1
|
Helmold M, Amann R. Advancing ORFV-Based Therapeutics to the Clinical Stage. Rev Med Virol 2025; 35:e70038. [PMID: 40346732 PMCID: PMC12064845 DOI: 10.1002/rmv.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/06/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
The Orf virus (ORFV) is the prototype member of the parapoxvirus family and has long been recognized for its robust immunogenicity, favourable safety profile and its ability to stimulate both cellular and humoural immune responses without inducing significant anti-vector immunity. Despite these inherent advantages, early applications of ORFV-based technologies were limited by challenges in manufacturing scalability and uncertainties regarding clinical safety in humans. However, recent breakthroughs have transformed this therapeutic landscape. A landmark achievement is the development of Prime-2-CoV, an ORFV-based anti-COVID-19 vaccine that has advanced into human clinical trials, providing the first clinical evidence of live ORFV's feasibility, safety and immunogenicity. This milestone, together with the establishment of a good manufacturing practice (GMP)-compliant production process and comprehensive preclinical evaluations, has laid a robust foundation for broader clinical applications of ORFV-based therapeutics. Moreover, the use of ORFV as an oncolytic virus therapy has shown promising results, effectively converting immunologically 'cold' tumours into 'hot' ones, underscoring its versatility as a therapeutic platform. In this review, we critically assess recent advances in ORFV-based therapeutics, with a particular focus on vaccine development and oncolytic virotherapy (OVT). We thoroughly discuss the milestones and impact of the first ORFV-based clinical trial, outline strategies for optimizing the technology and provide insights into overcoming remaining challenges. Collectively, these advancements position ORFV as a highly promising and versatile platform for next-generation prophylactic and therapeutic interventions in both human and veterinary medicine, while also providing a roadmap for future innovations.
Collapse
Affiliation(s)
- Matthias Helmold
- Institute of ImmunologyUniversity Hospital TübingenTübingenGermany
- Institute of Tropical MedicineUniversity Hospital TübingenTübingenGermany
| | - Ralf Amann
- Institute of ImmunologyUniversity Hospital TübingenTübingenGermany
| |
Collapse
|
2
|
Castro-Trujillo S, Castro-Meneses J, Rojas MC, Castro-Amaya M, Lastra G, Narváez CF. Regulatory cytokines modulate early isotype-specific response associated with COVID-19 survival. Front Immunol 2025; 16:1543626. [PMID: 40342417 PMCID: PMC12058664 DOI: 10.3389/fimmu.2025.1543626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/01/2025] [Indexed: 05/11/2025] Open
Abstract
Identifying immune markers driving early and effective antibody response in patients with severe coronavirus disease 2019 (COVID-19) is critical due to the threat of future coronavirus pandemics, incomplete global vaccination, and suboptimal booster coverage. Patients with life-threatening severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection are characterized by dysregulated thromboinflammation and cytokine storm that could influence the isotype virus-specific antibody response and the subsequent clinical outcome. We investigated the association between COVID-19-related mortality with the dynamics, magnitude, and relative avidity of nucleoprotein (N), spike (S), and receptor-binding domain (RBD)-specific IgM, IgA, and IgG in circulation. We also assessed the relationship between the virus-specific antibody responses and cytokine patterns, as well as systemic and pulmonary thromboinflammation markers. This multicenter study included COVID-19 patients hospitalized early in the pandemic, classified as survivors (n=62) and non-survivors (n=17). We developed indirect enzyme-linked immunosorbent assays (ELISAs) to evaluate each virus-specific isotype using well-characterized outpatient COVID-19 (n=180) and pre-pandemic cohorts (n=111). The pro-inflammatory interleukin (IL)-6 and tumor necrosis factor (TNF)-α, as well as the regulatory IL-10, transforming growth factor (TGF)-β1, and soluble tumor necrosis factor receptor I (sTNFRI) levels were evaluated. The ELISAs performed highly for all virus-specific isotypes, although modest for IgM-N. Non-survivors increased N-specific, but no S-specific, IgM and IgA responses throughout the disease course and, more notably, a delayed class switching to IgG-S and IgG-RBD compared to survivors. No differences were observed in the virus-specific IgG relative avidity. Survivors exhibited an antibody response proportional to the degree of systemic and pulmonary thromboinflammation, whereas non-survivors showed those dissociated because of their uncontrolled severe thromboinflammation. Only the survivors showed a dominant regulatory cytokine pattern in the early phase of infection (<10 days after symptoms onset), which strongly correlated with developing IgG-S and IgG-RBD protective antibodies. We developed easy-to-use immune assays that enable patient monitoring and identify at-risk populations in low- to middle-income regions. Non-survivors displayed an ineffective N-mediated antibody response, marked by an inability to control inflammation and a compromised time-dependent class switching toward S and RBD-specific IgG. The regulatory cytokine axis, including TGF-β1, maybe a critical immune correlate of effective antibody-mediated immunity in COVID-19.
Collapse
Affiliation(s)
- Sebastián Castro-Trujillo
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Juanita Castro-Meneses
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
- Programa de Biología Aplicada, Facultad de Ciencias Exactas y Naturales, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - María Clemencia Rojas
- Dirección Laboratorio de Salud Pública, Secretaría de Salud Departamental, Gobernación del Huila, Neiva, Huila, Colombia
| | - Marcela Castro-Amaya
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Giovani Lastra
- Departamento de Medicina Interna, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
- Servicio de Neumología, E.S.E. Hospital Universitario de Neiva. Programa de Medicina, Universidad Surcolombiana, Neiva, Huila, Colombia
| | - Carlos F. Narváez
- División de Inmunología, Programa de Medicina, Facultad de Ciencias de la Salud, Universidad Surcolombiana, Neiva, Huila, Colombia
| |
Collapse
|
3
|
Fahim C, Wang S, Paul N, Colwill K, Dayam R, Boyd JM, Ma H, Gruppuso V, Mrazovac A, Firman J, Patel A, Bach V, de Launay KQ, Takaoka A, Grubac V, Gingras AC, Straus SE, Mishra S. Patterns of SARS-CoV-2 seropositivity among essential workers in long term care and retirement homes in Ontario, Canada: A descriptive cross-sectional study. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0004294. [PMID: 40153448 PMCID: PMC11952236 DOI: 10.1371/journal.pgph.0004294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 01/27/2025] [Indexed: 03/30/2025]
Abstract
Understanding patterns of SARS-CoV-2 seroprevalence among Long-Term Care Home and Retirement Home (LTCH/RH) staff is critical to designing effective public health interventions. We estimated SARS-CoV-2 seroprevalence among LTCH/RH staff in Ontario, Canada between May 2021-October 2022 using a cross-sectional analysis. Eligible participants completed a demographic questionnaire and provided a dried blood spot sample. Positive seroprevalence was defined as the proportion of individuals in a population who were positive for a SARS-CoV-2 infection, determined using anti-nucleocapsid total IgG antibodies analyzed with a validated chemiluminescent ELISA. We report age-adjusted prevalence ratios [PR; confidence interval, CI] by participant socio-demographic, household, neighbourhood, and occupational characteristics and stratified the analyses over two time periods (period 1: 2021-05-17 to 2021-12-31; period 2: 2022-01-02 to 2022-10-25). A total of 603 staff were included in our analysis; n=235 (39%) were enrolled in period 1 and n=368 (61%) were enrolled in period 2. Seroprevalence was 24% and 44% in periods 1 and 2, respectively. Age-adjusted prevalence ratios were nearly 2-fold higher among Black [PR 1.78; CI 1.28-2.48], East and Southeast Asian [PR 1.55, CI 1.18-2.04] and other racialized participants [PR 1.42, CI 1.03-1.96] compared to White participants. We did not observe a pattern across household characteristics, although we observed a trend towards higher seropositivity among participants living in COVID-19 hotspots. Prevalence ratios were lower for participants in higher income neighbourhoods [PR 0.72, CI 0.58-0.98]. We did not observe variability in seroprevalence across occupational characteristics with the exception of paid sick leave which was higher among participants with home-provided paid sick leave at the time of the survey [PR 0.58, CI 0.45-0.75]. Among LTCH/RH staff, we found important sources of variability of SARS-CoV-2 seroprevalence and strong correlations with socioeconomic disparities. Our findings show the importance of designing equity-rooted health interventions that recognize the intersection between community and the workplace.
Collapse
Affiliation(s)
- Christine Fahim
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
| | - Siyi Wang
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Nimitha Paul
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Roya Dayam
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Jamie M. Boyd
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Huiting Ma
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Vincenza Gruppuso
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Ana Mrazovac
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Jessica Firman
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Anjali Patel
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Vanessa Bach
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | | | - Alyson Takaoka
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Vanja Grubac
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
| | - Anne-Claude Gingras
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health, Toronto, Canada
| | - Sharon E. Straus
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
- Dalla Lana School of Public Health, University of Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Sharmistha Mishra
- Li Ka Shing Knowledge Institute, Unity Health Toronto, Toronto, Canada
- Department of Medicine, University of Toronto, Toronto, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, Canada
- Institute of Clinical and Evaluative Sciences, Toronto, Canada
| |
Collapse
|
4
|
Oyebanji OA, Yin A, Sundheimer N, Ragavapuram V, Shea P, Cao Y, Chan PA, Nanda A, Tyagi R, Raza S, Mujahid N, Abul Y, Balazs AB, Bosch J, King CL, Klein SL, Gravenstein S, Canaday DH, Wilson BM. COVID-19 booster doses reduce sex disparities in antibody responses among nursing home residents. Aging Clin Exp Res 2025; 37:73. [PMID: 40055264 PMCID: PMC11889018 DOI: 10.1007/s40520-025-02990-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/20/2025] [Indexed: 03/12/2025]
Abstract
BACKGROUND Data suggest that antibody responses following COVID-19 vaccines are a correlate of protection. Some studies, including the clinical trials of COVID-19 mRNA vaccines, did not stratify and evaluate whether antibody responses to COVID-19 vaccines differed between the sexes or with aging. This gap in research is particularly relevant for older populations such as nursing home residents (NHR). We hypothesized that sex differences in vaccine-induced antibody responses may intersect with age and be diminished among older adults residing in nursing homes. METHODS We analyzed serum samples from 638 NHRs collected serially after the primary two-dose series and three subsequent booster doses of mRNA SARS-CoV-2 vaccinations. We analyzed anti-Spike IgG and neutralizing antibody titers to the Wuhan and Omicron BA.4/5 variant strains. Mixed-effects models predicting log-transformed titers were estimated to compare responses across vaccine doses, focusing on sex-differential responses. For detected post-dose sex differences, additional sample times were analyzed to assess the duration of the difference. RESULTS Following the primary series, female NHRs with a prior history of SARS-CoV-2 infection had significantly higher Wuhan anti-Spike antibodies and neutralizing antibody titers than male NHRs with differences persisting up to nine months post-vaccination. Subsequent monovalent booster doses and a bivalent booster dose eliminated this disparity. We did not detect any differential response to the Omicron BA.4/5 variant. CONCLUSIONS The blunting of sex differences in antibody response observed following the primary series by the 1st booster dose underscores the importance of booster vaccination in this population.
Collapse
Affiliation(s)
- Oladayo A Oyebanji
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Anna Yin
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Nicholas Sundheimer
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Vaishnavi Ragavapuram
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Patrick Shea
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Yi Cao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Philip A Chan
- Division of Infectious Diseases, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Aman Nanda
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Rohit Tyagi
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Sakeena Raza
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Nadia Mujahid
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Yasin Abul
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center of Innovation in Long-Term Services and Supports, Veterans Administration Medical Center, Providence, RI, USA
- Brown University School of Public Health Center for Gerontology and Healthcare Research, Providence, RI, USA
| | | | - Jürgen Bosch
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Christopher L King
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
| | - Sabra L Klein
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Stefan Gravenstein
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School of Brown University, Providence, RI, USA
- Center of Innovation in Long-Term Services and Supports, Veterans Administration Medical Center, Providence, RI, USA
- Brown University School of Public Health Center for Gerontology and Healthcare Research, Providence, RI, USA
| | - David H Canaday
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Geriatric Research Education and Clinical Center (GRECC), VA Northeast Ohio Healthcare System, Cleveland, OH, USA
| | - Brigid M Wilson
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Geriatric Research Education and Clinical Center (GRECC), VA Northeast Ohio Healthcare System, Cleveland, OH, USA
| |
Collapse
|
5
|
Bartholdsson S, Hergens MP, Hansson KE, Ragnarsson J, Hodosi P, Kus I, Insulander M, Vene S, Lindquist L, Askling HH, Gredmark-Russ S. Clinical Characteristics of Tick-Borne Encephalitis in Adult Patients: A 10-year Retrospective Study in Stockholm, Sweden. J Infect Dis 2025; 231:e195-e205. [PMID: 39316686 PMCID: PMC11793045 DOI: 10.1093/infdis/jiae463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND The incidence of tick-borne encephalitis (TBE) has increased during the last decades in Europe. Our aim was to assess the clinical characteristics and outcome of patients with TBE in Region Stockholm, as a high-risk area in Sweden. METHODS The notification database at the regional Department of Communicable Disease Control and Prevention was used to identify TBE cases during 2006-2015. Clinical data were retrieved from the included patients' medical records. The associations of specific variables to predefined outcomes of disease severity were evaluated with multivariate logistic regression models. RESULTS Of 1004 identified TBE cases, 703 adult patients were included. Sixty-one percent were men, and the median age was 50 years (range, 18-94 years). The majority of patients were nonvaccinated. Comorbidity was present in 34%, and 4% were receiving immunomodulatory therapy. Seventy-five percent were hospitalized, and 11% had severe disease. More than 70% of the 79 patients followed up for >6 months had persisting symptoms. The case fatality rate was 1.4%, 15% in the group with immunomodulatory treatment. In the multivariate analysis, severe disease was associated with underlying comorbid conditions, age ≥50 years, and previous complete TBE vaccination. CONCLUSIONS This is the largest cohort of patients with TBE in Scandinavia. Our findings of a more severe course of disease in older patients, those receiving immunomodulatory therapy, those with comorbid conditions, and those with vaccination breakthrough infections must be interpreted in the context of hospitalized patients. Optimized prevention is needed for patients receiving immunomodulatory therapy, given the considerable case fatality rate. Follow-up visits and rehabilitation should be better standardized.
Collapse
Affiliation(s)
- Sofia Bartholdsson
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Maria-Pia Hergens
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Communicable Disease Control and Prevention, Region Stockholm, Stockholm, Sweden
| | - Karin E Hansson
- Department of Infectious Diseases, Södersjukhuset Stockholm, Sweden
| | - Josef Ragnarsson
- Department of Infectious Diseases, University Hospital of Umeå, Umeå, Sweden
| | - Peter Hodosi
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ismail Kus
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mona Insulander
- Department of Communicable Disease Control and Prevention, Region Stockholm, Stockholm, Sweden
| | - Sirkka Vene
- The Public Health Agency of Sweden, Solna, Sweden
| | - Lars Lindquist
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Helena H Askling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Academic Specialist Centre, Stockholm Health Services, Region Stockholm, Stockholm, Sweden
| | - Sara Gredmark-Russ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Laboratory for Molecular Infection Medicine Sweden, Umeå, Sweden
| |
Collapse
|
6
|
Morgan G, Fung CYJ, Gingras AC, Colwill K, Briollais L, Frangione E, Wolday D, Qi F, Pasculescu A, Delgado-Brand M, Mailhot G, Tursun T, Arnoldo S, Bearss E, Binnie A, Borgundvaag B, Casalino S, Chowdhary S, Dagher M, Devine L, Elliott LT, Friedman SM, Khan Z, Lapadula E, MacDonald G, Mazzulli T, McLeod SL, Mighton C, Nirmalanathan K, Richardson D, Stern S, Taher A, Young J, Lerner-Ellis J, Taher J. Characterizing the SARS-CoV-2 antibody response and associations with patient factors: Serological profiling of participants enrolled in the GENCOV study. Clin Biochem 2025; 135:110859. [PMID: 39645018 DOI: 10.1016/j.clinbiochem.2024.110859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 11/09/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION The GENCOV study sought to evaluate serological differences between individuals with differing COVID-19 severity and outcomes. We assessed the SARS-CoV-2 antibody response of GENCOV participants cross-sectionally 1-, 6-, and 12-months following COVID-19 diagnosis to identify patient factors associated with more robust and durable humoral immune responses. MATERIALS AND METHODS COVID-19 patients and a control cohort of vaccinated infection-naïve participants were recruited at hospital sites across the Greater Toronto Area in Ontario, Canada. Commercially available and laboratory-developed serological assays were used to characterize features of participants' antibody responses, including both binding and neutralizing antibodies. Regression analyses were performed to identify associations between participant characteristics and features of the SARS-CoV-2 antibody response. RESULTS Samples were obtained from participants 1- (n = 938), 6- (n = 842), and 12-months (n = 662) post-infection or vaccination. At all time points, vaccinees, and to a greater extent those who were both infected and vaccinated, had significantly elevated anti-spike antibody levels compared to unvaccinated participants. Increasing age and/or illness severity were associated with significantly higher antibody levels among unvaccinated participants. Among vaccines, those who were vaccinated after infection (i.e., hybrid immunity) had consistently higher antibody levels compared to participants who were infection-naïve or vaccinated before their infection (i.e., breakthrough infections). Additionally, receiving more vaccine doses and having a more recent vaccination were strongly associated with higher antibody levels across all time points. CONCLUSIONS Our findings highlight various patient factors, including vaccination, which contribute to robust, durable SARS-CoV-2 antibody responses. Overall, the findings presented here may inform future vaccine development and rollout plans.
