed therapeutically. During early development, hematopoietic stem cells in the fetal liver undergo active expansion while simultaneously retaining robust engraftment capacity, yet the underlying molecular program responsible for their efficient engraftment remains unclear. Here, we profile 26,407 fetal liver cells at both the transcriptional and protein level including ~7,000 highly enriched and functional fetal liver hematopoietic stem cells to establish a detailed molecular signature of engraftment potential. Integration of transcript and linked cell surface marker expression reveals a generalizable signature defining functional fetal liver hematopoietic stem cells and allows for the stratification of enrichment strategies with high translational potential. More precisely, our integrated analysis identifies CD201 (endothelial protein C receptor (EPCR), encoded by PROCR) as a marker that can specifically enrich for engraftment potential. This comprehensive, multi-modal profiling of engraftment capacity connects a critical biological function at a key developmental timepoint with its underlying molecular drivers. As such, it serves as a useful resource for the field and forms the basis for further biological exploration of strategies to retain the engraftment potential of hematopoietic stem cells ex vivo or induce this potential during in vitro hematopoietic stem cell generation., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
43. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant.
- Author
-
Garcia-Beltran WF, St Denis KJ, Hoelzemer A, Lam EC, Nitido AD, Sheehan ML, Berrios C, Ofoman O, Chang CC, Hauser BM, Feldman J, Roederer AL, Gregory DJ, Poznansky MC, Schmidt AG, Iafrate AJ, Naranbhai V, and Balazs AB
- Abstract
Recent surveillance has revealed the emergence of the SARS-CoV-2 Omicron variant (BA.1/B.1.1.529) harboring up to 36 mutations in spike protein, the target of neutralizing antibodies. Given its potential to escape vaccine-induced humoral immunity, we measured the neutralization potency of sera from 88 mRNA-1273, 111 BNT162b, and 40 Ad26.COV2.S vaccine recipients against wild-type, Delta, and Omicron SARS-CoV-2 pseudoviruses. We included individuals that received their primary series recently (<3 months), distantly (6-12 months), or an additional "booster" dose, while accounting for prior SARS-CoV-2 infection. Remarkably, neutralization of Omicron was undetectable in most vaccinees. However, individuals boosted with mRNA vaccines exhibited potent neutralization of Omicron, only 4-6-fold lower than wild type, suggesting enhanced cross-reactivity of neutralizing antibody responses. In addition, we find that Omicron pseudovirus infects more efficiently than other variants tested. Overall, this study highlights the importance of additional mRNA doses to broaden neutralizing antibody responses against highly divergent SARS-CoV-2 variants., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
44. Neutralization breadth of SARS-CoV-2 viral variants following primary series and booster SARS-CoV-2 vaccines in patients with cancer.
- Author
-
Naranbhai V, St Denis KJ, Lam EC, Ofoman O, Garcia-Beltran WF, Mairena CB, Bhan AK, Gainor JF, Balazs AB, and Iafrate AJ
- Subjects
- Aged, Aging immunology, Antigens, Viral immunology, Female, Humans, Immunization, Secondary, Immunocompromised Host, Immunogenicity, Vaccine, In Vitro Techniques, Male, Middle Aged, Neoplasms therapy, Spike Glycoprotein, Coronavirus immunology, Viral Load, Antibodies, Neutralizing biosynthesis, Antibodies, Viral biosynthesis, COVID-19 prevention & control, COVID-19 Vaccines immunology, Neoplasms immunology, SARS-CoV-2 immunology
- Abstract
Patients with cancer are more likely to have impaired immune responses to SARS-CoV-2 vaccines. We study the breadth of responses against SARS-CoV-2 variants after primary vaccination in 178 patients with a variety of tumor types and after booster doses in a subset. Neutralization of alpha, beta, gamma, and delta SARS-CoV-2 variants is impaired relative to wildtype, regardless of vaccine type. Regardless of viral variant, mRNA1273 is the most immunogenic, followed by BNT162b2, and then Ad26.COV2.S. Neutralization of more variants (breadth) is associated with a greater magnitude of wildtype neutralization, and increases with time since vaccination; advancing age associates with a lower breadth. The concentrations of anti-spike protein antibody are a good surrogate for breadth (positive predictive value of =90% at >1,000 U/mL). Booster SARS-CoV-2 vaccines confer enhanced breadth. These data suggest that achieving a high antibody titer is desirable to achieve broad neutralization; a single