Collapse
Affiliation(s)
- Gregory Morgan
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chun Yiu Jordan Fung
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Karen Colwill
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Laurent Briollais
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Erika Frangione
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Dawit Wolday
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Freda Qi
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Adrian Pasculescu
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Melanie Delgado-Brand
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Genevieve Mailhot
- Princess Margaret Genomics Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Tulunay Tursun
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Saranya Arnoldo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; William Osler Health System, Brampton, ON L6R 3J, Canada
| | - Erin Bearss
- Mount Sinai Academic Family Health Team, Mount Sinai Hospital, Toronto, ON M5T 3L9, Canada; Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada
| | - Alexandra Binnie
- Department of Critical Care, William Osler Health System, Etobicoke, ON M9V 1R8, Canada
| | - Bjug Borgundvaag
- Schwartz/Reisman Emergency Medicine Institute, Sinai Health System, Toronto, ON M5G 2A2, Canada
| | - Selina Casalino
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Sunakshi Chowdhary
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Marc Dagher
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada; Women's College Hospital, Toronto, ON M5S 1B2, Canada
| | - Luke Devine
- Division of General Internal Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Lloyd T Elliott
- Department of Statistics and Actuarial Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Steven M Friedman
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada; Emergency Medicine, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Zeeshan Khan
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada
| | - Elisa Lapadula
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Georgia MacDonald
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Tony Mazzulli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Microbiology, Mount Sinai Hospital, Sinai Health, Toronto, ON M5G 1X5, Canada
| | - Shelley L McLeod
- Department of Family and Community Medicine, University of Toronto, Toronto, ON M5G 1V7, Canada; Schwartz/Reisman Emergency Medicine Institute, Sinai Health System, Toronto, ON M5G 2A2, Canada
| | - Chloe Mighton
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1A6, Canada; Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON M5T 3M6, Canada
| | | | | | - Seth Stern
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada
| | - Ahmed Taher
- Mackenzie Health, Richmond Hill, ON L4C 4Z3, Canada; Division of Emergency Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada
| | - Juliet Young
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Jordan Lerner-Ellis
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jennifer Taher
- Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
7
|
Parimi VP, Pyati A, Eerike M. Long-term antibody responses to COVAXIN and COVISHIELD vaccines in rheumatoid arthritis patients and healthy control population - A cross-sectional study. J Family Med Prim Care 2025; 14:107-114. [PMID: 39989580 PMCID: PMC11844958 DOI: 10.4103/jfmpc.jfmpc_907_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/04/2024] [Accepted: 07/18/2024] [Indexed: 02/25/2025] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation and damage in the joints. It often requires treatment with disease-modifying antirheumatic drugs (DMARDs) to manage symptoms and prevent progression. The study investigates the long-term antibody responses to COVAXIN and COVISHIELD vaccines in RA patients. Methodology This cross-sectional study (IEC approval no: AlIMS/BBN/IEC/AUG/2021/60-R dated Sept 05, 2022, and Ref No: 799/U/IEC/ESICMC/F490/09/2022 dated Oct 31, 2022) enrolled 103 diagnosed RA patients receiving DMARDs and 183 healthy controls. The participants who completed 1 year after the second dose of vaccination were included, and detailed information on demographic, medical, and vaccination were collected. Laboratory investigations included complete blood count, inflammatory markers, and antispike antibody levels. Statistical analyses assessed differences between COVAXIN and COVISHIELD subgroups, considering DMARDs usage and disease duration. Results Among RA patients, both COVAXIN and COVISHIELD groups exhibited low disease activity. No significant (P > 0.05) differences were found in IL-6, CRP, or antispike antibody levels between COVAXIN and COVISHIELD subgroups in RA patients and healthy controls. Notably, 89% of female RA patients received COVISHIELD. Co-morbidities, including hypothyroidism (44%), were prevalent in COVISHIELD-received RA patients. Antibody concentration varied significantly among DMARDs usage groups in COVAXIN-vaccinated RA patients, with a notable difference between three-drug and HCQ-alone regimens. However, no such difference was observed in the COVISHIELD group. Disease duration did not significantly impact antispike antibody concentration in either of the vaccination group. Conclusion RA patients had a decreased antibody response, 1 year after receiving the second dose of the COVID-19 vaccine. Nonetheless, there was no discernible difference in the antispike antibody concentration between the COVISHIELD and COVAXIN vaccination groups. Additionally, immunosuppressive medications significantly impact serological responses to these vaccines.
Collapse
Affiliation(s)
| | - Anand Pyati
- Department of Biochemistry, All India Institute of Medical Sciences, Bibinagar, Telangana, India
| | - Madhavi Eerike
- Department of Pharmacology, All India Institute of Medical Sciences, Bibinagar, Telangana, India
| |
Collapse
|
8
|
Klinkardt U, Schunk M, Ervin J, Schindler C, Sugimoto D, Rankin B, Amann R, Monti M, Kutschenko A, Schumacher C, Huber K, Zeder A, Heikkila N, Didierlaurent AM, Schwarz SE, Derouazi M. A novel orf virus vector-based COVID-19 booster vaccine shows cross-neutralizing activity in the absence of anti-vector neutralizing immunity. Hum Vaccin Immunother 2024; 20:2410574. [PMID: 39397784 PMCID: PMC11485980 DOI: 10.1080/21645515.2024.2410574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/12/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Next-generation COVID-19 vaccines are being developed to expand the breadth of coverage against existing and future variants and to extend the duration of protection. Prime-2-CoV_Beta is an orf virus (ORFV) based multi-antigen COVID-19 vaccine that co-expresses Spike (S) and Nucleocapsid (N) antigens. The safety and immunogenicity of Prime-2-CoV_Beta is investigated in a phase 1 first-in-human (FIH) dose-finding trial (ORFEUS study, ClinicalTrials.gov: NCT05367843). Participants of two age groups (18-55 and 65-85 years) who previously completed at least two doses of mRNA vaccines were enrolled and sequentially assigned to different dose groups to receive one intramuscular dose of 3 × 105, 3 × 106, 1.5 × 107, or 3 × 107 plaque-forming units (PFU) of Prime-2-CoV_Beta on day 1 and a second dose on day 29. Here, we report safety and immunogenicity data collected up to 6 months after the first study vaccination. Prime-2-CoV_Beta is safe and well tolerated and elicits immune responses at higher dose levels in participants aged 18-55. A single dose of 3 × 107 PFU boosted binding and cross-neutralizing antibody responses that are maintained through 6 months after the first booster vaccination. Polyfunctional S-specific CD4+ and CD8+ T cell responses are observed after vaccination. No pre-existing or vaccine-induced neutralizing anti-vector antibodies are detected. Our findings highlight the potential of the ORFV vector as a safe platform for future vaccine design, which provides the ability to deliver multiple antigens and allows for repeat immunization.
Collapse
Affiliation(s)
- Ute Klinkardt
- Speransa Therapeutics, TowerOne (Spaces), Frankfurt am Main, Germany
| | - Mirjam Schunk
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, Munich, Germany
| | - John Ervin
- AMR - Center for Pharmaceutical Research, Kansas City, MO, USA
| | - Christoph Schindler
- Medizinische Hochschule Hannover (MHH), Center for Clinical Trials (ZKS) – Early Clinical Trial Unit (ECTU), Hanover, Germany
| | | | - Bruce Rankin
- Accel Research Sites (ARS) – Corporate Office, DeLand, FL, USA
| | - Ralf Amann
- Department of Immunology, Institute for Cell Biology, University of Tübingen, Tübingen, Germany
| | | | - Anna Kutschenko
- Medizinische Hochschule Hannover (MHH), Center for Clinical Trials (ZKS) – Early Clinical Trial Unit (ECTU), Hanover, Germany
| | - Carsten Schumacher
- Medizinische Hochschule Hannover (MHH), Center for Clinical Trials (ZKS) – Early Clinical Trial Unit (ECTU), Hanover, Germany
| | - Kristina Huber
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, Munich, Germany
| | - Andreas Zeder
- Institute of Infectious Diseases and Tropical Medicine, LMU University Hospital, Munich, Germany
| | - Nelli Heikkila
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arnaud M. Didierlaurent
- Center of Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sylvia E. Schwarz
- Speransa Therapeutics, TowerOne (Spaces), Frankfurt am Main, Germany
| | - Madiha Derouazi
- Speransa Therapeutics, TowerOne (Spaces), Frankfurt am Main, Germany
| |
Collapse
|
9
|
Perfilyeva YV, Aquino AD, Borodin MA, Kali A, Abdolla N, Ostapchuk YO, Tleulieva R, Perfilyeva AV, Jainakbayev NT, Sharipov KO, Belyaev NN. Can interventions targeting MDSCs improve the outcome of vaccination in vulnerable populations? Int Rev Immunol 2024:1-17. [PMID: 39707917 DOI: 10.1080/08830185.2024.2443423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/26/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
Preventive vaccination is a crucial strategy for controlling and preventing infectious diseases, offering both effectiveness and cost-efficiency. However, despite the widespread success of vaccination programs, there are still certain population groups who struggle to mount adequate responses to immunization. These at-risk groups include but are not restricted to the elderly, overweight individuals, individuals with chronic infections and cancer patients. All of these groups are characterized by persistent chronic inflammation. Recent studies have demonstrated that one of the key players in immune regulation and the promotion of chronic inflammation are myeloid-derived suppressor cells (MDSCs). These cells possess a wide range of immunosuppressive mechanisms and are able to dampen immune responses in both antigen-specific and antigen-nonspecific manner, thus contributing to the establishment and maintenance of an inflammatory environment. Given their pivotal role in immune modulation, there is growing interest in understanding how MDSCs may influence the efficacy of vaccines, particularly in vulnerable populations. In this narrative review, we discuss whether MDSCs are able to regulate vaccine-induced immunity and whether their suppression can potentially enhance vaccine efficacy in vulnerable populations.
Collapse
Affiliation(s)
- Yuliya V Perfilyeva
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Arthur D Aquino
- Almazov National Medical Research Center, St. Petersburg, Russia
| | - Maxim A Borodin
- Almazov National Medical Research Center, St. Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg, Russia
| | - Aikyn Kali
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Nurshat Abdolla
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
- Al-Farabi, Kazakh National University, Almaty, Kazakhstan
| | | | - Raikhan Tleulieva
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | | | | - Kamalidin O Sharipov
- M.A. Aitkhozhin Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | |
Collapse
|
10
|
Fernández-Ciriza L, del Pozo JL, Betanzos N, González Á, Fernandez-Montero A, Carmona-Torre F, Vidaurreta M, Carlos S, Reina G. Impact of Repeated Variant Exposures on Cellular and Humoral Immunogenicity Induced by SARS-CoV-2 Vaccines. Vaccines (Basel) 2024; 12:1408. [PMID: 39772069 PMCID: PMC11680034 DOI: 10.3390/vaccines12121408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES The emergence of the Omicron variant has complicated COVID-19 control and prompted vaccine updates. Recent studies have shown that a fourth dose significantly protects against infection and severe disease, though long-term immunity data remain limited. This study aimed to assess Anti-S-RBD antibodies and interferon-γ levels in healthcare workers 12 months after receiving bivalent Original/Omicron BA.4-5 fourth SARS-CoV-2 vaccine. METHODS In this prospective cohort study, 549 healthcare workers were categorized by the initial vaccination schedule, with 229 individuals having received the fourth SARS-CoV-2 vaccine dose. Blood samples were collected from all participants 12 months post-vaccination. RESULTS Significant differences in Anti-S-RBD antibody levels were observed between those receiving a fourth dose and those who did not, while no differences were seen in interferon-γ levels. After 12 months, there were no significant differences in humoral and cellular immunity response between volunteers primoinfected or reinfected across different periods by the Omicron variant. A multivariable analysis revealed an association between high antibody levels (>6000 U/mL) and interferon-γ levels (OR: 3.13; 95% CI: 1.3-7.7; p < 0.05). Regarding primary vaccine schedules, participants vaccinated with ChAdOx1 (a single or double dose) had notably lower antibody levels compared to those who received mRNA-based vaccines. Additionally, the study shows a higher frequency of multiple infections among those with a single-dose ChAdOx1 primary schedule (OR: 6.24; 95% CI: 1.25-31.15; p < 0.01). CONCLUSIONS Overall, mRNA-based vaccines exhibited stronger long-term immunogenicity. Repeated exposure to the Omicron variant seems to mitigate immune imprinting from the wild-type SARS-CoV-2. An association was observed between high antibody levels and a strong cellular response, although the correlation was not linear.
Collapse
Affiliation(s)
- Leire Fernández-Ciriza
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
| | - José Luis del Pozo
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Infectious Diseases Division, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Nazaret Betanzos
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
| | - Álvaro González
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alejandro Fernandez-Montero
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Occupational Medicine, Universidad de Navarra, 31008 Pamplona, Spain
| | - Francisco Carmona-Torre
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Infectious Diseases Division, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Marta Vidaurreta
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Biochemistry, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Silvia Carlos
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
- Department of Preventive Medicine and Public Health, Universidad de Navarra, 31008 Pamplona, Spain
| | - Gabriel Reina
- Department of Microbiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (L.F.-C.); (N.B.); (M.V.); (G.R.)
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Spain; (Á.G.); (A.F.-M.); (F.C.-T.); (S.C.)
| |
Collapse
|
11
|
Aguirre F, Marro MJ, Rodriguez PE, Rall P, Miglietta EA, Miranda LAL, Poncet V, Pascuale CA, Ballejo CA, Ricardo T, Miragaya Y, Gamarnik A, Rossi AH, Silva AP. [Effect of previous exposure to COVID-19, occurrence of spikes, and type of vaccine on the humoral immune response of institutionalized older adults]. CAD SAUDE PUBLICA 2024; 40:e00155023. [PMID: 39417469 PMCID: PMC11469675 DOI: 10.1590/0102-311xes155023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 06/03/2024] [Accepted: 06/14/2024] [Indexed: 10/19/2024] Open
Abstract
This study evaluated the explanatory factors of humoral immune response in older adults admitted to long-term care institutions in Buenos Aires, Argentina, up to 180 days after vaccination. An open-label, prospective, multicenter cohort study was conducted with volunteers who received two doses of the Sputnik V, Sinopharm, or AZD1222 vaccines. Plasma samples were analyzed at 0 and 21 days after the first dose, 21 days after the second dose, and 120 and 180 days after the first dose. Marginal linear models and generalized additives mixed models were adjusted to determine the behavior of anti-spike IgG antibody concentration over time according to exposure group (naïve/no-naïve) and vaccine. Occurrence of an outbreak of COVID-19 in long-term care institutions and comorbidities were the covariates analyzed. A total of 773 participants were included, with a mean age of 83 years (IQR: 76-89). Results showed that antibody levels in the naïve: Sinopharm group were significantly lower to the other groups (p < 0.05). Antibody levels in the no-naïve: Sinopharm group were similar to those in the naïve group who received AZD1222 (p = 0.945) or Sputnik V (p = 1). Participants exposed to outbreaks in long-term care institutions had significantly higher antibody levels, regardless of exposure group and vaccine (p < 0.001). In conclusion, previous exposure to COVID-19, type of vaccine, and admittance into a long-term care institution with a history of outbreaks are factors to be considered in future epidemic events with transmission dynamics and immunological mechanisms similar to COVID-19, in populations similar to the one analyzed.
Collapse
Affiliation(s)
- Fernanda Aguirre
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - María Jimena Marro
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - Pamela E Rodriguez
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Pablo Rall
- Instituto Nacional de Servicios Sociales para Jubilados y Pensionados, Buenos Aires, Argentina
| | - Esteban A Miglietta
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lucía A López Miranda
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - Verónica Poncet
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - Carla A Pascuale
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Christian A Ballejo
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - Tamara Ricardo
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| | - Yanina Miragaya
- Instituto Nacional de Servicios Sociales para Jubilados y Pensionados, Buenos Aires, Argentina
| | - Andrea Gamarnik
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Andrés H Rossi
- Fundación Instituto Leloir, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Andrea P Silva
- Instituto Nacional de Epidemiología "Dr. Juan H. Jara", Administración Nacional de Laboratorios e Institutos de Salud "Dr. Carlos G. Malbrán", Mar del Plata, Argentina
| |
Collapse
|
12
|
Alirezaylavasani A, Skeie LG, Egner IM, Chopra A, Dahl TB, Prebensen C, Vaage JT, Halvorsen B, Lund-Johansen F, Tonby K, Reikvam DH, Stiksrud B, Holter JC, Dyrhol-Riise AM, Munthe LA, Kared H. Vaccine responses and hybrid immunity in people living with HIV after SARS-CoV-2 breakthrough infections. NPJ Vaccines 2024; 9:185. [PMID: 39384763 PMCID: PMC11464709 DOI: 10.1038/s41541-024-00972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
The COVID-19 pandemic posed a challenge for people living with HIV (PLWH), particularly immune non-responders (INR) with compromised CD4 T-cell reconstitution following antiretroviral therapy (CD4 count <350 cells per mm3). Their diminished vaccine responses raised concerns about their vulnerability to SARS-CoV-2 breakthrough infections (BTI). Our in-depth study here revealed chronic inflammation in PLWH and a limited anti-Spike IgG response after vaccination in INR. Nevertheless, the imprinting of Spike-specific B cells by vaccination significantly enhanced the humoral responses after BTI. Notably, the magnitude of cellular CD4 response in all PLWH was comparable to that in healthy donors (HD). However, the polyfunctionality and phenotype of Spike-specific CD8 T cells in INR differed from controls. The findings highlight the need for additional boosters with variant vaccines, and for monitoring ART adherence and the durability of both humoral and cellular anti-SARS-CoV-2 immunity in INR.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Linda Gail Skeie
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
13
|
Fabiani M, Mateo-Urdiales A, Sacco C, Fotakis EA, Battilomo S, Petrone D, Del Manso M, Bella A, Riccardo F, Stefanelli P, Palamara AT, Pezzotti P. Effectiveness against severe COVID-19 of a seasonal booster dose of bivalent (original/Omicron BA.4-5) mRNA vaccines in persons aged ≥60 years: Estimates over calendar time and by time since administration during prevalent circulation of different Omicron subvariants, Italy, 2022-2023. Vaccine 2024; 42:126026. [PMID: 38834428 DOI: 10.1016/j.vaccine.2024.05.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/21/2024] [Accepted: 05/31/2024] [Indexed: 06/06/2024]
Abstract
Evaluating how a COVID-19 seasonal vaccination program performed might help to plan future campaigns. This study aims to estimate the relative effectiveness (rVE) against severe COVID-19 of a seasonal booster dose over calendar time and by time since administration. We conducted a retrospective cohort analysis among 13,083,855 persons aged ≥60 years who were eligible to receive a seasonal booster at the start of the 2022-2023 vaccination campaign in Italy. We estimated rVE against severe COVID-19 (hospitalization or death) of a seasonal booster dose of bivalent (original/Omicron BA.4-5) mRNA vaccines by two-month calendar interval and at different times post-administration. We used multivariable Cox regression models, including vaccination as time-dependent exposure, to estimate adjusted hazard ratios (HR) and rVEs as [(1-HR)X100]. The rVE of a seasonal booster decreased from 64.9% (95% CI: 59.8-69.4) in October-November 2022 to 22.0% (95% CI: 15.4-28.0) in April-May 2023, when the majority of vaccinated persons (67%) had received the booster at least 4-6 months earlier. During the epidemic phase with prevalent circulation of the Omicron BA.5 subvariant, rVE of a seasonal booster received ≤90 days earlier was 83.0% (95% CI: 79.1-86.1), compared to 37.4% (95% CI: 25.5-47.5) during prevalent circulation of the Omicron XBB subvariant. During the XBB epidemic phase, rVE was estimated at 15.8% (95% CI: 9.1-20.1) 181-369 days post-administration of the booster dose. In all the analyses we observed similar trends of rVE between persons aged 60-79 and those ≥80 years, although estimates were somewhat lower for the oldest group. A seasonal booster dose received during the vaccination campaign provided additional protection against severe COVID-19 up to April-May 2023, after which the incidence of severe COVID-19 was much reduced. The results also suggest that the Omicron XBB subvariant might have partly escaped the immunity provided by the seasonal booster targeting the original and Omicron BA.4-5 strains of SARS-CoV-2.
Collapse
Affiliation(s)
- Massimo Fabiani
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Alberto Mateo-Urdiales
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Chiara Sacco
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy; European Programme on Intervention Epidemiology Training (EPIET), European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Emmanouil Alexandros Fotakis
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy; European Programme on Intervention Epidemiology Training (EPIET), European Centre for Disease Prevention and Control, Stockholm, Sweden
| | - Serena Battilomo
- General Directorate of Health Information System and Statistics, Italian Ministry of Health, Viale Giorgio Ribotta 5, 00144 Rome, Italy
| | - Daniele Petrone
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Martina Del Manso
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonino Bella
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Flavia Riccardo
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Anna Teresa Palamara
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| | - Patrizio Pezzotti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
14
|
Xu S, Sy LS, Hong V, Holmquist KJ, Qian L, Farrington P, Bruxvoort KJ, Klein NP, Fireman B, Han B, Lewin BJ. Ischemic Stroke After Bivalent COVID-19 Vaccination: Self-Controlled Case Series Study. JMIR Public Health Surveill 2024; 10:e53807. [PMID: 38916940 PMCID: PMC11234065 DOI: 10.2196/53807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 05/21/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND The potential association between bivalent COVID-19 vaccination and ischemic stroke remains uncertain, despite several studies conducted thus far. OBJECTIVE This study aimed to evaluate the risk of ischemic stroke following bivalent COVID-19 vaccination during the 2022-2023 season. METHODS A self-controlled case series study was conducted among members aged 12 years and older who experienced ischemic stroke between September 1, 2022, and March 31, 2023, in a large health care system. Ischemic strokes were identified using International Classification of Diseases, Tenth Revision codes in emergency departments and inpatient settings. Exposures were Pfizer-BioNTech or Moderna bivalent COVID-19 vaccination. Risk intervals were prespecified as 1-21 days and 1-42 days after bivalent vaccination; all non-risk-interval person-time served as the control interval. The incidence of ischemic stroke was compared in the risk interval and control interval using conditional Poisson regression. We conducted overall and subgroup analyses by age, history of SARS-CoV-2 infection, and coadministration of influenza vaccine. When an elevated risk was detected, we performed a chart review of ischemic strokes and analyzed the risk of chart-confirmed ischemic stroke. RESULTS With 4933 ischemic stroke events, we found no increased risk within the 21-day risk interval for the 2 vaccines and by subgroups. However, risk of ischemic stroke was elevated within the 42-day risk interval among individuals aged younger than 65 years with coadministration of Pfizer-BioNTech bivalent and influenza vaccines on the same day; the relative incidence (RI) was 2.13 (95% CI 1.01-4.46). Among those who also had a history of SARS-CoV-2 infection, the RI was 3.94 (95% CI 1.10-14.16). After chart review, the RIs were 2.34 (95% CI 0.97-5.65) and 4.27 (95% CI 0.97-18.85), respectively. Among individuals aged younger than 65 years who received Moderna bivalent vaccine and had a history of SARS-CoV-2 infection, the RI was 2.62 (95% CI 1.13-6.03) before chart review and 2.24 (95% CI 0.78-6.47) after chart review. Stratified analyses by sex did not show a significantly increased risk of ischemic stroke after bivalent vaccination. CONCLUSIONS While the point estimate for the risk of chart-confirmed ischemic stroke was elevated in a risk interval of 1-42 days among individuals younger than 65 years with coadministration of Pfizer-BioNTech bivalent and influenza vaccines on the same day and among individuals younger than 65 years who received Moderna bivalent vaccine and had a history of SARS-CoV-2 infection, the risk was not statistically significant. The potential association between bivalent vaccination and ischemic stroke in the 1-42-day analysis warrants further investigation among individuals younger than 65 years with influenza vaccine coadministration and prior SARS-CoV-2 infection. Furthermore, the findings on ischemic stroke risk after bivalent COVID-19 vaccination underscore the need to evaluate monovalent COVID-19 vaccine safety during the 2023-2024 season.
Collapse
Affiliation(s)
- Stanley Xu
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Lina S Sy
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Vennis Hong
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Kimberly J Holmquist
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Lei Qian
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Paddy Farrington
- School of Mathematics and Statistics, The Open University, United Kingdom
| | - Katia J Bruxvoort
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Nicola P Klein
- Kaiser Permanente Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, United States
| | - Bruce Fireman
- Kaiser Permanente Vaccine Study Center, Kaiser Permanente Northern California, Oakland, CA, United States
| | - Bing Han
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| | - Bruno J Lewin
- Department of Research & Evaluation, Southern California Permanente Medical Group, Pasadena, CA, United States
| |
Collapse
|
15
|
Mallory M, Munt JE, Narowski TM, Castillo I, Cuadra E, Pisanic N, Fields P, Powers JM, Dickson A, Harris R, Wargowsky R, Moran S, Allabban A, Raphel K, McCaffrey TA, Brien JD, Heaney CD, Lafleur JE, Baric RS, Premkumar L. COVID-19 point-of-care tests can identify low-antibody individuals: In-depth immunoanalysis of boosting benefits in a healthy cohort. SCIENCE ADVANCES 2024; 10:eadi1379. [PMID: 38865463 PMCID: PMC11168476 DOI: 10.1126/sciadv.adi1379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
The recommended COVID-19 booster vaccine uptake is low. At-home lateral flow assay (LFA) antigen tests are widely accepted for detecting infection during the pandemic. Here, we present the feasibility and potential benefits of using LFA-based antibody tests as a means for individuals to detect inadequate immunity and make informed decisions about COVID-19 booster immunization. In a health care provider cohort, we investigated the changes in the breadth and depth of humoral and T cell immune responses following mRNA vaccination and boosting in LFA-positive and LFA-negative antibody groups. We show that negative LFA antibody tests closely reflect the lack of functional humoral immunity observed in a battery of sophisticated immune assays, while positive results do not necessarily reflect adequate immunity. After booster vaccination, both groups gain depth and breadth of systemic antibodies against evolving SARS-CoV-2 and related viruses. Our findings show that LFA-based antibody tests can alert individuals about inadequate immunity against COVID-19, thereby increasing booster shots and promoting herd immunity.
Collapse
Affiliation(s)
- Michael Mallory
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jennifer E. Munt
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tara M. Narowski
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Izabella Castillo
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Edwing Cuadra
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Nora Pisanic
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | | | - John M. Powers
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandria Dickson
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, USA
| | - Rohan Harris
- Department Emergency Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Richard Wargowsky
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, Washington, DC, USA
| | - Seamus Moran
- Department Emergency Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Ahmed Allabban
- Department Emergency Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Kristin Raphel
- Department Emergency Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Timothy A. McCaffrey
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, Washington, DC, USA
| | - James D. Brien
- Department of Molecular Microbiology and Immunology, Saint Louis University, Saint Louis, MO, USA
| | - Christopher D. Heaney
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - John E. Lafleur
- Department Emergency Medicine, George Washington University School of Medicine, Washington, DC, USA
| | - Ralph S. Baric
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
- Department of Epidemiology, UNC Chapel Hill School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
16
|
Yang Y, Li F, Du L. Therapeutic nanobodies against SARS-CoV-2 and other pathogenic human coronaviruses. J Nanobiotechnology 2024; 22:304. [PMID: 38822339 PMCID: PMC11140877 DOI: 10.1186/s12951-024-02573-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 05/20/2024] [Indexed: 06/02/2024] Open
Abstract
Nanobodies, single-domain antibodies derived from variable domain of camelid or shark heavy-chain antibodies, have unique properties with small size, strong binding affinity, easy construction in versatile formats, high neutralizing activity, protective efficacy, and manufactural capacity on a large-scale. Nanobodies have been arisen as an effective research tool for development of nanobiotechnologies with a variety of applications. Three highly pathogenic coronaviruses (CoVs), SARS-CoV-2, SARS-CoV, and MERS-CoV, have caused serious outbreaks or a global pandemic, and continue to post a threat to public health worldwide. The viral spike (S) protein and its cognate receptor-binding domain (RBD), which initiate viral entry and play a critical role in virus pathogenesis, are important therapeutic targets. This review describes pathogenic human CoVs, including viral structures and proteins, and S protein-mediated viral entry process. It also summarizes recent advances in development of nanobodies targeting these CoVs, focusing on those targeting the S protein and RBD. Finally, we discuss potential strategies to improve the efficacy of nanobodies against emerging SARS-CoV-2 variants and other CoVs with pandemic potential. It will provide important information for rational design and evaluation of therapeutic agents against emerging and reemerging pathogens.
Collapse
MESH Headings
- Single-Domain Antibodies/immunology
- Single-Domain Antibodies/pharmacology
- Single-Domain Antibodies/therapeutic use
- Single-Domain Antibodies/chemistry
- Humans
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/metabolism
- Animals
- COVID-19/virology
- COVID-19/immunology
- COVID-19/therapy
- Coronavirus Infections/drug therapy
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Middle East Respiratory Syndrome Coronavirus/immunology
- Virus Internalization/drug effects
- Pandemics
- Betacoronavirus/immunology
- Antibodies, Viral/immunology
- Antibodies, Viral/therapeutic use
- Pneumonia, Viral/drug therapy
- Pneumonia, Viral/virology
- Pneumonia, Viral/immunology
- Severe acute respiratory syndrome-related coronavirus/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/therapeutic use
Collapse
Affiliation(s)
- Yang Yang
- Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames, IA, USA
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, MN, USA.
- Center for Coronavirus Research, University of Minnesota, Minneapolis, MN, USA.
| | - Lanying Du
- Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
17
|
Hyun H, Nham E, Seong H, Yoon JG, Noh JY, Cheong HJ, Kim WJ, Yoon SK, Park SJ, Gwak W, Lee JW, Kim B, Song JY. Long-term humoral and cellular immunity against vaccine strains and Omicron subvariants (BQ.1.1, BN.1, XBB.1, and EG.5) after bivalent COVID-19 vaccination. Front Immunol 2024; 15:1385135. [PMID: 38756783 PMCID: PMC11096540 DOI: 10.3389/fimmu.2024.1385135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
Background The assessment of long-term humoral and cellular immunity post-vaccination is crucial for establishing an optimal vaccination strategy. Methods This prospective cohort study evaluated adults (≥18 years) who received a BA.4/5 bivalent vaccine. We measured the anti-receptor binding domain immunoglobulin G antibody and neutralizing antibodies (NAb) against wild-type and Omicron subvariants (BA.5, BQ.1.1, BN.1, XBB.1 and EG.5) up to 9 months post-vaccination. T-cell immune responses were measured before and 4 weeks after vaccination. Results A total of 108 (28 SARS-CoV-2-naïve and 80 previously infected) participants were enrolled. Anti-receptor binding domain immunoglobulin G (U/mL) levels were higher at 9 months post-vaccination than baseline in SAR-CoV-2-naïve individuals (8,339 vs. 1,834, p<0.001). NAb titers against BQ.1.1, BN.1, and XBB.1 were significantly higher at 9 months post-vaccination than baseline in both groups, whereas NAb against EG.5 was negligible at all time points. The T-cell immune response (median spot forming unit/106 cells) was highly cross-reactive at both baseline (wild-type/BA.5/XBB.1.5, 38.3/52.5/45.0 in SARS-CoV-2-naïve individuals; 51.6/54.9/54.9 in SARS-CoV-2-infected individuals) and 4 weeks post-vaccination, with insignificant boosting post-vaccination. Conclusion Remarkable cross-reactive neutralization was observed against BQ.1.1, BN.1, and XBB.1 up to 9 months after BA.4/5 bivalent vaccination, but not against EG.5. The T-cell immune response was highly cross-reactive.
Collapse
Affiliation(s)
- Hakjun Hyun
- Department of Infectious Diseases, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Eliel Nham
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hye Seong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jin Gu Yoon
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sun Kyung Yoon
- Division of Vaccine Clinical Research, Center for Vaccine Research National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Se-Jin Park
- Division of Vaccine Clinical Research, Center for Vaccine Research National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - WonSeok Gwak
- Division of Vaccine Clinical Research, Center for Vaccine Research National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - June-Woo Lee
- Division of Vaccine Clinical Research, Center for Vaccine Research National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Byoungguk Kim
- Division of Vaccine Clinical Research, Center for Vaccine Research National Institute of Infectious Diseases, Korea National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
- Asia Pacific Influenza Institute, Korea University College of Medicine, Seoul, Republic of Korea
- Department of Research and Development, Vaccine Innovation Center, Korea University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Korosec CS, Dick DW, Moyles IR, Watmough J. SARS-CoV-2 booster vaccine dose significantly extends humoral immune response half-life beyond the primary series. Sci Rep 2024; 14:8426. [PMID: 38637521 PMCID: PMC11026522 DOI: 10.1038/s41598-024-58811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
SARS-CoV-2 lipid nanoparticle mRNA vaccines continue to be administered as the predominant prophylactic measure to reduce COVID-19 disease pathogenesis. Quantifying the kinetics of the secondary immune response from subsequent doses beyond the primary series and understanding how dose-dependent immune waning kinetics vary as a function of age, sex, and various comorbidities remains an important question. We study anti-spike IgG waning kinetics in 152 individuals who received an mRNA-based primary series (first two doses) and a subset of 137 individuals who then received an mRNA-based booster dose. We find the booster dose elicits a 71-84% increase in the median Anti-S half life over that of the primary series. We find the Anti-S half life for both primary series and booster doses decreases with age. However, we stress that although chronological age continues to be a good proxy for vaccine-induced humoral waning, immunosenescence is likely not the mechanism, rather, more likely the mechanism is related to the presence of noncommunicable diseases, which also accumulate with age, that affect immune regulation. We are able to independently reproduce recent observations that those with pre-existing asthma exhibit a stronger primary series humoral response to vaccination than compared to those that do not, and further, we find this result is sustained for the booster dose. Finally, via a single-variate Kruskal-Wallis test we find no difference between male and female humoral decay kinetics, however, a multivariate approach utilizing Least Absolute Shrinkage and Selection Operator (LASSO) regression for feature selection reveals a statistically significant (p < 1 × 10 - 3 ), albeit small, bias in favour of longer-lasting humoral immunity amongst males.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
| | - James Watmough
- Department of Mathematics and Statistics, University of New Brunswick, 3 Bailey Dr, Fredericton, E3B 5A3, NB, Canada
| |
Collapse
|
19
|
Hasani-Sadrabadi MM, Majedi FS, Zarubova J, Thauland TJ, Arumugaswami V, Hsiai TK, Bouchard LS, Butte MJ, Li S. Harnessing Biomaterials to Amplify Immunity in Aged Mice through T Memory Stem Cells. ACS NANO 2024; 18:6908-6926. [PMID: 38381620 DOI: 10.1021/acsnano.3c08559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
The durability of a protective immune response generated by a vaccine depends on its ability to induce long-term T cell immunity, which tends to decline in aging populations. The longest protection appears to arise from T memory stem cells (TMSCs) that confer high expandability and effector functions when challenged. Here we engineered artificial antigen presenting cells (aAPC) with optimized size, stiffness and activation signals to induce human and mouse CD8+ TMSCs in vitro. This platform was optimized as a vaccine booster of TMSCs (Vax-T) with prolonged release of small-molecule blockade of the glycogen synthase kinase-3β together with target antigens. By using SARS-CoV-2 antigen as a model, we show that a single injection of Vax-T induces durable antigen-specific CD8+ TMSCs in young and aged mice, and generates humoral responses at a level stronger than or similar to soluble vaccines. This Vax-T approach can boost long-term immunity to fight infectious diseases, cancer, and other diseases.
Collapse
Affiliation(s)
| | - Fatemeh S Majedi
- Department of Bioengineering, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Jana Zarubova
- Department of Bioengineering, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Timothy J Thauland
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, University of California Los Angeles, Los Angeles, California 90095 United States
| | - Vaithilingaraja Arumugaswami
- Jonsson Comprehensive Cancer Center, University of California Los Angeles; Los Angeles, California 90095 United States
- Department of Molecular and Medical Pharmacology, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Tzung K Hsiai
- Department of Bioengineering, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Louis-S Bouchard
- Department of Bioengineering, University of California Los Angeles; Los Angeles, California 90095 United States
- Jonsson Comprehensive Cancer Center, University of California Los Angeles; Los Angeles, California 90095 United States
- Department of Chemistry and Biochemistry, University of California Los Angeles; Los Angeles, California 90095 United States
- The Molecular Biology Institute, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Manish J Butte
- Department of Pediatrics, Division of Immunology, Allergy, and Rheumatology, University of California Los Angeles, Los Angeles, California 90095 United States
- Jonsson Comprehensive Cancer Center, University of California Los Angeles; Los Angeles, California 90095 United States
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles; Los Angeles, California 90095 United States
| | - Song Li
- Department of Bioengineering, University of California Los Angeles; Los Angeles, California 90095 United States
- Jonsson Comprehensive Cancer Center, University of California Los Angeles; Los Angeles, California 90095 United States
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles; Los Angeles, California 90095 United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles; Los Angeles, California 90095 United States
| |
Collapse
|
20
|
Poniedziałek B, Sikora D, Hallmann E, Brydak L, Rzymski P. Influenza vaccination as a prognostic factor of humoral IgA responses to SARS-CoV-2 infection. Cent Eur J Immunol 2024; 49:11-18. [PMID: 38812601 PMCID: PMC11130984 DOI: 10.5114/ceji.2024.135462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 01/18/2024] [Indexed: 05/31/2024] Open
Abstract
There is evidence that influenza vaccination may provide additional benefits by inducing training of innate immunity and increasing humoral responses to heterologous challenges. Immunoglobulin A (IgA) antibodies dominate the early phase of the adaptive response to SARS-CoV-2 infection, but whether their production may be associated with previous influenza vaccination has not been a subject of any study. This study compared serum SARS-CoV-2-specific IgA responses, measured with Microblot-Array assay, in individuals who experienced COVID-19 (N = 1318) and differed in the status of the seasonal influenza vaccine, age, sex, and disease severity. Influenza-vaccinated individuals had a higher seroprevalence of IgA antibodies against nucleocapsid (anti-NP; by 10.1%), receptor-binding domain of spike protein (anti-RBD; by 11.8%) and the S2 subunit of spike protein (anti-S2; by 6.8%). Multivariate analysis, including age, sex, and COVID-19 severity, confirmed that receiving the influenza vaccine was associated with higher odds of being seropositive for anti-NP (OR, 95% CI = 1.57, 1.2-2.0), anti-RBD (OR, 95% CI = 1.6, 1.3-2.0), and anti-S2 (OR, 95% CI = 1.9, 1.4-2.7), as well as being seropositive for at least one anti-SARS-CoV-2 IgA antibody (OR, 95% CI = 1.7, 1.3-2.1) and all three of them (OR, 95% CI = 2.6, 1.7-4.0). Age ≥ 50 years was an additional factor predicting better IgA responses. However, the concentration of particular antibodies in seropositive subjects did not differ in relation to the influenza vaccination status. The study evidenced that influenza vaccination was associated with improved serum IgA levels produced in response to SARS-CoV-2 infection. Further studies are necessary to assess whether trained immunity is involved in the observed phenomenon.