booster dose with the current vaccines increases the breadth of responses against variants., Competing Interests: Declaration of interests J.F.G. has served as a compensated consultant or received honoraria from Bristol-Myers Squibb, Genentech, Ariad/Takeda, Loxo/Lilly, Blueprint, Oncorus, Regeneron, Gilead, Moderna, AstraZeneca, Pfizer, Novartis, Merck, and GlydeBio; research support from Novartis, Genentech/Roche, and Ariad/Takeda; institutional research support from Bristol-Myers Squibb, Tesaro, Moderna, Blueprint, Jounce, Array Biopharma, Merck, Adaptimmune, Novartis, and Alexo; and has an immediate family member who is an employee with equity at Ironwood Pharmaceuticals. A.J.I. has served as a compensated consultant for Oncoclinicas Brasil, Kinnate, Repare, and Paige.ai., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
45. T cell reactivity to the SARS-CoV-2 Omicron variant is preserved in most but not all prior infected and vaccinated individuals.
- Author
-
Naranbhai V, Nathan A, Kaseke C, Berrios C, Khatri A, Choi S, Getz MA, Tano-Menka R, Ofoman O, Gayton A, Senjobe F, Denis KJS, Lam EC, Garcia-Beltran WF, Balazs AB, Walker BD, Iafrate AJ, and Gaiha GD
- Abstract
The SARS-CoV-2 Omicron variant (B.1.1.529) contains mutations that mediate escape from infection and vaccine-induced antibody responses, although the extent to which these substitutions in spike and non-spike proteins affect T cell recognition is unknown. Here we show that T cell responses in individuals with prior infection, vaccination, both prior infection and vaccination, and boosted vaccination are largely preserved to Omicron spike and non-spike proteins. However, we also identify a subset of individuals (∼21%) with a >50% reduction in T cell reactivity to the Omicron spike. Evaluation of functional CD4
+ and CD8+ memory T cell responses confirmed these findings and reveal that reduced recognition to Omicron spike is primarily observed within the CD8+ T cell compartment. Booster vaccination substantially enhanced T cell responses to Omicron spike. In contrast to neutralizing immunity, these findings suggest preservation of T cell responses to the Omicron variant, although with reduced reactivity in some individuals.- Published
- 2022
- Full Text
- View/download PDF
46. Comprehensive antibody profiling of mRNA vaccination in children.
- Author
-
Bartsch YC, St Denis KJ, Kaplonek P, Kang J, Lam EC, Burns MD, Farkas EJ, Davis JP, Boribong BP, Edlow AG, Fasano A, Shreffler W, Zavadska D, Johnson M, Goldblatt D, Balazs AB, Yonker LM, and Alter G
- Abstract
While children have been largely spared from COVID-19 disease, the emergence of viral variants of concern (VOC) with increased transmissibility, combined with fluctuating mask mandates and school re-openings have led to increased infections and disease among children. Thus, there is an urgent need to roll out COVID-19 vaccines to children of all ages. However, whether children respond equivalently to adults to mRNA vaccines and whether dosing will elicit optimal immunity remains unclear. Given the recent announcement of incomplete immunity induced by the pediatric dose of the BNT162b2 vaccine in young children, here we aimed to deeply profile and compare the vaccine-induced humoral immune response in 6-11 year old children receiving the pediatric (50μg) or adult (100μg) dose of the mRNA-1273 vaccine compared to adults and naturally infected children or children that experienced multi inflammatory syndrome in children (MIS-C) for the first time. Children elicited an IgG dominant vaccine induced immune response, surpassing adults at a matched 100μg dose, but more variable immunity at a 50μg dose. Irrespective of titer, children generated antibodies with enhanced Fc-receptor binding capacity. Moreover, like adults, children generated cross-VOC humoral immunity, marked by a decline of omicron receptor binding domain-binding, but robustly preserved omicron Spike-receptor binding, with robustly preserved Fc-receptor binding capabilities, in a dose dependent manner. These data indicate that while both 50μg and 100μg of mRNA vaccination in children elicits robust cross-VOC antibody responses, 100ug of mRNA in children results in highly preserved omicron-specific functional humoral immunity., Competing Interests: Competing interests G.A. is a founder of Seromyx Systems, a company developing a platform technology that describes the antibody immune response. G.A.’s interests were reviewed and are managed by Massachusetts General Hospital and Partners HealthCare in accordance with their conflict of interest policies. All other authors have declared that no conflicts of interest exist.