Collapse
Affiliation(s)
- Barbara Poniedziałek
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Dominika Sikora
- Doctoral School, Poznan University of Medical Sciences, Poznań, Poland
| | - Ewelina Hallmann
- Laboratory of Influenza Viruses and Respiratory Infection Viruses, Department of Virology at the National Institute of Public Health NIH – National Research Institute in Warsaw, Poland
| | - Lidia Brydak
- Laboratory of Influenza Viruses and Respiratory Infection Viruses, Department of Virology at the National Institute of Public Health NIH – National Research Institute in Warsaw, Poland
| | - Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
21
|
Xie Y, Xia Y, Xu H, Wang J, Zhang W, Li L, Liu Z. Analysis of related factors of plasma antibody levels in patients with severe and critical COVID-19. Sci Rep 2024; 14:2581. [PMID: 38297067 PMCID: PMC10831068 DOI: 10.1038/s41598-024-52572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 01/20/2024] [Indexed: 02/02/2024] Open
Abstract
Coronavirus disease 2019 (COVID-19) continues to impact global public health. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become less virulent as it mutates, prompting China to ease restrictions at the end of 2022. With the complete reopening, a surge in COVID-19 cases has ensued. Therefore, we conducted a study to explore the correlation between plasma antibody levels and baseline conditions or clinical outcomes in severe and critical patients. We collected the basic information of 79 included patients. Enzyme-linked immunosorbent assay (ELISA) tests were performed on plasma samples. The receptor-binding domain (RBD) IgG antibody level of the mild group was significantly higher than that of the severe/critical group (P = 0.00049). And in the severe/critical group, there existed an association between plasma antibody levels and age (P < 0.001, r = - 0.471), as well as plasma antibody levels and vaccination status (P = 0.00147, eta2 = 0.211). Besides, the level of plasma antibody seemed to be moderately correlated with the age, indicating the need for heightened attention to infections in the elderly. And plasma antibody levels were strongly associated with vaccination status in the severe/critical patients.
Collapse
Affiliation(s)
- Yudi Xie
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, 26 Huacai Road, Longtan Industry Zone, Chenghua District, Chengdu, Sichuan Province, People's Republic of China
- Key Laboratory of Transfusion Adverse Reactions, CAMS, Chengdu, Sichuan Province, People's Republic of China
| | - Yang Xia
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Second Military Medical University, Shanghai, People's Republic of China
| | - Haixia Xu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, 26 Huacai Road, Longtan Industry Zone, Chenghua District, Chengdu, Sichuan Province, People's Republic of China
- Key Laboratory of Transfusion Adverse Reactions, CAMS, Chengdu, Sichuan Province, People's Republic of China
| | - Jue Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, 26 Huacai Road, Longtan Industry Zone, Chenghua District, Chengdu, Sichuan Province, People's Republic of China
- Key Laboratory of Transfusion Adverse Reactions, CAMS, Chengdu, Sichuan Province, People's Republic of China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, The First Affiliated Hospital of Second Military Medical University, Shanghai, People's Republic of China
| | - Ling Li
- Department of Blood Transfusion, The Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, 82 Qinglong Street, Qingyang District, Chengdu, Sichuan Province, People's Republic of China.
- College of Public, Hygiene of Anhui Medical University, Hefei, Anhui Province, People's Republic of China.
| | - Zhong Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, 26 Huacai Road, Longtan Industry Zone, Chenghua District, Chengdu, Sichuan Province, People's Republic of China.
- College of Public, Hygiene of Anhui Medical University, Hefei, Anhui Province, People's Republic of China.
| |
Collapse
|
22
|
Kim YK, Choi Y, Jung JI, Kim JY, Kim MH, Curtis J, Lee EB. Modeling of antibody responses to COVID-19 vaccination in patients with rheumatoid arthritis. Sci Rep 2024; 14:1335. [PMID: 38228736 PMCID: PMC10791674 DOI: 10.1038/s41598-024-51535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 01/06/2024] [Indexed: 01/18/2024] Open
Abstract
To construct a model of the antibody response to COVID-19 vaccination in patients with rheumatoid arthritis (RA), and to identify clinical factors affecting the antibody response. A total of 779 serum samples were obtained from 550 COVID-19-naïve RA patients who were vaccinated against COVID-19. Antibody titers for the receptor binding domain (anti-RBD) and nucleocapsid (anti-N) were measured. The time from vaccination, and the log-transformed anti-RBD titer, were modeled using a fractional polynomial method. Clinical factors affecting antibody responses were analyzed by a regression model using generalized estimating equation. The anti-RBD titer peaked at about 2 weeks post-vaccination and decreased exponentially to 36.5% of the peak value after 2 months. Compared with the first vaccination, the 3rd or 4th vaccinations shifted the peaks of anti-RBD antibody response curves significantly upward (by 28-fold [4-195] and 32-fold [4-234], respectively). However, there was no significant shift in the peak from the 3rd vaccination to the 4th vaccination (p = 0.64). Multivariable analysis showed that sulfasalazine increased the vaccine response (by 1.49-fold [1.13-1.97]), but abatacept or JAK inhibitor decreased the vaccine response (by 0.13-fold [0.04-0.43] and 0.44-fold [0.26-0.74], respectively). Age was associated with lower ln [anti-RBD] values (coefficient: - 0.03 [- 0.04 to - 0.02]). In conclusion, the anti-RBD response of RA patients peaked at 2 weeks after COVID-19 vaccination, and then decreased exponentially, with the maximum peak increase observed after the 3rd vaccination. The antibody response was affected by age and the medications used to treat RA.
Collapse
Affiliation(s)
- Yun Kyu Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yunhee Choi
- Medical Research Collaborating Center, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ji In Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ju Yeon Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Mi Hyeon Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jeffrey Curtis
- Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, USA
| | - Eun Bong Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul, Republic of Korea.
- Division of Rheumatology, Department of Internal Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 110-744, Republic of Korea.
| |
Collapse
|
23
|
He Y, Xue X, Dalbeth N, Terkeltaub R, Chen Y, Yan F, Pang L, Li X, Yuan X, Cheng X, Li C, Sun M. COVID-19 Vaccination and Gout Flare Risk in Patients With Infrequent or Frequent Flares: A Prospective Cohort Study. Arthritis Care Res (Hoboken) 2024; 76:131-139. [PMID: 37553607 DOI: 10.1002/acr.25215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/07/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
OBJECTIVE To assess post-COVID-19 vaccination gout flare risk with differing baseline flare burden. METHODS We prospectively studied gout patients with infrequent or frequent flares, defined as ≤1 flare/year or ≥2 flares/year, respectively. COVID-19 vaccine-naive patients managed with urate-lowering therapy between February and June 2021 were included and voluntarily decided on vaccination. Participants were followed for 12 weeks after enrollment or first vaccine dose. Gout flares and risk factors were compared between groups. RESULTS Of 530 participants, 308 (58.1%) had infrequent flares and 222 (41.9%) had frequent flares at baseline, with 248 (142 infrequent and 106 frequent) receiving two-dose COVID-19 vaccination. Vaccination increased cumulative flare incidence at 12 weeks in the infrequent but not the frequent flare group (26.1% vs 10.8%, P = 0.001, compared with 60.4% vs 65.5%, P = 0.428). Flare incidence in the final 4 weeks of observation decreased significantly only in the vaccinated infrequent flare group (4.3% vs 12.0%, P = 0.017). Multivariable analyses showed that vaccination (odds ratio [OR] 2.82, 95% confidence interval [95% CI] 1.50-5.30, P = 0.001), flare in the preceding year (OR 1.95, 95% CI 1.03-3.71, P = 0.04), and body mass index (OR 1.09, 95% CI 1.01-1.19, P = 0.03) were independently associated with increased flare risk in the infrequent flare group. Baseline serum urate (mg/dl) was an independent risk factor in the frequent flare group (OR 1.23, 95% CI 1.05-1.45, P = 0.012). CONCLUSION COVID-19 vaccination was associated with increased early gout flares only in patients with previously infrequent flares.
Collapse
Affiliation(s)
- Yuwei He
- The Affiliated Hospital of Qingdao University, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, and Qingdao University, Qingdao, China
| | - Xiaomei Xue
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Robert Terkeltaub
- VA San Diego Medical Center, San Diego, and University of California San Diego, La Jolla, California
| | - Ying Chen
- The Affiliated Hospital of Qingdao University and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| | - Fei Yan
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Pang
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinde Li
- The Affiliated Hospital of Qingdao University and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| | | | - Xiaoyu Cheng
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Changgui Li
- The Affiliated Hospital of Qingdao University, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, and Qingdao University, Qingdao, China
| | - Mingshu Sun
- The Affiliated Hospital of Qingdao University and Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Qingdao, China
| |
Collapse
|
24
|
Keeshan A, Galipeau Y, Heiskanen A, Collins E, McCluskie PS, Arnold C, Saginur R, Booth R, Little J, McGuinty M, Buchan CA, Crawley A, Langlois MA, Cooper C. Results of the Stop the Spread Ottawa (SSO) cohort study: a Canadian urban-based prospective evaluation of antibody responses and neutralisation efficiency to SARS-CoV-2 infection and vaccination. BMJ Open 2023; 13:e077714. [PMID: 37907304 PMCID: PMC10619119 DOI: 10.1136/bmjopen-2023-077714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Predictors of COVID-19 vaccine immunogenicity and the influence of prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection require elucidation. METHODS Stop the Spread Ottawa is a prospective cohort of individuals at-risk for or who have been infected with SARS-CoV-2, initially enrolled for 10 months beginning October 2020. This cohort was enriched for public-facing workers. This analysis focuses on safety and immunogenicity of the initial two doses of COVID-19 vaccine. RESULTS Post-vaccination data with blood specimens were available for 930 participants. 22.8% were SARS-CoV2 infected prior to the first vaccine dose. Cohort characteristics include: median age 44 (IQR: 22-56), 66.6% women, 89.0% white, 83.2% employed. 38.1% reported two or more comorbidities and 30.8% reported immune compromising condition(s). Over 95% had detectable IgG levels against the spike and receptor binding domain (RBD) 3 months post second vaccine dose. By multivariable analysis, increasing age and high-level immune compromise predicted diminishing IgG spike and RBD titres at month 3 post second dose. IgG spike and RBD titres were higher immediately post vaccination in those with SARS-CoV-2 infection prior to first vaccination and spike titres were higher at 6 months in those with wider time intervals between dose 1 and 2. IgG spike and RBD titres and neutralisation were generally similar by sex, weight and whether receiving homogeneous or heterogeneous combinations of vaccines. Common symptoms post dose 1 vaccine included fatigue (64.7%), injection site pain (47.5%), headache (27.2%), fever/chills (26.2%) and body aches (25.3%). These symptoms were similar with subsequent doses. CONCLUSION The initial two COVID-19 vaccine doses are safe, well-tolerated and highly immunogenic across a broad spectrum of vaccine recipients including those working in public facing environments.
Collapse
Affiliation(s)
- Alexa Keeshan
- Dept of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Yannick Galipeau
- Department of Biochemistry, University of Ottawa, Ottawa, Ontario, Canada
| | - Aliisa Heiskanen
- Dept of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Erin Collins
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Pauline S McCluskie
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Corey Arnold
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Raphael Saginur
- Department of Medicine, Ottawa Hospital, Ottawa, Ontario, Canada
| | - Ronald Booth
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa Faculty of Medicine, Ottawa, Ontario, Canada
| | - Michaeline McGuinty
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital, Ottawa, Ontario, Canada
| | - C Arianne Buchan
- Dept of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Department of Medicine, Ottawa Hospital, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Anglea Crawley
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network, Ottawa, Ontario, Canada
| | - Marc-Andre Langlois
- Coronavirus Variants Rapid Response Network, Ottawa, Ontario, Canada
- Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Curtis Cooper
- Dept of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Coronavirus Variants Rapid Response Network, Ottawa, Ontario, Canada
- Division of Infectious Diseases, Ottawa Hospital Research Institute Clinical Epidemiology Program, Ottawa,Canada, Ontario, Canada
| |
Collapse
|
25
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. iScience 2023; 26:107967. [PMID: 37822504 PMCID: PMC10562792 DOI: 10.1016/j.isci.2023.107967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
As SARS-CoV-2 becomes endemic, it is critical to understand immunity following early-life infection. We evaluated humoral responses to SARS-CoV-2 in 23 infants/young children. Antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with spike and RBD IgG antibody half-life nearly 4X as long as in adults. IgG subtype analysis revealed that while IgG1 formed the majority of the response in both groups, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
Affiliation(s)
- Devyani Joshi
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Lindsay E. Nyhoff
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | | | - Alberto Moreno
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Kelly Manning
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Susanne Linderman
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Allison R. Burrell
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Environmental and Public Health Sciences, Division of Epidemiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathy Stephens
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Carson Norwood
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Grace Mantus
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Mary A. Staat
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mehul S. Suthar
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Jens Wrammert
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| |
Collapse
|
26
|
Ferreira IATM, Lee CYC, Foster WS, Abdullahi A, Dratva LM, Tuong ZK, Stewart BJ, Ferdinand JR, Guillaume SM, Potts MOP, Perera M, Krishna BA, Peñalver A, Cabantous M, Kemp SA, Ceron-Gutierrez L, Ebrahimi S, Lyons P, Smith KGC, Bradley J, Collier DA, McCoy LE, van der Klaauw A, Thaventhiran JED, Farooqi IS, Teichmann SA, MacAry PA, Doffinger R, Wills MR, Linterman MA, Clatworthy MR, Gupta RK. Atypical B cells and impaired SARS-CoV-2 neutralization following heterologous vaccination in the elderly. Cell Rep 2023; 42:112991. [PMID: 37590132 DOI: 10.1016/j.celrep.2023.112991] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 05/15/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
Suboptimal responses to a primary vaccination course have been reported in the elderly, but there is little information regarding the impact of age on responses to booster third doses. Here, we show that individuals 70 years or older (median age 73, range 70-75) who received a primary two-dose schedule with AZD1222 and booster third dose with mRNA vaccine achieve significantly lower neutralizing antibody responses against SARS-CoV-2 spike pseudotyped virus compared with those younger than 70 (median age 66, range 54-69) at 1 month post booster. Impaired neutralization potency and breadth post third dose in the elderly is associated with circulating "atypical" spike-specific B cells expressing CD11c and FCRL5. However, when considering individuals who received three doses of mRNA vaccine, we did not observe differences in neutralization or enrichment in atypical B cells. This work highlights the finding that AdV and mRNA COVID-19 vaccine formats differentially instruct the memory B cell response.
Collapse
Affiliation(s)
- Isabella A T M Ferreira
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Colin Y C Lee
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - William S Foster
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Adam Abdullahi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - John R Ferdinand
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Stephane M Guillaume
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK
| | - Martin O P Potts
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Benjamin A Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ana Peñalver
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Mia Cabantous
- Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK
| | - Steven A Kemp
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Soraya Ebrahimi
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Paul Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - John Bradley
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Dami A Collier
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Agatha van der Klaauw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, Cambridge, UK
| | | | - Paul A MacAry
- National University of Singapore, Singapore, Singapore
| | - Rainer Doffinger
- Department of Clinical Biochemistry and Immunology, Cambridge University Hospital NHS Trust, Cambridge, UK
| | - Mark R Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michelle A Linterman
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK; Molecular Immunity Unit, Department of Medicine, Medical Research Council Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK; Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK.
| | - Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
27
|
Clairon Q, Prague M, Planas D, Bruel T, Hocqueloux L, Prazuck T, Schwartz O, Thiébaut R, Guedj J. Modeling the kinetics of the neutralizing antibody response against SARS-CoV-2 variants after several administrations of Bnt162b2. PLoS Comput Biol 2023; 19:e1011282. [PMID: 37549192 PMCID: PMC10434962 DOI: 10.1371/journal.pcbi.1011282] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/17/2023] [Accepted: 06/20/2023] [Indexed: 08/09/2023] Open
Abstract
Because SARS-CoV-2 constantly mutates to escape from the immune response, there is a reduction of neutralizing capacity of antibodies initially targeting the historical strain against emerging Variants of Concern (VoC)s. That is why the measure of the protection conferred by vaccination cannot solely rely on the antibody levels, but also requires to measure their neutralization capacity. Here we used a mathematical model to follow the humoral response in 26 individuals that received up to three vaccination doses of Bnt162b2 vaccine, and for whom both anti-S IgG and neutralization capacity was measured longitudinally against all main VoCs. Our model could identify two independent mechanisms that led to a marked increase in measured humoral response over the successive vaccination doses. In addition to the already known increase in IgG levels after each dose, we identified that the neutralization capacity was significantly increased after the third vaccine administration against all VoCs, despite large inter-individual variability. Consequently, the model projects that the mean duration of detectable neutralizing capacity against non-Omicron VoC is between 348 days (Beta variant, 95% Prediction Intervals PI [307; 389]) and 587 days (Alpha variant, 95% PI [537; 636]). Despite the low neutralization levels after three doses, the mean duration of detectable neutralizing capacity against Omicron variants varies between 173 days (BA.5 variant, 95% PI [142; 200]) and 256 days (BA.1 variant, 95% PI [227; 286]). Our model shows the benefit of incorporating the neutralization capacity in the follow-up of patients to better inform on their level of protection against the different SARS-CoV-2 variants. Trial registration: This clinical trial is registered with ClinicalTrials.gov, Trial IDs NCT04750720 and NCT05315583.