- Published
- 2022
- Full Text
- View/download PDF
47. Immunogenicity and Reactogenicity of SARS-CoV-2 Vaccines in Patients With Cancer: The CANVAX Cohort Study.
- Author
-
Naranbhai V, Pernat CA, Gavralidis A, St Denis KJ, Lam EC, Spring LM, Isakoff SJ, Farmer JR, Zubiri L, Hobbs GS, How J, Brunner AM, Fathi AT, Peterson JL, Sakhi M, Hambelton G, Denault EN, Mortensen LJ, Perriello LA, Bruno MN, Bertaux BY, Lawless AR, Jackson MA, Niehoff E, Barabell C, Nambu CN, Nakajima E, Reinicke T, Bowes C, Berrios-Mairena CJ, Ofoman O, Kirkpatrick GE, Thierauf JC, Reynolds K, Willers H, Beltran WG, Dighe AS, Saff R, Blumenthal K, Sullivan RJ, Chen YB, Kim A, Bardia A, Balazs AB, Iafrate AJ, and Gainor JF
- Subjects
- Aged, Cohort Studies, Female, Humans, Male, Middle Aged, Prospective Studies, COVID-19 Vaccines immunology, COVID-19 Vaccines therapeutic use, Neoplasms immunology, SARS-CoV-2 immunology
- Abstract
Purpose: The immunogenicity and reactogenicity of SARS-CoV-2 vaccines in patients with cancer are poorly understood., Methods: We performed a prospective cohort study of adults with solid-organ or hematologic cancers to evaluate anti-SARS-CoV-2 immunoglobulin A/M/G spike antibodies, neutralization, and reactogenicity ≥ 7 days following two doses of mRNA-1273, BNT162b2, or one dose of Ad26.COV2.S. We analyzed responses by multivariate regression and included data from 1,638 healthy controls, previously reported, for comparison., Results: Between April and July 2021, we enrolled 1,001 patients; 762 were eligible for analysis (656 had neutralization measured). mRNA-1273 was the most immunogenic (log
10 geometric mean concentration [GMC] 2.9, log10 geometric mean neutralization titer [GMT] 2.3), followed by BNT162b2 (GMC 2.4; GMT 1.9) and Ad26.COV2.S (GMC 1.5; GMT 1.4; P < .001). The proportion of low neutralization (< 20% of convalescent titers) among Ad26.COV2.S recipients was 69.9%. Prior COVID-19 infection (in 7.1% of the cohort) was associated with higher responses ( P < .001). Antibody titers and neutralization were quantitatively lower in patients with cancer than in comparable healthy controls, regardless of vaccine type ( P < .001). Receipt of chemotherapy in the prior year or current steroids were associated with lower antibody levels and immune checkpoint blockade with higher neutralization. Systemic reactogenicity varied by vaccine and correlated with immune responses ( P = .002 for concentration, P = .016 for neutralization). In 32 patients who received an additional vaccine dose, side effects were similar to prior doses, and 30 of 32 demonstrated increased antibody titers (GMC 1.05 before additional dose, 3.17 after dose)., Conclusion: Immune responses to SARS-CoV-2 vaccines are modestly impaired in patients with cancer. These data suggest utility of antibody testing to identify patients for whom additional vaccine doses may be effective and appropriate, although larger prospective studies are needed., Competing Interests: Laura M. SpringConsulting or Advisory Role: Novartis, AvrobioResearch Funding: Tesaro (Inst), Merck (Inst)Travel, Accommodations, Expenses: Merck, Tesaro Steven J. IsakoffConsulting or Advisory Role: AbbVie, OncoPep, Puma Biotechnology, Seattle Genetics, Novartis, Paxman Coolers LtdResearch Funding: Genentech (Inst), PharmaMar (Inst), AbbVie (Inst), OncoPep (Inst), Merck (Inst), AstraZeneca/MedImmune (Inst), Outcomes4Me (Inst) Jocelyn R. FarmerConsulting or Advisory Role: Bristol Myers Squibb FoundationResearch Funding: Bristol Myers Squibb Foundation Leyre ZubiriConsulting or Advisory Role: Merck Gabriela S. HobbsConsulting or Advisory Role: Incyte, AbbVie, Novatis, Blueprint Medicines, Keros TherapeuticsResearch Funding: Incyte, Constellation Pharmaceuticals Andrew M. BrunnerConsulting or Advisory Role: Celgene, Novartis, Takeda, Agios, Bristol Myers Squibb/Celgene, Acceleron PharmaResearch Funding: Celgene, Takeda, Novartis, GlaxoSmithKline, AstraZeneca Amir T. FathiConsulting or Advisory Role: Agios, Novartis, Takeda, Astellas Pharma, Daiichi Sankyo, Bristol Myers Squibb, Forty Seven, AbbVie, Kite, a Gilead Company, Trovagene, Pfizer, Seattle Genetics, Amgen, Trillium Therapeutics, Blueprint Medicines, Kura Oncology, Foghorn Therapeutics, Genentech, Ipsen, MorphoSys, ServierResearch Funding: Takeda (Inst), Agios (Inst), Bristol Myers Squibb (Inst), AbbVie (Inst), Servier (Inst) Brittany Y. BertauxEmployment: Partners (I) Elizabeth NiehoffStock and Other Ownership Interests: Biogen Inc, Blueprint Medicines, Crispr Therapeutics, InVitae, LabCorp, Natera Inc, Pacific Biosciences Christian N. NambuEmployment: Massachusetts General Hospital Cancer Center, AFC Urgent Care (I)Stock and Other Ownership Interests: Moderna Therapeutics Onosereme OfomanEmployment: Massachusetts General Hospital Kerry ReynoldsEmployment: TeladocStock and Other Ownership Interests: Biogen (I)Other Relationship: Project DataSphere Henning WillersResearch Funding: Apple Inc Wilfredo-Garcia BeltranPatents, Royalties, Other Intellectual Property: European Patent—New therapy for treating graft-versus-host disease (EP3575320A1) Kimberly BlumenthalLeadership: Novocardia (I)Stock and Other Ownership Interests: Devoted Health (I), Novocardia (I)Honoraria: UpToDate, GA²LEN ANACAREResearch Funding: National Institute of Health (NIH)—K01AI125631, Massachusetts General Hospital, Transformative Scholar Award Ryan J. SullivanConsulting or Advisory Role: Novartis, Merck, Replimune, Asana Biosciences, Alkermes, Eisai, Pfizer, Iovance Biotherapeutics, OncoSec, AstraZeneca, Bristol Myers SquibbResearch Funding: Amgen (Inst), Lilly (Inst), BioMed Valley Discoveries (Inst), Merck (Inst), Deciphera (Inst), Roche/Genentech (Inst), Moderna Therapeutics (Inst), Sanofi (Inst), Aeglea Biotherapeutics (Inst), Asana Biosciences (Inst), Viralytics (Inst), Compugen (Inst), Neon Therapeutics (Inst), Pfizer (Inst), BeiGene (Inst), Rubius Therapeutics (Inst), Strategia (Inst) Yi-Bin ChenConsulting or Advisory Role: Magenta Therapeutics, Incyte, Kiadis Pharma, AbbVie, Equillium, Daiichi Sankyo/Lilly, Celularity, Actinium Pharmaceuticals Arthur KimConsulting or Advisory Role: Kintor PharmaceuticalPatents, Royalties, Other Intellectual Property: Uptodate, Chapter Royalties Aditya BardiaConsulting or Advisory Role: Novartis (Inst), Genentech, Pfizer (Inst), Spectrum Pharmaceuticals, bioTheranostics, Merck, Radius Health (Inst), Immunomedics (Inst), Genentech/Roche (Inst), Innocrin Pharma (Inst), Sanofi, Puma Biotechnology, Daiichi Sankyo/Astra Zeneca, Foundation Medicine, PhilipsResearch Funding: Genentech (Inst), Novartis (Inst), Pfizer (Inst), Merck (Inst), Sanofi (Inst), Radius Health (Inst), Immunomedics (Inst), AstraZeneca/Daiichi Sankyo (Inst)Open Payments Link: https://openpaymentsdata.cms.gov/physician/523675 A. John IafrateStock and Other Ownership Interests: Archer BiosciencesConsulting or Advisory Role: Repare Therapeutics, Kinnate Biopharma, Oncoclinicas Brasil, PAIGE.AIPatents, Royalties, Other Intellectual Property: ArcherDx exclusive license to AMP technology Justin F. GainorThis author is a member of the JCO Editorial Board. Journal policy recused the author from having any role in the peer review of this manuscript.Employment: Ironwood Pharmaceuticals (I)Stock and Other Ownership Interests: Ironwood Pharmaceuticals (I)Honoraria: Merck, Incyte, ARIAD, Novartis, Pfizer, TakedaConsulting or Advisory Role: Genentech, Bristol Myers Squibb, Theravance, Loxo, Takeda, Array BioPharma, Amgen, Merck, Agios, Regeneron, Oncorus, Jounce Therapeutics, Blueprint Medicines, Gilead Sciences, Lilly, Moderna TherapeuticsResearch Funding: Genentech, ARIAD, Merck, Novartis, Bristol Myers Squibb, Adaptimmune, AstraZeneca, Jounce Therapeutics, Blueprint Medicines, Moderna Therapeutics, Tesaro, Alexo Therapeutics, Array BioPharmaNo other potential conflicts of interest were reported.- Published
- 2022
- Full Text
- View/download PDF
48. Heterogeneous immunogenicity of SARS-CoV-2 vaccines in cancer patients receiving radiotherapy.
- Author
-
Bowes CL, Naranbhai V, St Denis KJ, Lam EC, Bertaux B, Keane FK, Khandekar MJ, Balazs AB, Iafrate JA, Gainor JF, and Willers H
- Subjects
- BNT162 Vaccine, COVID-19 Vaccines, Humans, Prospective Studies, SARS-CoV-2, COVID-19, Neoplasms radiotherapy
- Abstract
The immunogenicity of SARS-CoV-2 vaccines in cancer patients receiving radiotherapy is unknown. This prospective cohort study demonstrates that anti-SARS-CoV-2 spike antibody and neutralization titers are reduced in a subset of thoracic radiotherapy patients, possibly due to immunosuppressive conditions. Antibody testing may be useful to identify candidates for additional vaccine doses., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Dr. Gainor has served as a compensated consultant or received honoraria from Bristol-Myers Squibb, Genentech, Ariad/Takeda, Loxo/Lilly, Blueprint, Oncorus, Regeneron, Gilead, Moderna, AstraZeneca, Pfizer, Novartis, Merck, and GlydeBio; research support from Novartis, Genentech/Roche, and Ariad/Takeda; institutional research support from Bristol-Myers Squibb, Tesaro, Moderna, Blueprint, Jounce, Array Biopharma, Merck, Adaptimmune, Novartis, and Alexo; and has an immediate family member who is an employee with equity at Ironwood Pharmaceuticals. None of the other authors has declared a competing interest., (Copyright © 2021 Elsevier B.V. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
49. mRNA-based COVID-19 vaccine boosters induce neutralizing immunity against SARS-CoV-2 Omicron variant.