Collapse
Affiliation(s)
- Quentin Clairon
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mélanie Prague
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Delphine Planas
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Timothée Bruel
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Laurent Hocqueloux
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional, Orléans, France
| | - Thierry Prazuck
- Service des Maladies Infectieuses et Tropicales, Centre Hospitalier Régional, Orléans, France
| | - Olivier Schwartz
- Vaccine Research Institute, Créteil, France
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, CNRS UMR3569, Paris, France
| | - Rodolphe Thiébaut
- Université de Bordeaux, Inria Bordeaux Sud-Ouest, Bordeaux, France
- Inserm, Bordeaux Population Health Research Center, SISTM Team, UMR1219, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | | |
Collapse
|
28
|
Lee J, Ko M, Kim S, Lim D, Park G, Lee SE. Household secondary attack rates and risk factors during periods of SARS-CoV-2 Delta and Omicron variant predominance in the Republic of Korea. Osong Public Health Res Perspect 2023; 14:263-271. [PMID: 37652681 PMCID: PMC10493696 DOI: 10.24171/j.phrp.2023.0133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND The household secondary attack rate (SAR) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an important indicator for community transmission. This study aimed to characterize transmission by comparing household SARs and identifying risk factors during the periods of Delta and Omicron variant predominance in Republic of Korea. METHODS We defined the period of Delta variant predominance (Delta period) as July 25, 2021 to January 15, 2022, and the period of Omicron variant predominance (Omicron period) as February 7 to September 3, 2022. The number of index cases included was 214,229 for the Delta period and 5,521,393 for the Omicron period. To identify the household SARs and risk factors for each period, logistic regression was performed to determine the adjusted odds ratio (aOR). RESULTS The SAR was 35.2% for the Delta period and 43.1% for the Omicron period. The aOR of infection was higher in 2 groups, those aged 0 to 18 years and ≥75 years, compared to those aged 19 to 49 years. Unvaccinated individuals (vs. vaccinated individuals) and individuals experiencing initial infection (vs. individuals experiencing a second or third infection) had an increased risk of infection with SARS-CoV-2. CONCLUSION This study analyzed the household SARs and risk factors. We hope that the results can help develop age-specific immunization plans and responses to reduce the SAR in preparation for emerging infectious diseases or potential new variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Jin Lee
- Central Disease Control Headquarters, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Mijeong Ko
- Capital Regional Center for Disease Control and Prevention, Korea Disease Control and Prevention Agency, Seoul, Republic of Korea
| | - Seontae Kim
- Central Disease Control Headquarters, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Dosang Lim
- Bureau of Chronic Disease Prevention and Control, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| | - Gemma Park
- Chungcheong Regional Center for Disease Control and Prevention, Korea Disease Control and Prevention Agency, Daejeon, Republic of Korea
| | - Sang-Eun Lee
- Central Disease Control Headquarters, Korea Disease Control and Prevention Agency, Cheongju, Republic of Korea
| |
Collapse
|
29
|
Asmar I, Almahmoud O, Yaseen K, Jamal J, Omar A, Naseef H, Hasan S. Assessment of immunoglobin G (spike and nucleocapsid protein) response to COVID-19 vaccination in Palestine. CLINICAL EPIDEMIOLOGY AND GLOBAL HEALTH 2023; 22:101330. [PMID: 37293133 PMCID: PMC10239151 DOI: 10.1016/j.cegh.2023.101330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/13/2023] [Accepted: 05/25/2023] [Indexed: 06/10/2023] Open
Abstract
INTRODUCTION Many countries have begun immunization programs and established protocols to combat pandemics caused by the SARS-CoV-2 virus. Six months after vaccination, the antibody titers produced by the immunization begin to decline, and individuals whose first immunization (either one or two doses) did not provide adequate protection may require a booster dose. METHODS A quantitative cross-sectional survey of 18-year-olds and older was undertaken in the West Bank from June 15 to June 27, 2022. Each participant had 5 mL of blood drawn to be tested for IgG-S, IgG-N, and blood group. RESULTS All participants had positive IgG-S results; IgG-S values ranged between 77 and 40,000 AU/ml, with a mean value of 1254 AU/ml. The value of IgG-N ranged from 0 to 139.3 U/ml for all participants, with a mean value of 22.4 U/ml. 64 (37.2%) of the participants demonstrated positive IgG-N screening results, with mean values of 51.2 U/ml. Female participants' mean IgG concentration was higher than male participants. Furthermore, the results revealed that smokers had lower levels of vaccine-induced antibodies than nonsmokers. High significance was found in the time from the last vaccine till the blood sample test (T = 3.848, P < .001), and the group between 6 and 9 months was found to have higher mean values than the 9-months group (M = 15952). CONCLUSIONS Participants vaccinated with a higher number of vaccines tend to have higher IgG-S. To elevate total antibodies, booster doses are essential. Additional researchers are needed to examine the positive correlation between IgG-S and IgG-N.
Collapse
Affiliation(s)
- Imad Asmar
- Birzeit University, Palestine
- Department of Nursing, P. O. Box Birzeit 14, Palestine
| | - Omar Almahmoud
- Birzeit University, Palestine
- Department of Nursing, P. O. Box Birzeit 14, Palestine
| | - Khalid Yaseen
- Birzeit University, Palestine
- Department of Nursing, P. O. Box Birzeit 14, Palestine
| | - Jehad Jamal
- Birzeit University, Palestine
- Department of Nursing, P. O. Box Birzeit 14, Palestine
| | - Ahmad Omar
- Birzeit University, Palestine
- Department of Nursing, P. O. Box Birzeit 14, Palestine
| | - Hani Naseef
- Birzeit University, Palestine
- Pharmacy Department, P. O. Box Birzeit 14, Palestine
| | - Shadi Hasan
- Birzeit University, Palestine
- Master program in Clinical Laboratory Science, P. O. Box Birzeit 14, Palestine
| |
Collapse
|
30
|
Giuliano A, Kuter B, Pilon-Thomas S, Whiting J, Mo Q, Leav B, Sirak B, Cubitt C, Dukes C, Isaacs-Soriano K, Kennedy K, Ball S, Dong N, Jain A, Hwu P, Lancet J. Safety and immunogenicity of a third dose of mRNA-1273 vaccine among cancer patients. Cancer Commun (Lond) 2023. [PMID: 37377402 PMCID: PMC10354405 DOI: 10.1002/cac2.12453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 05/15/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Compared to the general population, cancer patients are at higher risk of morbidity and mortality following SARS-CoV-2 infection. The immune response to a two-dose regimen of mRNA vaccines in cancer patients is generally lower than in immunocompetent individuals. Booster doses may meaningfully augment immune response in this population. We conducted an observational study with the primary objective of determining the immunogenicity of vaccine dose three (100 μg) of mRNA-1273 among cancer patients and a secondary objective of evaluating safety at 14 and 28 days. METHODS The mRNA-1273 vaccine was administered ∼7 to 9 months after administering two vaccine doses (i.e., the primary series). Immune responses (enzyme-linked immunosorbent assay [ELISA]) were assessed 28 days post-dose three. Adverse events were collected at days 14 (± 5) and 28 (+5) post-dose three. Fisher exact or X2 tests were used to compare SARS-CoV-2 antibody positivity rates, and paired t-tests were used to compare SARS-CoV-2 antibody geometric mean titers (GMTs) across different time intervals. RESULTS Among 284 adults diagnosed with solid tumors or hematologic malignancies, dose three of mRNA-1273 increased the percentage of patients seropositive for SARS-CoV-2 antibody from 81.7% pre-dose three to 94.4% 28 days post-dose three. GMTs increased 19.0-fold (15.8-22.8). Patients with lymphoid cancers or solid tumors had the lowest and highest antibody titers post-dose three, respectively. Antibody responses after dose three were reduced among those who received anti-CD20 antibody treatment, had lower total lymphocyte counts and received anticancer therapy within 3 months. Among patients seronegative for SARS-CoV-2 antibody pre-dose three, 69.2% seroconverted after dose three. A majority (70.4%) experienced mostly mild, transient adverse reactions within 14 days of dose three, whereas severe treatment-emergent events within 28 days were very rare (<2%). CONCLUSION Dose three of the mRNA-1273 vaccine was well-tolerated and augmented SARS-CoV-2 seropositivity in cancer patients, especially those who did not seroconvert post-dose two or whose GMTs significantly waned post-dose two. Lymphoid cancer patients experienced lower humoral responses to dose three of the mRNA-1273 vaccine, suggesting that timely access to boosters is important for this population.
Collapse
Affiliation(s)
- Anna Giuliano
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | | | - Junmin Whiting
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, USA
| | - Qianxing Mo
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Bradley Sirak
- Department of Cancer Epidemiology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Christopher Dukes
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | | | - Kayoko Kennedy
- Non-Therapeutic Research Office (NTRO), Moffitt Cancer Center, Tampa, Florida, USA
| | - Somedeb Ball
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Ning Dong
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Akriti Jain
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Patrick Hwu
- Department of Immunology, Moffitt Cancer Center, Tampa, Florida, USA
| | - Jeffrey Lancet
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
31
|
Emeksiz HC, Hepokur MN, Şahin SE, Şirvan BN, Çiçek B, Önder A, Yıldız M, Aksakal DK, Bideci A, Ovalı HF, İşman F. Immunogenicity, safety and clinical outcomes of the SARS-CoV-2 BNT162b2 vaccine in adolescents with type 1 diabetes. Front Pediatr 2023; 11:1191706. [PMID: 37435175 PMCID: PMC10331611 DOI: 10.3389/fped.2023.1191706] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Introduction The mRNA-based BNT162b2 (Pfizer-BioNTech) vaccine has been shown to elicit robust systemic immune response and confer substantial protection against the severe coronavirus disease (COVID-19), with a favorable safety profile in adolescents. However, no data exist regarding immunogenicity, reactogenicity and clinical outcomes of COVID-19 vaccines in adolescents with type 1 diabetes (T1D). In this prospective observational cohort study, we examined the humoral immune responses and side effects induced by the BNT162b2 vaccine, as well as, the rate and symptomatology of laboratory-confirmed COVID-19 vaccine breakthrough infections after completion of dual-dose BNT162b2 vaccination in adolescents with T1D and compared their data with those of healthy control adolescents. The new data obtained after the vaccination of adolescents with T1D could guide their further COVID-19 vaccination schedule. Methods A total of 132 adolescents with T1D and 71 controls were enrolled in the study, of whom 81 COVID-19 infection-naive adolescents with T1D (patient group) and 40 COVID-19 infection-naive controls (control group) were eligible for the final analysis. The response of participants to the BNT162b2 vaccine was assessed by measuring their serum IgG antibodies to the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), 4-6 weeks after the receipt of first and second vaccine doses. Data about the adverse events of the vaccine was collected after the receipt of each vaccine dose. The rate of COVID-19 vaccine breakthrough infections was evaluated in the 6-month period following second vaccination. Results After vaccinations, adolescents with T1D and controls exhibited similar, highly robust increments in anti-SARS-CoV-2 IgG titers. All the participants in the patient and control groups developed anti-SARS-CoV-2 IgG titers over 1,050 AU/ml after the second vaccine dose which is associated with a neutralizing effect. None of the participants experienced severe adverse events. The rate of breakthrough infections in the patient group was similar to that in the control group. Clinical symptomatology was mild in all cases. Conclusion Our findings suggest that two-dose BNT162b2 vaccine administered to adolescents with T1D elicits robust humoral immune response, with a favorable safety profile and can provide protection against severe SARS-CoV-2 infection similar to that in healthy adolescents.
Collapse
Affiliation(s)
- Hamdi Cihan Emeksiz
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Merve Nur Hepokur
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Sibel Ergin Şahin
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Banu Nursoy Şirvan
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Burçin Çiçek
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Aşan Önder
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Metin Yıldız
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Derya Karaman Aksakal
- Department of Pediatric Endocrinology, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Aysun Bideci
- Department of Pediatric Endocrinology, Gazi University Hospital, Ankara, Türkiye
| | - Hüsnü Fahri Ovalı
- Department of Pediatrics, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| | - Ferruh İşman
- Department of Biochemistry, Professor Doctor Süleyman Yalçın City Hospital, Istanbul Medeniyet University, Istanbul, Türkiye
| |
Collapse
|
32
|
Serra N, Andriolo M, Butera I, Mazzola G, Sergi CM, Fasciana TMA, Giammanco A, Gagliano MC, Cascio A, Di Carlo P. A Serological Analysis of the Humoral Immune Responses of Anti-RBD IgG, Anti-S1 IgG, and Anti-S2 IgG Levels Correlated to Anti-N IgG Positivity and Negativity in Sicilian Healthcare Workers (HCWs) with Third Doses of the mRNA-Based SARS-CoV-2 Vaccine: A Retrospective Cohort Study. Vaccines (Basel) 2023; 11:1136. [PMID: 37514952 PMCID: PMC10384738 DOI: 10.3390/vaccines11071136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/07/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND With SARS-CoV-2 antibody tests on the market, healthcare providers must be confident that they can use the results to provide actionable information to understand the characteristics and dynamics of the humoral response and antibodies (abs) in SARS-CoV-2-vaccinated patients. In this way, the study of the antibody responses of healthcare workers (HCWs), a population that is immunocompetent, adherent to vaccination, and continuously exposed to different virus variants, can help us understand immune protection and determine vaccine design goals. METHODS We retrospectively evaluated antibody responses via multiplex assays in a sample of 538 asymptomatic HCWs with a documented complete vaccination cycle of 3 doses of mRNA vaccination and no previous history of infection. Our sample was composed of 49.44% males and 50.56% females, with an age ranging from 21 to 71 years, and a mean age of 46.73 years. All of the HCWs' sera were collected from April to July 2022 at the Sant'Elia Hospital of Caltanissetta to investigate the immunologic responses against anti-RBD, anti-S1, anti-S2, and anti-N IgG abs. RESULTS A significant difference in age between HCWs who were positive and negative for anti-N IgG was observed. For anti-S2 IgG, a significant difference between HCWs who were negative and positive compared to anti-N IgG was observed only for positive HCWs, with values including 10 (U/mL)-100 (U/mL); meanwhile, for anti-RBD IgG and anti-S1 IgG levels, there was only a significant difference observed for positive HCWs with diluted titers. For the negative values of anti-N IgG, among the titer dilution levels of anti-RBD, anti-S1, and anti-S2 IgG, the anti-S2 IgG levels were significantly lower than the anti-RBD and anti-S1 levels; in addition, the anti-S1 IgG levels were significantly lower than the anti-RBD IgG levels. For the anti-N IgG positive levels, only the anti-S2 IgG levels were significantly lower than the anti-RBD IgG and anti-S1 IgG levels. Finally, a logistic regression analysis showed that age and anti-S2 IgG were negative and positive predictors of anti-N IgG levels, respectively. The analysis between the vaccine type and mixed mRNA combination showed higher levels of antibodies in mixed vaccinated HCWs. This finding disappeared in the anti-N positive group. CONCLUSIONS Most anti-N positive HCWs showed antibodies against the S2 domain and were young subjects. Therefore, the authors suggest that including the anti-SARS-CoV-2-S2 in antibody profiles can serve as a complementary testing approach to qRT-PCR for the early identification of asymptomatic infections in order to reduce the impact of potential new SARS-CoV-2 variants. Our serological investigation on the type of mRNA vaccine and mixed mRNA vaccines shows that future investigations on the serological responses in vaccinated asymptomatic patients exposed to previous infection or reinfection are warranted for updated vaccine boosters.
Collapse
Affiliation(s)
- Nicola Serra
- Department of Public Health, University Federico II of Naples, 80131 Napoli, Italy
| | - Maria Andriolo
- Clinical Pathology Laboratory, Provincial Health Authority of Caltanissetta, 93100 Caltanissetta, Italy
| | - Ignazio Butera
- Degree Course in Medicine and Surgery, Medical Scholl of Hypatia, University of Palermo, 93100 Caltanissetta, Italy
| | - Giovanni Mazzola
- Infectious Disease Unit, Provincial Health Authority of Caltanissetta, 93100 Caltanissetta, Italy
| | - Consolato Maria Sergi
- Department of Pathology and Laboratory Medicine, University of Ottawa, 401 Smyth Road, Ottawa, ON K1H 8L1, Canada
| | - Teresa Maria Assunta Fasciana
- Department of Health Promotion, Maternal-Childhood, Internal Medicine of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| | - Anna Giammanco
- Department of Health Promotion, Maternal-Childhood, Internal Medicine of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| | - Maria Chiara Gagliano
- Infectious Disease Unit, Department of Health Promotion, Maternal-Childhood, Internal Medicine of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| | - Antonio Cascio
- Infectious Disease Unit, Department of Health Promotion, Maternal-Childhood, Internal Medicine of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| | - Paola Di Carlo
- Infectious Disease Unit, Department of Health Promotion, Maternal-Childhood, Internal Medicine of Excellence “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
33
|
Batra G, Murugesan DR, Raghavan S, Chattopadhyay S, Mehdi F, Ayushi, Gosain M, Singh S, Das SJ, Deshpande S, Sonar S, Jakhar K, Bhattacharya J, Mani S, Pandey AK, Sankalp, Goswami S, Das A, Dwivedi T, Sharma N, Kumar S, Sharma P, Kapoor S, Kshetrapal P, Wadhwa N, Thiruvengadam R, Kumar R, Gupta R, Garg PK, Bhatnagar S. Trends of humoral immune responses to heterologous antigenic exposure due to vaccination & omicron SARS-CoV-2 infection: Implications for boosting. Indian J Med Res 2023; 157:509-518. [PMID: 37322634 PMCID: PMC10466496 DOI: 10.4103/ijmr.ijmr_2521_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Indexed: 06/17/2023] Open
Abstract
Background & objectives Vaccination and natural infection can both augment the immune responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but how omicron infection has affected the vaccine-induced and hybrid immunity is not well studied in Indian population. The present study was aimed to assess the durability and change in responses of humoral immunity with age, prior natural infection, vaccine type and duration with a minimum gap of six months post-two doses with either ChAdOx1 nCov-19 or BBV152 prior- and post-emergence of the omicron variant. Methods A total of 1300 participants were included in this observational study between November 2021 and May 2022. Participants had completed at least six months after vaccination (2 doses) with either ChAdOx1 nCoV-19 or an inactivated whole virus vaccine BBV152. They were grouped according to their age (≤ or ≥60 yr) and prior exposure of SARS-CoV-2 infection. Five hundred and sixteen of these participants were followed up after emergence of the Omicron variant. The main outcome was durability and augmentation of the humoral immune response as determined by anti-receptor-binding domain (RBD) immunoglobulin G (IgG) concentrations, anti-nucleocapsid antibodies and anti-omicron RBD antibodies. Live virus neutralization assay was conducted for neutralizing antibodies against four variants - ancestral, delta and omicron and omicron sublineage BA.5. Results Before the omicron surge, serum anti-RBD IgG antibodies were detected in 87 per cent participants after a median gap of eight months from the second vaccine dose, with a median titre of 114 [interquartile range (IQR) 32, 302] BAU/ml. The levels increased to 594 (252, 1230) BAU/ml post-omicron surge (P<0.001) with 97 per cent participants having detectable antibodies, although only 40 had symptomatic infection during the omicron surge irrespective of vaccine type and previous history of infection. Those with prior natural infection and vaccination had higher anti-RBD IgG titre at baseline, which increased further [352 (IQR 131, 869) to 816 (IQR 383, 2001) BAU/ml] (P<0.001). The antibody levels remained elevated after a mean time gap of 10 months, although there was a decline of 41 per cent. The geometric mean titre was 452.54, 172.80, 83.1 and 76.99 against the ancestral, delta, omicron and omicron BA.5 variants in the live virus neutralization assay. Interpretation & conclusions Anti-RBD IgG antibodies were detected in 85 per cent of participants after a median gap of eight months following the second vaccine dose. Omicron infection probably resulted in a substantial proportion of asymptomatic infection in the first four months in our study population and boosted the vaccine-induced humoral immune response, which declined but still remained durable over 10 months.