- Author
-
Garcia-Beltran WF, St Denis KJ, Hoelzemer A, Lam EC, Nitido AD, Sheehan ML, Berrios C, Ofoman O, Chang CC, Hauser BM, Feldman J, Gregory DJ, Poznansky MC, Schmidt AG, Iafrate AJ, Naranbhai V, and Balazs AB
- Abstract
Recent surveillance has revealed the emergence of the SARS-CoV-2 Omicron variant (BA.1/B.1.1.529) harboring up to 36 mutations in spike protein, the target of vaccine-induced neutralizing antibodies. Given its potential to escape vaccine-induced humoral immunity, we measured neutralization potency of sera from 88 mRNA-1273, 111 BNT162b, and 40 Ad26.COV2.S vaccine recipients against wild type, Delta, and Omicron SARS-CoV-2 pseudoviruses. We included individuals that were vaccinated recently (<3 months), distantly (6-12 months), or recently boosted, and accounted for prior SARS-CoV-2 infection. Remarkably, neutralization of Omicron was undetectable in most vaccinated individuals. However, individuals boosted with mRNA vaccines exhibited potent neutralization of Omicron only 4-6-fold lower than wild type, suggesting that boosters enhance the cross-reactivity of neutralizing antibody responses. In addition, we find Omicron pseudovirus is more infectious than any other variant tested. Overall, this study highlights the importance of boosters to broaden neutralizing antibody responses against highly divergent SARS-CoV-2 variants.
- Published
- 2021
- Full Text
- View/download PDF
50. Naive human B cells engage the receptor binding domain of SARS-CoV-2, variants of concern, and related sarbecoviruses.
- Author
-
Feldman J, Bals J, Altomare CG, St Denis K, Lam EC, Hauser BM, Ronsard L, Sangesland M, Moreno TB, Okonkwo V, Hartojo N, Balazs AB, Bajic G, Lingwood D, and Schmidt AG
- Subjects
- Antibodies, Neutralizing immunology, Antigens, Viral immunology, B-Lymphocytes metabolism, COVID-19 immunology, COVID-19 Vaccines immunology, Epitopes, Humans, Lymphocyte Activation, SARS-CoV-2 classification, SARS-CoV-2 metabolism, Spike Glycoprotein, Coronavirus chemistry, Spike Glycoprotein, Coronavirus metabolism, B-Lymphocytes cytology, B-Lymphocytes immunology, Coronavirus immunology, SARS-CoV-2 immunology
- Abstract
Initial exposure to a pathogen elicits an adaptive immune response to control and eradicate the threat. Interrogating the abundance and specificity of the naive B cell repertoire drives understanding of how to mount protective responses. Here, we isolated naive B cells from eight seronegative human donors targeting the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor binding domain (RBD). Single-cell B cell receptor (BCR) sequencing identified diverse gene usage and no restriction on complementarity determining region length. A subset of recombinant antibodies produced by naive B cell precursors bound to SARS-CoV-2 RBD and engaged circulating variants including B.1.1.7, B.1.351, and B.1.617.2, as well as preemergent bat-derived coronaviruses RaTG13, SHC104, and WIV1. By structural characterization of a naive antibody in complex with SARS-CoV-2 spike, we identified a conserved mode of recognition shared with infection-induced antibodies. We found that representative naive antibodies could signal in a B cell activation assay, and by using directed evolution, we could select for a higher-affinity RBD interaction, conferred by a single amino acid change. The minimally mutated, affinity-matured antibodies also potently neutralized SARS-CoV-2. Understanding the SARS-CoV-2 RBD–specific naive repertoire may inform potential responses capable of recognizing future SARS-CoV-2 variants or emerging coronaviruses, enabling the development of pan-coronavirus vaccines aimed at engaging protective germline responses.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.