Collapse
Affiliation(s)
- Gaurav Batra
- Centre for Bio Design and Diagnostics, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Deepika Rathna Murugesan
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sreevatsan Raghavan
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Souvick Chattopadhyay
- Centre for Bio Design and Diagnostics, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Farha Mehdi
- Centre for Bio Design and Diagnostics, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ayushi
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Mudita Gosain
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Savita Singh
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Soon Jyoti Das
- Centre for Bio Design and Diagnostics, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Suprit Deshpande
- Centre for Viral Therapeutics and Vaccine, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Sudipta Sonar
- Centre for Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Kamini Jakhar
- Centre for Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Jayanta Bhattacharya
- Centre for Viral Therapeutics and Vaccine, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Shailendra Mani
- Centre for Infection and Immunity, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Anil Kumar Pandey
- Department of Physiology, ESIC Medical College & Hospital, Faridabad, Haryana, India
| | - Sankalp
- Department of Physiology, ESIC Medical College & Hospital, Faridabad, Haryana, India
| | - Shweta Goswami
- Department of Community Medicine, ESIC Medical College & Hospital, Faridabad, Haryana, India
| | - Asim Das
- ESIC Medical College & Hospital, Faridabad, Haryana, India
| | - Tanima Dwivedi
- Department of Laboratory Medicine, National Cancer Center, All India Institute of Medical Science, Jhajjar, Haryana, India
| | - Nandini Sharma
- Department of Community Medicine, Maulana Azad Medical College & Lok Nayak Hospital, New Delhi, India
| | - Suresh Kumar
- Maulana Azad Medical College & Lok Nayak Hospital, New Delhi, India
| | - Pragya Sharma
- Andaman & Nicobar Islands Institute of Medical Sciences, Port Blair, Andaman and Nicobar Islands, India
| | - Seema Kapoor
- Department of Pediatrics, Maulana Azad Medical College & Lok Nayak Hospital, New Delhi, India
| | - Pallavi Kshetrapal
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Nitya Wadhwa
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ramachandran Thiruvengadam
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Rakesh Kumar
- Centre for Community Medicine, All India Institute of Medical Science, New Delhi, India
| | - Ritu Gupta
- Department of Laboratory Oncology, Dr. B.R.A Institute-Rotary Cancer Hospital, New Delhi, India
| | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Shinjini Bhatnagar
- Centre for Maternal and Child Health, Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | | |
Collapse
|
34
|
Allen JC, Toapanta FR, Baliban SM, Sztein MB, Tennant SM. Reduced immunogenicity of a live Salmonella enterica serovar Typhimurium vaccine in aged mice. Front Immunol 2023; 14:1190339. [PMID: 37207226 PMCID: PMC10188964 DOI: 10.3389/fimmu.2023.1190339] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 04/13/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction Non-typhoidal Salmonella (NTS) is responsible for a high burden of foodborne infections and deaths worldwide. In the United States, NTS infections are the leading cause of hospitalizations and deaths due to foodborne illnesses, and older adults (≥65 years) are disproportionately affected by Salmonella infections. Due to this public health concern, we have developed a live attenuated vaccine, CVD 1926 (I77 ΔguaBA ΔclpP ΔpipA ΔhtrA), against Salmonella enterica serovar Typhimurium, a common serovar of NTS. Little is known about the effect of age on oral vaccine responses, and due to the decline in immune function with age, it is critical to evaluate vaccine candidates in older age groups during early product development. Methods In this study, adult (six-to-eight-week-old) and aged (18-month-old) C57BL/6 mice received two doses of CVD 1926 (109 CFU/dose) or PBS perorally, and animals were evaluated for antibody and cell-mediated immune responses. A separate set of mice were immunized and then pre-treated with streptomycin and challenged orally with 108 CFU of wild-type S. Typhimurium SL1344 at 4 weeks postimmunization. Results Compared to PBS-immunized mice, adult mice immunized with CVD 1926 had significantly lower S. Typhimurium counts in the spleen, liver, and small intestine upon challenge. In contrast, there were no differences in bacterial loads in the tissues of vaccinated versus PBS aged mice. Aged mice exhibited reduced Salmonella-specific antibody titers in the serum and feces following immunization with CVD 1926 compared to adult mice. In terms of T cell responses (T-CMI), immunized adult mice showed an increase in the frequency of IFN-γ- and IL-2-producing splenic CD4 T cells, IFN-γ- and TNF-α-producing Peyer's Patch (PP)-derived CD4 T cells, and IFN-γ- and TNF-α-producing splenic CD8 T cells compared to adult mice administered PBS. In contrast, in aged mice, T-CMI responses were similar in vaccinated versus PBS mice. CVD 1926 elicited significantly more PP-derived multifunctional T cells in adult compared to aged mice. Conclusion These data suggest that our candidate live attenuated S. Typhimurium vaccine, CVD 1926, may not be sufficiently protective or immunogenic in older humans and that mucosal responses to live-attenuated vaccines decrease with increasing age.
Collapse
Affiliation(s)
- Jessica C. Allen
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Franklin R. Toapanta
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Scott M. Baliban
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
35
|
Syrimi N, Sourri F, Giannakopoulou MC, Karamanis D, Pantousas A, Georgota P, Rokka E, Vladeni Z, Tsiantoula E, Soukara E, Lavda N, Gkaragkanis D, Zisaki A, Vakalidis P, Goula V, Loupou E, Palaiodimos L, Hatzigeorgiou D. Humoral and Cellular Response and Associated Variables Nine Months following BNT162b2 Vaccination in Healthcare Workers. J Clin Med 2023; 12:jcm12093172. [PMID: 37176612 PMCID: PMC10179201 DOI: 10.3390/jcm12093172] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/15/2023] Open
Abstract
In this study, we aimed to illustrate the trajectory of humoral and cellular immunity nine months after primary vaccination with the BNT162b2 mRNA vaccine among 189 healthcare workers (HCWs). Additionally, we endeavored to identify correlations between immunity parameters and a number of common variables and comorbidities. A total of 189 healthcare workers (HCWs), vaccinated against COVID-19, were finally included in the study. All of the subjects had received two doses of the BNT162b2 vaccine; had undergone antibody tests one, four and nine months post-vaccination; and had completed a medical questionnaire. Further samples taken at nine months were tested for cellular immunity. No participants had evidence of COVID-19 infection pre- or post-vaccination. An anti-S1 receptor binding domain (RBD) antibody assay was used to assess humoral response, and cellular immunity was estimated with an INF-γ release assay (IGRA). Statistical analysis was performed using STATA. We report a statistically significant antibody drop over time. Being above the age of 40 or a smoker reduces the rise of antibodies by 37% and 28%, respectively. More than half of the participants did not demonstrate T-cell activation at nine months. Female gender and antibody levels at four months predispose detection of cellular immunity at nine months post-immunization. This study furthers the qualitative, quantitative, and temporal understanding of the immune response to the BNT162b2 mRNA vaccine and the effect of correlated factors.
Collapse
Affiliation(s)
- Natalia Syrimi
- Paediatric Department, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
- Infection Prevention and Control Department, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Flora Sourri
- Infection Prevention and Control Department, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Maria-Christina Giannakopoulou
- COVID-19 Ward, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
- Medical Directorate, Hellenic National and Defence General Staff, Mesogeion 227-231, 15561 Athens, Greece
| | - Dimitrios Karamanis
- Department of Health Informatics, Rutgers School of Health Professions, 65 Bergen St., Newark, NJ 07107, USA
- Department of Economics, University of Piraeus, Karaoli and Dimitriou 80, 18534 Piraeus, Greece
| | - Asterios Pantousas
- Department of Electrical and Computer Engineering, Democritus University of Thrace, 69100 Komotini, Greece
| | - Persefoni Georgota
- Immunology Laboratory, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Eleni Rokka
- Oncology Ward, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Zoe Vladeni
- Infection Prevention and Control Department, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Euaggelia Tsiantoula
- Biochemistry Laboratory, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Evangelia Soukara
- COVID-19 Ward, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Nikoletta Lavda
- COVID-19 Ward, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Dimitrios Gkaragkanis
- COVID-19 Ward, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Aikaterini Zisaki
- Infection Prevention and Control Department, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Panagiotis Vakalidis
- Biochemistry Laboratory, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Vasiliki Goula
- Biochemistry Laboratory, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Evdokia Loupou
- Biochemistry Laboratory, 251 Hellenic Air Force General Hospital, P. Kanellopoulou Avenue, 11525 Athens, Greece
| | - Leonidas Palaiodimos
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Dimitrios Hatzigeorgiou
- Medical Directorate, Hellenic National and Defence General Staff, Mesogeion 227-231, 15561 Athens, Greece
| |
Collapse
|
36
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.10.23288360. [PMID: 37090559 PMCID: PMC10120804 DOI: 10.1101/2023.04.10.23288360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Since the emergence of SARS-CoV-2, research has shown that adult patients mount broad and durable immune responses to infection. However, response to infection remains poorly studied in infants/young children. In this study, we evaluated humoral responses to SARS-CoV-2 in 23 infants/young children before and after infection. We found that antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with Spike and RBD IgG antibody half-life nearly 4X as long as in adults. The functional breadth of adult and infant/young children SARS-CoV-2 responses were comparable, with similar reactivity against panel of recent and previously circulating viral variants. Notably, IgG subtype analysis revealed that while IgG1 formed the majority of both adults' and infants/young children's response, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
|
37
|
Mallory M, Munt JE, Narowski TM, Castillo I, Cuadra E, Pisanic N, Fields P, Powers JM, Dickson A, Harris R, Wargowsky R, Moran S, Allabban A, Raphel K, McCaffrey TA, Brien JD, Heaney CD, Lafleur JE, Baric RS, Premkumar L. Longitudinal Analysis of Humoral and Cellular Immune Response Following SARS-CoV-2 Vaccination Supports Utilizing Point-Of-Care Tests to Enhance COVID-19 Booster Uptake. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.03.23287498. [PMID: 37066219 PMCID: PMC10104219 DOI: 10.1101/2023.04.03.23287498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Individuals with weaker neutralizing responses show reduced protection with SARS-CoV-2 variants. Booster vaccines are recommended for vaccinated individuals, but the uptake is low. We present the feasibility of utilizing point-of-care tests (POCT) to support evidence-based decision-making around COVID-19 booster vaccinations. Using infectious virus neutralization, ACE2 blocking, spike binding, and TCR sequencing assays, we investigated the dynamics of changes in the breadth and depth of blood and salivary antibodies as well as T-cell clonal response following mRNA vaccination in a cohort of healthcare providers. We evaluated the accuracy of two POCTs utilizing either blood or saliva to identify those in whom humoral immunity was inadequate. >4 months after two doses of mRNA vaccine, SARS-CoV-2 binding and neutralizing Abs (nAbs) and T-cell clones declined 40-80%, and 2/3rd lacked Omicron nAbs. After the third mRNA booster, binding and neutralizing Abs increased overall in the systemic compartment; notably, individuals with previously weak nAbs gained sharply. The third dose failed to stimulate secretory IgA, but salivary IgG closely tracked systemic IgG levels. Vaccine boosting increased Ab breadth against a divergent bat sarbecovirus, SHC014, although the TCR-beta sequence breadth was unchanged. Post 3rd booster dose, Ab avidity increased for the Wuhan and Delta strains, while avidity against Omicron and SHC014 increased to levels seen for Wuhan after the second dose. Negative results on POCTs strongly correlated with a lack of functional humoral immunity. The third booster dose helps vaccinees gain depth and breadth of systemic Abs against evolving SARS-CoV-2 and related viruses. Our findings show that POCTs are useful and easy-to-access tools to inform inadequate humoral immunity accurately. POCTs designed to match the circulating variants can help individuals with booster vaccine decisions and could serve as a population-level screening platform to preserve herd immunity. One Sentence Summary SARS-CoV-2 point-of-care antibody tests are valuable and easy-to-access tools to inform inadequate humoral immunity and to support informed decision-making regarding the current and future booster vaccination.
Collapse
|
38
|
Kim HM, Lee EJ, O SW, Choi YJ, Lee H, Oh SJ, Kim JM, Park AK, Kim JA, Lee CY, Kim JM, Park H, Park YJ, Yu JH, Kim EY, Ko HP, Kim EJ. A Seroprevalence Study on Residents in a Senior Care Facility with Breakthrough SARS-CoV-2 Omicron Infection. Viral Immunol 2023; 36:203-208. [PMID: 36951666 PMCID: PMC10621659 DOI: 10.1089/vim.2022.0133] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023] Open
Abstract
The Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) began spreading rapidly in the community in November 2021, becoming the dominant variant in the Republic of Korea in 2022. Although its pathogenesis in healthy individuals was low, the severity and hospitalization rate was higher in the elderly and immunocompromised patients. We aimed to investigate the immunogenicity in acute and convalescent phases of breakthrough infection by Omicron in elderly individuals. Serological data were assessed by electrochemiluminescence immunoassay, enzyme-linked immunosorbent assay, and plaque-reduction neutralization tests. SARS-CoV-2-specific antibody and immunoglobulin G levels in the acute phase were higher in third dose-vaccinated elderly than in first and second dose-vaccinated patients. The neutralization antibody titer was detected only in third dose-vaccinated patients, and the titer was higher for the Delta than the Omicron variant. In the convalescent phase of Omicron infection, the neutralization antibody titer of vaccinated patients was higher for the Delta than the Omicron variant except in unvaccinated individuals. We demonstrated that the cause of the vulnerability to Omicron variant infection in third dose-vaccinated elderly was due to the low neutralization antibody level against Omicron. A fourth dose of vaccination is required in the elderly to reduce hospitalization and mortality caused by the Omicron variant.
Collapse
Affiliation(s)
- Heui Man Kim
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Eun Ju Lee
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Sang Won O
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Yong Jun Choi
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Hyeokjin Lee
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Sae Jin Oh
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Jeong-Min Kim
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Ae Kyung Park
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Jeong-Ah Kim
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Chae young Lee
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Jong Mu Kim
- Epidemiological Investigation Team, Epidemiological Investigation and Analysis Task Force, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Hanul Park
- Epidemiological Investigation Team, Epidemiological Investigation and Analysis Task Force, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Young Joon Park
- Epidemiological Investigation Team, Epidemiological Investigation and Analysis Task Force, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| | - Jeong-Hee Yu
- Honam Regional Center for Disease Control and Prevention, Gwangju-si, Republic of Korea
| | - Eun-Young Kim
- Honam Regional Center for Disease Control and Prevention, Gwangju-si, Republic of Korea
| | - Hwa-Pyeong Ko
- Gwangju Metropolitan Government, Gwangju-si, Republic of Korea
| | - Eun-Jin Kim
- Division of Emerging Infectious Diseases, Bureau of Infectious Diseases Diagnosis Control, Korea Disease Control and Prevention Agency (KDCA), Cheongju-si, Republic of Korea
| |
Collapse
|
39
|
Kalyoncu S, Yilmaz S, Kuyucu AZ, Sayili D, Mert O, Soyturk H, Gullu S, Akinturk H, Citak E, Arslan M, Taskinarda MG, Tarman IO, Altun GY, Ozer C, Orkut R, Demirtas A, Tilmensagir I, Keles U, Ulker C, Aralan G, Mercan Y, Ozkan M, Caglar HO, Arik G, Ucar MC, Yildirim M, Yildirim TC, Karadag D, Bal E, Erdogan A, Senturk S, Uzar S, Enul H, Adiay C, Sarac F, Ekiz AT, Abaci I, Aksoy O, Polat HU, Tekin S, Dimitrov S, Ozkul A, Wingender G, Gursel I, Ozturk M, Inan M. Process development for an effective COVID-19 vaccine candidate harboring recombinant SARS-CoV-2 delta plus receptor binding domain produced by Pichia pastoris. Sci Rep 2023; 13:5224. [PMID: 36997624 PMCID: PMC10062263 DOI: 10.1038/s41598-023-32021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/21/2023] [Indexed: 04/01/2023] Open
Abstract
Recombinant protein-based SARS-CoV-2 vaccines are needed to fill the vaccine equity gap. Because protein-subunit based vaccines are easier and cheaper to produce and do not require special storage/transportation conditions, they are suitable for low-/middle-income countries. Here, we report our vaccine development studies with the receptor binding domain of the SARS-CoV-2 Delta Plus strain (RBD-DP) which caused increased hospitalizations compared to other variants. First, we expressed RBD-DP in the Pichia pastoris yeast system and upscaled it to a 5-L fermenter for production. After three-step purification, we obtained RBD-DP with > 95% purity from a protein yield of > 1 g/L of supernatant. Several biophysical and biochemical characterizations were performed to confirm its identity, stability, and functionality. Then, it was formulated in different contents with Alum and CpG for mice immunization. After three doses of immunization, IgG titers from sera reached to > 106 and most importantly it showed high T-cell responses which are required for an effective vaccine to prevent severe COVID-19 disease. A live neutralization test was performed with both the Wuhan strain (B.1.1.7) and Delta strain (B.1.617.2) and it showed high neutralization antibody content for both strains. A challenge study with SARS-CoV-2 infected K18-hACE2 transgenic mice showed good immunoprotective activity with no viruses in the lungs and no lung inflammation for all immunized mice.
Collapse
Affiliation(s)
| | - Semiramis Yilmaz
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | | | - Dogu Sayili
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Olcay Mert
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | - Seyda Gullu
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | - Erhan Citak
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Merve Arslan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | | | | | - Ceren Ozer
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ridvan Orkut
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | | | - Umur Keles
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Lund University, Lund, Sweden
| | - Ceren Ulker
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Gizem Aralan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Yavuz Mercan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Muge Ozkan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Hasan Onur Caglar
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Erzurum Technical University, Erzurum, Turkey
| | - Gizem Arik
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Ankara Medipol University, Ankara, Turkey
| | - Mehmet Can Ucar
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Imperial College London, London, UK
| | | | | | | | - Erhan Bal
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir Tinaztepe University, Izmir, Turkey
| | - Aybike Erdogan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Serdar Uzar
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Hakan Enul
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Cumhur Adiay
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Fahriye Sarac
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | | | - Irem Abaci
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | - Ozge Aksoy
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | | | - Saban Tekin
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
- University of Health Sciences, Istanbul, Turkey
| | | | | | | | - Ihsan Gursel
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Mehmet Ozturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir Tinaztepe University, Izmir, Turkey
| | - Mehmet Inan
- Izmir Biomedicine and Genome Center, Izmir, Turkey.
- Akdeniz University, Antalya, Turkey.
| |
Collapse
|
40
|
Parker E, Thomas J, Roper KJ, Ijaz S, Edwards T, Marchesin F, Katsanovskaja K, Lett L, Jones C, Hardwick HE, Davis C, Vink E, McDonald SE, Moore SC, Dicks S, Jegatheesan K, Cook NJ, Hope J, Cherepanov P, McClure MO, Baillie JK, Openshaw PJM, Turtle L, Ho A, Semple MG, Paxton WA, Tedder RS, Pollakis G. SARS-CoV-2 antibody responses associate with sex, age and disease severity in previously uninfected people admitted to hospital with COVID-19: An ISARIC4C prospective study. Front Immunol 2023; 14:1146702. [PMID: 37056776 PMCID: PMC10087108 DOI: 10.3389/fimmu.2023.1146702] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/17/2023] Open
Abstract
The SARS-CoV-2 pandemic enables the analysis of immune responses induced against a novel coronavirus infecting immunologically naïve individuals. This provides an opportunity for analysis of immune responses and associations with age, sex and disease severity. Here we measured an array of solid-phase binding antibody and viral neutralising Ab (nAb) responses in participants (n=337) of the ISARIC4C cohort and characterised their correlation with peak disease severity during acute infection and early convalescence. Overall, the responses in a Double Antigen Binding Assay (DABA) for antibody to the receptor binding domain (anti-RBD) correlated well with IgM as well as IgG responses against viral spike, S1 and nucleocapsid protein (NP) antigens. DABA reactivity also correlated with nAb. As we and others reported previously, there is greater risk of severe disease and death in older men, whilst the sex ratio was found to be equal within each severity grouping in younger people. In older males with severe disease (mean age 68 years), peak antibody levels were found to be delayed by one to two weeks compared with women, and nAb responses were delayed further. Additionally, we demonstrated that solid-phase binding antibody responses reached higher levels in males as measured via DABA and IgM binding against Spike, NP and S1 antigens. In contrast, this was not observed for nAb responses. When measuring SARS-CoV-2 RNA transcripts (as a surrogate for viral shedding) in nasal swabs at recruitment, we saw no significant differences by sex or disease severity status. However, we have shown higher antibody levels associated with low nasal viral RNA indicating a role of antibody responses in controlling viral replication and shedding in the upper airway. In this study, we have shown discernible differences in the humoral immune responses between males and females and these differences associate with age as well as with resultant disease severity.
Collapse
Affiliation(s)
- Eleanor Parker
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Jordan Thomas
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Kelly J. Roper
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Samreen Ijaz
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
| | - Tansy Edwards
- Medical Research Council (MRC) International Statistics and Epidemiology Group, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Federica Marchesin
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Ksenia Katsanovskaja
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Lauren Lett
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Christopher Jones
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Hayley E. Hardwick
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Chris Davis
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Elen Vink
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Sarah E. McDonald
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Shona C. Moore
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Steve Dicks
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
- National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Keerthana Jegatheesan
- Blood Borne Virus Unit, Reference Department, UK Health Security Agency, London, United Kingdom
- National Health Service (NHS) Blood and Transplant, London, United Kingdom
| | - Nicola J. Cook
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Joshua Hope
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Myra O. McClure
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | | | - Lance Turtle
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Antonia Ho
- Medical Research Council, University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Malcolm G. Semple
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - William A. Paxton
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Richard S. Tedder
- Department of Infectious Disease, Imperial College London, London, United Kingdom
| | - Georgios Pollakis
- National Institute of Health and Care Research (NIHR) Health Protection Research Unit in Emerging and Zoonotic Infections, Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | | |
Collapse
|
41
|
Jugler C, Sun H, Nguyen K, Palt R, Felder M, Steinkellner H, Chen Q. A novel plant-made monoclonal antibody enhances the synergetic potency of an antibody cocktail against the SARS-CoV-2 Omicron variant. PLANT BIOTECHNOLOGY JOURNAL 2023; 21:549-559. [PMID: 36403203 PMCID: PMC9946148 DOI: 10.1111/pbi.13970] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/06/2022] [Accepted: 11/12/2022] [Indexed: 06/01/2023]
Abstract
This study describes a novel, neutralizing monoclonal antibody (mAb), 11D7, discovered by mouse immunization and hybridoma generation, against the parental Wuhan-Hu-1 RBD of SARS-CoV-2. We further developed this mAb into a chimeric human IgG and recombinantly expressed it in plants to produce a mAb with human-like, highly homogenous N-linked glycans that has potential to impart greater potency and safety as a therapeutic. The epitope of 11D7 was mapped by competitive binding with well-characterized mAbs, suggesting that it is a Class 4 RBD-binding mAb that binds to the RBD outside the ACE2 binding site. Of note, 11D7 maintains recognition against the B.1.1.529 (Omicron) RBD, as well neutralizing activity. We also provide evidence that this novel mAb may be useful in providing additional synergy to established antibody cocktails, such as Evusheld™ containing the antibodies tixagevimab and cilgavimab, against the Omicron variant. Taken together, 11D7 is a unique mAb that neutralizes SARS-CoV-2 through a mechanism that is not typical among developed therapeutic mAbs and by being produced in ΔXFT Nicotiana benthamiana plants, highlights the potential of plants to be an economic and safety-friendly alternative platform for generating mAbs to address the evolving SARS-CoV-2 crisis.
Collapse
Affiliation(s)
- Collin Jugler
- The Biodesign InstituteArizona State UniversityTempeArizonaUSA
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| | - Haiyan Sun
- The Biodesign InstituteArizona State UniversityTempeArizonaUSA
| | - Katherine Nguyen
- School of Molecular SciencesArizona State UniversityTempeArizonaUSA
| | - Roman Palt
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | | | - Herta Steinkellner
- Department of Applied Genetics and Cell BiologyUniversity of Natural Resources and Life SciencesViennaAustria
| | - Qiang Chen
- The Biodesign InstituteArizona State UniversityTempeArizonaUSA
- School of Life SciencesArizona State UniversityTempeArizonaUSA
| |
Collapse
|
42
|
Mwimanzi F, Lapointe HR, Cheung PK, Sang Y, Yaseen F, Kalikawe R, Datwani S, Burns L, Young L, Leung V, Ennis S, Brumme CJ, Montaner JSG, Dong W, Prystajecky N, Lowe CF, DeMarco ML, Holmes DT, Simons J, Niikura M, Romney MG, Brumme ZL, Brockman MA. Impact of Age and Severe Acute Respiratory Syndrome Coronavirus 2 Breakthrough Infection on Humoral Immune Responses After Three Doses of Coronavirus Disease 2019 mRNA Vaccine. Open Forum Infect Dis 2023; 10:ofad073. [PMID: 36910697 PMCID: PMC10003738 DOI: 10.1093/ofid/ofad073] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/08/2023] [Indexed: 02/11/2023] Open
Abstract
Background Longer-term immune response data after 3 doses of coronavirus disease 2019 (COVID-19) mRNA vaccine remain limited, particularly among older adults and after Omicron breakthrough infection. Methods We quantified wild-type- and Omicron-specific serum immunoglobulin (Ig)G levels, angiotensin-converting enzyme 2 displacement activities, and live virus neutralization up to 6 months after third dose in 116 adults aged 24-98 years who remained COVID-19 naive or experienced their first severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during this time. Results Among the 78 participants who remained COVID-19 naive throughout follow up, wild-type- and Omicron-BA.1-specific IgG concentrations were comparable between younger and older adults, although BA.1-specific responses were consistently significantly lower than wild-type-specific responses in both groups. Wild-type- and BA.1-specific IgG concentrations declined at similar rates in COVID-19-naive younger and older adults, with median half-lives ranging from 69 to 78 days. Antiviral antibody functions declined substantially over time in COVID-19-naive individuals, particularly in older adults: by 6 months, BA.1-specific neutralization was undetectable in 96% of older adults, versus 56% of younger adults. Severe acute respiratory syndrome coronavirus 2 infection, experienced by 38 participants, boosted IgG levels and neutralization above those induced by vaccination alone. Nevertheless, BA.1-specific neutralization remained significantly lower than wild-type, with BA.5-specific neutralization lower still. Higher Omicron BA.1-specific neutralization 1 month after third dose was an independent correlate of lower SARS-CoV-2 infection risk. Conclusions Results underscore the immune benefits of the third COVID-19 mRNA vaccine dose in adults of all ages and identify vaccine-induced Omicron-specific neutralization as a correlate of protective immunity. Systemic antibody responses and functions however, particularly Omicron-specific neutralization, decline rapidly in COVID-19-naive individuals, particularly in older adults, supporting the need for additional booster doses.
Collapse
Affiliation(s)
- Francis Mwimanzi
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Hope R Lapointe
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Peter K Cheung
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Yurou Sang
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Fatima Yaseen
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Rebecca Kalikawe
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Sneha Datwani
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Laura Burns
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
| | - Landon Young
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
| | - Victor Leung
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Siobhan Ennis
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | - Chanson J Brumme
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Julio S G Montaner
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Winnie Dong
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver, Canada
| | - Natalie Prystajecky
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
- British Columbia Centre for Disease Control Public Health Laboratory, Vancouver, Canada
| | - Christopher F Lowe
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Mari L DeMarco
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel T Holmes
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Janet Simons
- Division of Medical Microbiology and Virology, St. Paul's Hospital, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Masahiro Niikura
- Faculty of Health Sciences, Simon Fraser University, Burnaby, Canada
| | | | | | - Mark A Brockman
- Correspondence: Mark A. Brockman, PhD, Professor, Faculty of Health Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada ()
| |
Collapse
|
43
|
Kulkarni PS, Kadam A, Godbole S, Bhatt V, Raut A, Kohli S, Tripathi S, Kulkarni P, Ludam R, Prabhu M, Bavdekar A, Gogtay NJ, Meshram S, Kadhiravan T, Kar S, Narayana DA, Samuel C, Kulkarni G, Gaidhane A, Sathyapalan D, Raut S, Hadda V, Bhalla HL, Bhamare C, Dharmadhikari A, Plested JS, Cloney-Clarke S, Zhu M, Pryor M, Hamilton S, Thakar M, Shete A, Gautam M, Gupta N, Panda S, Shaligram U, Poonawalla CS, Bhargava B, Gunale B, Kapse D. Safety and immunogenicity of SII-NVX-CoV2373 (COVID-19 vaccine) in adults in a phase 2/3, observer-blind, randomised, controlled study. THE LANCET REGIONAL HEALTH. SOUTHEAST ASIA 2023; 10:100139. [PMID: 36647543 PMCID: PMC9833646 DOI: 10.1016/j.lansea.2022.100139] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND NVX-CoV2373, a Covid-19 vaccine was developed in the USA with ∼90% efficacy. The same vaccine is manufactured in India after technology transfer (called as SII-NVX-CoV2373), was evaluated in this phase 2/3 immuno-bridging study. METHODS This was an observer-blind, randomised, phase 2/3 study in 1600 adults. In phase 2, 200 participants were randomized 3:1 to SII-NVX-CoV2373 or placebo. In phase 3, 1400 participants were randomized 3:1 to SII-NVX-CoV2373 or NVX-CoV2373 (940 safety cohort and 460 immunogenicity cohort). Two doses of study products (SII-NVX-CoV2373, NVX-CoV2373 or placebo) were given 3 weeks apart. Primary objectives were to demonstrate non-inferiority of SII-NVX-CoV2373 to NVX-CoV2373 in terms of geometric mean ELISA units (GMEU) ratio of anti-S IgG antibodies 14 days after the second dose (day 36) and to determine the incidence of causally related serious adverse events (SAEs) through 180 days after the first dose. Anti-S IgG response was assessed using an Enzyme-Linked Immunosorbent Assay (ELISA) and neutralizing antibodies (nAb) were assessed by a microneutralization assay using wild type SARS CoV-2 in participants from the immunogenicity cohort at baseline, day 22, day 36 and day 180. Cell mediated immune (CMI) response was assessed in a subset of 28 participants from immunogenicity cohort by ELISpot assay at baseline, day 36 and day 180. The total follow-up was for 6 months. Trial registration: CTRI/2021/02/031554. FINDINGS Total 1596 participants (200 in Phase 2 and 1396 in Phase 3) received the first dose. SII-NVX-CoV2373 was found non-inferior to NVX-CoV2373 (anti-S IgG antibodies GMEU ratio 0.91; 95% CI: 0.79, 1.06). At day 36, there was more than 58-fold rise in anti-S IgG and nAb titers compared to baseline in both the groups. On day 180 visit, these antibody titers declined to levels slightly lower than those after the first dose (13-22 fold-rise above baseline). Incidence of unsolicited and solicited AEs was similar between the SII-NVX-CoV2373 and NVX-CoV2373 groups. No adverse event of special interest (AESI) was reported. No causally related SAE was reported. INTERPRETATION SII-NVX-CoV2373 induced a non-inferior immune response compared to NVX-CoV2373 and has acceptable safety profile. FUNDING SIIPL, Indian Council of Medical Research, Novavax.
Collapse
Affiliation(s)
| | - Abhijit Kadam
- Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | - Sheela Godbole
- Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | - Varsha Bhatt
- Dr. D. Y. Patil Medical College Hospital and Research Centre, Pune, India
| | - Abhishek Raut
- Mahatma Gandhi Institute of Medical Sciences, Sewagram, Wardha, India
| | - Sunil Kohli
- Hamdard Institute of Medical Sciences and Research, New Delhi, India
| | | | | | - Rakhi Ludam
- Institution of Medical Science and SUM Hospital, Bhubaneswar, India
| | - Madhav Prabhu
- KLES Dr. Prabhakar Kore Hospital and Medical Research Center, Belgavi, India
| | | | | | | | - Tamilarasu Kadhiravan
- Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, India
| | - Sonali Kar
- Kalinga Institute of Medical Sciences, Bhubaneswar, India
| | | | | | | | - Abhay Gaidhane
- Acharya Vinoba Bhave Rural Hospital and Datta Meghe Institute of Medical Sciences, Sawangi (M), Wardha, India
| | | | | | - Vijay Hadda
- All India Institute of Medical Sciences, New Delhi, India
| | | | | | | | | | | | - Mingzhu Zhu
- Clinical Immunology Laboratory, Novavax, Gaithersburg, MD, USA
| | - Melinda Pryor
- 360biolabs, 85 Commercial Road, Melbourne, Victoria, Australia
| | | | - Madhuri Thakar
- Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | - Ashwini Shete
- Indian Council of Medical Research-National AIDS Research Institute, Pune, India
| | | | | | - Samiran Panda
- Indian Council of Medical Research, New Delhi, India
| | | | | | | | | | | |
Collapse
|
44
|
Ellison TJ, Talbott GC, Henderson DR. Intradermal delivery of a quadrivalent cell-based seasonal influenza vaccine using an adjuvanted skin patch vaccination platform. Vaccine 2023; 41:304-314. [PMID: 36587961 DOI: 10.1016/j.vaccine.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/26/2022] [Accepted: 10/05/2022] [Indexed: 12/15/2022]
Abstract
All seasonal influenza vaccines for 2021-2022 in the US were quadrivalent and the market continues to be dominated by intramuscular delivery of non-adjuvanted, virion-derived antigens grown in chicken eggs. Up to four new egg-adapted production influenza vaccine strains must be generated each year. The introduction in 2012 of Flucelvax®, which is grown in mammalian suspension cell culture and uses vaccine production strains without adaptive mutations for efficient growth in eggs, represented a major advance in vaccine production technology. Here we demonstrate that Flucelvax can be reformulated and combined with a liposomal adjuvant containing QS-21 (Verndari Adjuvant System 1.1, VAS1.1) or QS-21 and 3D-PHAD (VAS1.2) for intradermal administration using a painless skin patch, VaxiPatch™. VAS1.2 is similar to AS01B, the adjuvant system used in Shingrix® and Mosquirix™. We show that Flucelvax, when reformulated and concentrated using tangential flow filtration (TFF), maintains hemagglutination and single radial immunodiffusion (SRID) potency. Loading the reformulated Flucelvax material onto VaxiPatch arrays conferred high levels of resistance to heat stress and room temperature stability. TFF enriched vaccine antigens were combined with VAS1.1 or VAS1.2 and dispensed in 10nL drops into the pockets of 36 (total 360 nL) stainless steel microneedles arranged in a microarray 1.2 cm in diameter. Using VaxiPatch delivery of 2 µg of antigen, we demonstrated intramusuclar-comparable IgG and hemagglutination inhibition (HAI) immune responses in Sprague Dawley® rats. With addition of VAS1.2, antigen-specific IgG titers were increased as much as 68-fold (47-fold for VAS1.1) with improvements in seroconversion for three of four strains (all four were improved by VAS1.1). TFF-reformulated antigens combined with VAS1.1 or VAS1.2 and delivered by VaxiPatch showed only minor skin reactogenicity after 1 h and no skin reactogenicity after 24 h. These data indicate that VaxiPatch and the VAS system have the potential to be transformative for vaccine delivery.
Collapse
Affiliation(s)
- Thomas J Ellison
- Verndari Inc., 2700 Stockton Blvd., Suite 1104, Sacramento, CA 95817, United States.
| | - George C Talbott
- Verndari Inc., 2700 Stockton Blvd., Suite 1104, Sacramento, CA 95817, United States.
| | - Daniel R Henderson
- Verndari Inc., 2700 Stockton Blvd., Suite 1104, Sacramento, CA 95817, United States.
| |
Collapse
|
45
|
Costiniuk CT, Singer J, Lee T, Langlois MA, Arnold C, Galipeau Y, Needham J, Kulic I, Jenabian MA, Burchell AN, Shamji H, Chambers C, Walmsley S, Ostrowski M, Kovacs C, Tan DH, Harris M, Hull M, Brumme ZL, Lapointe HR, Brockman MA, Margolese S, Mandarino E, Samarani S, Vulesevic B, Lebouché B, Angel JB, Routy JP, Cooper CL, Anis AH. COVID-19 vaccine immunogenicity in people with HIV. AIDS 2023; 37:F1-F10. [PMID: 36476452 PMCID: PMC9794000 DOI: 10.1097/qad.0000000000003429] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 10/31/2022] [Accepted: 11/01/2022] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Many vaccines require higher/additional doses or adjuvants to provide adequate protection for people with HIV (PWH). Our objective was to compare COVID-19 vaccine immunogenicity in PWH to HIV-negative individuals. DESIGN In a Canadian multi-center prospective, observational cohort of PWH receiving at least two COVID-19 vaccinations, we measured vaccine-induced immunity at 3 and 6 months post 2nd and 1-month post 3rd doses. METHODS The primary outcome was the percentage of PWH mounting vaccine-induced immunity [co-positivity for anti-IgG against SARS-CoV2 Spike(S) and receptor-binding domain proteins] 6 months post 2nd dose. Univariable and multivariable logistic regressions were used to compare COVID-19-specific immune responses between groups and within subgroups. RESULTS Data from 294 PWH and 267 controls were analyzed. Immunogenicity was achieved in over 90% at each time point in both groups. The proportions of participants achieving comparable anti-receptor-binding domain levels were similar between the group at each time point. Anti-S IgG levels were similar by group at month 3 post 2nd dose and 1-month post 3rd dose. A lower proportion of PWH vs. controls maintained vaccine-induced anti-S IgG immunity 6 months post 2nd dose [92% vs. 99%; odds ratio: 0.14 (95% confidence interval: 0.03, 0.80; P = 0.027)]. In multivariable analyses, neither age, immune non-response, multimorbidity, sex, vaccine type, or timing between doses were associated with reduced IgG response. CONCLUSION Vaccine-induced IgG was elicited in the vast majority of PWH and was overall similar between groups. A slightly lower proportion of PWH vs. controls maintained vaccine-induced anti-S IgG immunity 6 months post 2nd dose demonstrating the importance of timely boosting in this population.
Collapse
Affiliation(s)
- Cecilia T. Costiniuk
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Institute of the McGill University Health Centre
- Department of Microbiology and Immunology, McGill University, Montreal, QC
| | - Joel Singer
- School of Population and Public Health, University of British Columbia
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC
| | - Terry Lee
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa
| | - Corey Arnold
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa
| | - Yannick Galipeau
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa
| | - Judy Needham
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC
| | - Iva Kulic
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC
| | | | - Ann N. Burchell
- Department of Family and Community Medicine, St Michael's Hospital, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, ON
| | - Hasina Shamji
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Disease Control, Vancouver, BC
| | - Catharine Chambers
- Department of Family and Community Medicine, St Michael's Hospital, Unity Health Toronto and Dalla Lana School of Public Health, University of Toronto, Toronto, ON
| | - Sharon Walmsley
- Division of Infectious Diseases, Department of Medicine, University of Toronto
| | - Mario Ostrowski
- Clinical Sciences Division and Department of Immunology, University of Toronto, Li Ka Shing Knowledge Institute, St. Michael's Hospital
| | | | - Darrell H.S. Tan
- Division of Infectious Diseases, Department of Medicine, University of Toronto
- MAP Centre for Urban Health Solutions, St Michael's Hospital
- Institute of Public Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, ON
| | - Marianne Harris
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | - Mark Hull
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | - Zabrina L. Brumme
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
| | | | - Mark A. Brockman
- Faculty of Health Sciences, Simon Fraser University, Burnaby
- British Columbia Centre for Excellence in HIV/AIDS, Vancouver
- Department of Molecular Biology and Biochemistry, Faculty of Science, Simon Fraser University, Burnaby, BC
| | - Shari Margolese
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
| | - Enrico Mandarino
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
| | - Suzanne Samarani
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
| | - Branka Vulesevic
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, ON
| | - Bertrand Lebouché
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Institute of the McGill University Health Centre
- Department of Family Medicine, McGill University
- Canadian Institutes of Health Research Strategy for Patient-Oriented Research Mentorship Chair in Innovative Clinical Trials
| | - Jonathan B. Angel
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, ON
| | - Jean-Pierre Routy
- Division of Infectious Diseases/Chronic Viral Illness Service, McGill University Health Centre, Royal Victoria Hospital
- Infectious Diseases and Immunity in Global Health Research Institute of the McGill University Health Centre
- Division of Hematology, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
| | - Curtis L. Cooper
- Division of Infectious Diseases, Department of Medicine, University of Ottawa and the Ottawa Hospital Research Institute, Ottawa, ON
| | - Aslam H. Anis
- School of Population and Public Health, University of British Columbia
- Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN)
- Centre for Health Evaluation and Outcome Sciences, St. Paul's Hospital, Vancouver, BC
| |
Collapse
|
46
|
Neutralizing Antibody and T-Cell Responses against SARS-CoV-2 Wild-Type and Variants of Concern in Chronic Obstructive Pulmonary Disease Subjects after ChAdOx-1/ChAdOx-1 Homologous Vaccination: A Preliminary Study. Vaccines (Basel) 2022; 10:vaccines10122176. [PMID: 36560586 PMCID: PMC9781239 DOI: 10.3390/vaccines10122176] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Data on immunogenicity of adenovirus-vectored vaccine in chronic obstructive pulmonary disease (COPD) patients is limited. Therefore, we aimed to determine the humoral and cellular immune responses after homologous ChAdOx-1 vaccination in subjects with COPD. COPD subjects and age- and sex-matched healthy elderly receiving ChAdOx-1 homologous vaccination were included. The levels of neutralizing antibodies (NAb) and specific CD4 and CD8 T-cell responses against SARS-CoV-2 wild-type (WT) and variants of concern (VOCs: Alpha, Beta, Delta, and Omicron) were measured. Eight COPD patients were matched with eight control participants. After vaccination for 4 and 12 weeks, % inhibition of NAb against Alpha, Beta, and Delta in both groups were comparable and significantly higher than baseline. The percentage inhibition of NAb at the 12th week was significantly dropped from the 4th week in each group. The NAb against the Omicron variant, however, were much lower than Alpha, Beta, Delta variants. The increasing trend in the number of CD4 T-cells producing TNF-α, IFN-γ, IL-10, and FasL upon stimulation with spike peptides of WT and VOCs was observed in COPD patients compared to the healthy group. These responses were not observed in CD8 T-cells. Homologous ChAdOx-1 vaccination could induce comparable NAb against the SARS-CoV-2 WT, Alpha, Beta, Delta, and Omicron variants between COPD and healthy elderly. The CD4 T-cell responses did not differ between COPD patients and healthy control.
Collapse
|
47
|
Maeda H, Saito N, Igarashi A, Ishida M, Suami K, Yagiuchi A, Kimura Y, Komino M, Arai H, Morikawa T, Motohashi I, Miyazawa R, Moriyama T, Kamura H, Terada M, Kuwamitsu O, Hayakawa T, Sando E, Ohara Y, Teshigahara O, Suzuki M, Morimoto K. Effectiveness of Messenger RNA Coronavirus Disease 2019 Vaccines Against Symptomatic Severe Acute Respiratory Syndrome Coronavirus 2 Infections During the Delta Variant Epidemic in Japan: Vaccine Effectiveness Real-time Surveillance for SARS-CoV-2 (VERSUS). Clin Infect Dis 2022; 75:1971-1979. [PMID: 35438137 PMCID: PMC9047210 DOI: 10.1093/cid/ciac292] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/04/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Although high vaccine effectiveness of messenger RNA (mRNA) coronavirus disease 2019 (COVID-19) vaccines has been reported in studies in several countries, data are limited from Asian countries, especially against the Delta (B.1.617.2) variant. METHODS We conducted a multicenter test-negative case-control study in patients aged ≥16 years visiting hospitals or clinics with signs or symptoms consistent with COVID-19 from 1 July to 30 September 2021, when the Delta variant was dominant (≥90% of SARS-CoV-2 infections) nationwide in Japan. Vaccine effectiveness of BNT162b2 or mRNA-1273 against symptomatic SARS-CoV-2 infections was evaluated. Waning immunity among patients aged 16-64 years was also assessed. RESULTS We enrolled 1936 patients, including 396 test-positive cases and 1540 test-negative controls for SARS-CoV-2. The median age was 49 years, 53.4% were male, and 34.0% had underlying medical conditions. Full vaccination (receiving 2 doses ≥14 days before symptom onset) was received by 6.6% of cases and 38.8% of controls. Vaccine effectiveness of full vaccination against symptomatic SARS-CoV-2 infections was 88.7% (95% confidence interval [CI], 78.8%-93.9%) among patients aged 16-64 years and 90.3% (95% CI, 73.6%-96.4%) among patients aged ≥65 years. Among patients aged 16-64 years, vaccine effectiveness was 91.8% (95% CI, 80.3%-96.6%) within 1-3 months after full vaccination, and 86.4% (95% CI, 56.9%-95.7%) within 4-6 months. CONCLUSIONS mRNA COVID-19 vaccines had high effectiveness against symptomatic SARS-CoV-2 infections in Japan during July-September 2021, when the Delta variant was dominant nationwide.
Collapse
Affiliation(s)
- Haruka Maeda
- Department of Respiratory Infections, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
- Department of Clinical Tropical Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Nobuo Saito
- Department of Microbiology, Faculty of Medicine, Oita University, Oita, Japan
| | - Ataru Igarashi
- Unit of Public Health and Preventive Medicine, Yokohama City University School of Medicine, Kanagawa, Japan
- Department of Health Economics and Outcomes Research, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | | | - Yuya Kimura
- Saiseikai Kurihashi Hospital, Saitama, Japan
| | | | - Hiromi Arai
- Saiseikai Kurihashi Hospital, Saitama, Japan
| | | | | | - Rei Miyazawa
- Kawasaki Municipal Tama Hospital, Kanagawa, Japan
| | | | | | | | | | | | - Eiichiro Sando
- Kita-Fukushima Medical Center, Fukushima, Japan
- Fukushima Medical University, Fukushima, Japan
| | | | | | - Motoi Suzuki
- Infectious Disease Surveillance Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Konosuke Morimoto
- Department of Respiratory Infections, Institute of Tropical Medicine, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
48
|
Breznik JA, Huynh A, Zhang A, Bilaver L, Bhakta H, Stacey HD, Ang JC, Bramson JL, Nazy I, Miller MS, Denburg J, Costa AP, Bowdish DME. Cytomegalovirus Seropositivity in Older Adults Changes the T Cell Repertoire but Does Not Prevent Antibody or Cellular Responses to SARS-CoV-2 Vaccination. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1892-1905. [PMID: 36426948 PMCID: PMC9666329 DOI: 10.4049/jimmunol.2200369] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/03/2022] [Indexed: 12/14/2022]
Abstract
Chronic infection with human CMV may contribute to poor vaccine efficacy in older adults. We assessed the effects of CMV serostatus on Ab quantity and quality, as well as cellular memory recall responses, after two and three SARS-CoV-2 mRNA vaccine doses, in older adults in assisted living facilities. CMV serostatus did not affect anti-Spike and anti-receptor-binding domain IgG Ab levels, nor neutralization capacity against wild-type or β variants of SARS-CoV-2 several months after vaccination. CMV seropositivity altered T cell expression of senescence-associated markers and increased effector memory re-expressing CD45RA T cell numbers, as has been previously reported; however, this did not impact Spike-specific CD4+ T cell memory recall responses. CMV-seropositive individuals did not have a higher incidence of COVID-19, although prior infection influenced humoral immunity. Therefore, CMV seropositivity may alter T cell composition but does not impede the durability of humoral protection or cellular memory responses after SARS-CoV-2 mRNA vaccination in older adults.
Collapse
Affiliation(s)
- Jessica A Breznik
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Institute for Research on Aging, McMaster University, Hamilton, Ontario, Canada
| | - Angela Huynh
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Ali Zhang
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Lucas Bilaver
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hina Bhakta
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hannah D Stacey
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jann C Ang
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Jonathan L Bramson
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Ishac Nazy
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- McMaster Centre for Transfusion Research, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Judah Denburg
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Andrew P Costa
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
- Centre for Integrated Care, St. Joseph's Health System, Hamilton, Ontario, Canada; and
| | - Dawn M E Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
- Firestone Institute for Respiratory Health, St. Joseph's Healthcare, Hamilton, Ontario, Canada
| |
Collapse
|
49
|
Toledo-Romani ME, García-Carmenate M, Verdecia-Sánchez L, Pérez-Rodríguez S, Rodriguez-González M, Valenzuela-Silva C, Paredes-Moreno B, Sanchez-Ramirez B, González-Mugica R, Hernández-Garcia T, Orosa-Vázquez I, Díaz-Hernández M, Pérez-Guevara MT, Enriquez-Puertas J, Noa-Romero E, Palenzuela-Diaz A, Baro-Roman G, Mendoza-Hernández I, Muñoz Y, Gómez-Maceo Y, Santos-Vega BL, Fernandez-Castillo S, Climent-Ruiz Y, Rodríguez-Noda L, Santana-Mederos D, García-Vega Y, Chen GW, Doroud D, Biglari A, Boggiano-Ayo T, Valdés-Balbín Y, Rivera DG, García-Rivera D, Vérez-Bencomo V. Safety and immunogenicity of anti-SARS-CoV-2 heterologous scheme with SOBERANA 02 and SOBERANA Plus vaccines: Phase IIb clinical trial in adults. MED (NEW YORK, N.Y.) 2022; 3:760-773.e5. [PMID: 35998623 PMCID: PMC9359498 DOI: 10.1016/j.medj.2022.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 05/17/2022] [Accepted: 08/02/2022] [Indexed: 12/25/2022]
Abstract
BACKGROUND SOBERANA 02 has been evaluated in phase I and IIa studies comparing homologous versus heterologous schedule (this one, including SOBERANA Plus). Here, we report results of immunogenicity, safety, and reactogenicity of SOBERANA 02 in a two- or three-dose heterologous scheme in adults. METHOD Phase IIb was a parallel, multicenter, adaptive, double-blind, randomized, and placebo-controlled trial. Subjects (n = 810) aged 19-80 years were randomized to receive two doses of SARS-CoV-2 RBD conjugated to tetanus toxoid (SOBERANA 02) and a third dose of dimeric RBD (SOBERANA Plus) 28 days apart; two production batches of active ingredients of SOBERANA 02 were evaluated. Primary outcome was the percentage of seroconverted subjects with ≥4-fold the anti-RBD immunoglobulin G (IgG) concentration. Secondary outcomes were safety, reactogenicity, and neutralizing antibodies. FINDINGS Seroconversion rate in vaccinees was 76.3% after two doses and 96.8% after the third dose of SOBERANA Plus (7.3% in the placebo group). Neutralizing IgG antibodies were detected against D614G and variants of concern (VOCs) Alpha, Beta, Delta, and Omicron. Specific, functional antibodies were detected 7-8 months after the third dose. The frequency of serious adverse events (AEs) associated with vaccination was very low (0.1%). Local pain was the most frequent AE. CONCLUSIONS Two doses of SOBERANA 02 were safe and immunogenic in adults. The heterologous combination with SOBERANA Plus increased neutralizing antibodies, detectable 7-8 months after the third dose. TRIAL REGISTRY https://rpcec.sld.cu/trials/RPCEC00000347 FUNDING: This work was supported by Finlay Vaccine Institute, BioCubaFarma, and the Fondo Nacional de Ciencia y Técnica (FONCI-CITMA-Cuba, contract 2020-20).
Collapse
Affiliation(s)
| | - Mayra García-Carmenate
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | | | - Suzel Pérez-Rodríguez
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | | | - Carmen Valenzuela-Silva
- Cybernetics, Mathematics and Physics Institute, 15th St. #55, Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | - Beatriz Paredes-Moreno
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | | | - Raúl González-Mugica
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Tays Hernández-Garcia
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Ivette Orosa-Vázquez
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | | | | | | | - Enrique Noa-Romero
- National Civil Defense Research Laboratory, San José de las Lajas, Mayabeque, Cuba
| | | | - Gerardo Baro-Roman
- Centre for Immunoassays, 134 St. and 25, Cubanacán, Playa, Havana 11600 Cuba
| | - Ivis Mendoza-Hernández
- National Clinical Trials Coordinating Center, 5th Avenue and 62, Miramar, Playa, Havana, Cuba
| | - Yaima Muñoz
- National Clinical Trials Coordinating Center, 5th Avenue and 62, Miramar, Playa, Havana, Cuba
| | | | - Bertha Leysi Santos-Vega
- "19 de Abril" Polyclinic, Tulipan St. between Panorama y Oeste, Nuevo Vedado, Plaza de la Revolución, Havana 10400, Cuba
| | - Sonsire Fernandez-Castillo
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba,Corresponding author
| | - Yanet Climent-Ruiz
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Laura Rodríguez-Noda
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Darielys Santana-Mederos
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Yanelda García-Vega
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Guang-Wu Chen
- Chengdu Olisynn Biotech. Co. Ltd., Chengdu 610041, People’s Republic of China,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Delaram Doroud
- Pasteur Institute of Iran, No. 69, Pasteur Avenue, Tehran 1316943551, Islamic Republic of Iran
| | - Alireza Biglari
- Pasteur Institute of Iran, No. 69, Pasteur Avenue, Tehran 1316943551, Islamic Republic of Iran
| | - Tammy Boggiano-Ayo
- Center of Molecular Immunology, 15th Avenue and 216 St, Siboney, Playa, Havana, Cuba
| | - Yury Valdés-Balbín
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Daniel G. Rivera
- Laboratory of Synthetic and Biomolecular Chemistry, Faculty of Chemistry, University of Havana, Havana 10400, Cuba
| | - Dagmar García-Rivera
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba
| | - Vicente Vérez-Bencomo
- Finlay Vaccine Institute, 21st Avenue Nº 19810 Between 198 and 200 St, Atabey, Playa, Havana, Cuba,Corresponding author
| | | |
Collapse
|
50
|
Şan H, Alagöz E. The Effects of CoronaVac (Sinovac) and BNT162b2 (BioNTech/Pfizer) Vaccination on Oncologic 18F-FDG PET/CT Studies. Mol Imaging Radionucl Ther 2022; 31:179-190. [PMID: 36268854 PMCID: PMC9586001 DOI: 10.4274/mirt.galenos.2022.86570] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objectives: BioNTech (Pfizer) and CoronaVac (Sinovac) vaccines are two of the most administered coronavirus disease-2019 (COVID-19) vaccines worldwide. Vaccination against severe acute respiratory syndrome-coronavirus-2 has caused a diagnostic challenge in oncological 18F-fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) studies. The aim of our study was to evaluate the 18F-FDG PET/CT findings of the two most commonly administered vaccines worldwide. Methods: Patients over 18 years old who underwent 18F-FDG PET/CT for oncological purposes in our institution between January 13, 2021 and January 31, 2022, who received a single or second dose of the BioNTech or CoronaVac vaccines in the last two months, were included in the study. Descriptive analyses were presented as mean, standard deviation, frequency and ratio. Additionally, chi-square test was used to analyze categorical variables. Results: Ipsilateral deltoid muscle hypermetabolism was observed in 6.9% (n=15) and 14.3% (n=22) patients who received CoronaVac and BioNTech vaccines, respectively. Ipsilateral axillary lymph node hypermetabolism was observed in 11% (n=24) and 41.6% (n=64) patients who received CoronaVac and BioNTech vaccines, respectively. Synchronous deltoid muscle and axillary lymph node hypermetabolism was observed in 4.14% (n=9) and 12.33% (n=19) patients who received CoronaVac and BioNTech vaccines, respectively. Significant differences were detected between CoronaVac and BioNTech vaccines in terms of ipsilateral deltoid muscle hypermetabolism, ipsilateral axillary lymph node hypermetabolism and synchronous deltoid muscle and axillary lymph node hypermetabolism (p<0.05). Conclusion: COVID-19 vaccination may result in ipsilateral axillary lymph node hypermetabolism, ipsilateral deltoid muscle hypermetabolism, or synchronous deltoid muscle and axillary lymph node hypermetabolism with different frequencies depending on the type of vaccination. Although synchronous deltoid muscle and axillary lymph node hypermetabolism can reduce misinterpretation of 18F-FDG PET/CT, to avoid misinterpretation, it is important to question the vaccination history during ongoing COVID-19 vaccination process.
Collapse
Affiliation(s)
- Hüseyin Şan
- University of Health Sciences Turkey, Ankara City Hospital, Department of Nuclear Medicine, Ankara, Turkey
| | - Engin Alagöz
- University of Health Sciences Turkey, Gülhane Training and Research Hospital, Department of Nuclear Medicine, Ankara, Turkey
| |
Collapse
|