201
|
Fang F, Clemens JD, Zhang ZF, Brewer TF. Impact of SARS-CoV-2 vaccines on Covid-19 incidence and mortality in the United States. PLoS One 2024; 19:e0301830. [PMID: 38656933 PMCID: PMC11042718 DOI: 10.1371/journal.pone.0301830] [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: 08/08/2022] [Accepted: 03/19/2024] [Indexed: 04/26/2024] Open
Abstract
BACKGROUND Given the waning of vaccine effectiveness and the shifting of the most dominant strains in the U.S., it is imperative to understand the association between vaccination coverage and Severe Acute Respiratory Syndrome coronavirus 2 (SARS-CoV-2) disease and mortality at the community levels and whether that association might vary according to the dominant SARS-CoV-2 strains in the U.S. METHODS Generalized estimating equations were used to estimate associations between U.S. county-level cumulative vaccination rates and booster distribution and the daily change in county-wide Coronavirus 2019 disease (COVID-19) risks and mortality during Alpha, Delta and Omicron predominance. Models were adjusted for potential confounders at both county and state level. A 2-week lag and a 4-week lag were introduced to assess vaccination rate impact on incidence and mortality, respectively. RESULTS Among 3,073 counties in 48 states, the average county population complete vaccination rate of all age groups was 50.79% as of March 11th, 2022. Each percentage increase in vaccination rates was associated with reduction of 4% (relative risk (RR) 0.9607 (95% confidence interval (CI): 0.9553, 0.9661)) and 3% (RR 0.9694 (95% CI: 0.9653, 0.9736)) in county-wide COVID-19 cases and mortality, respectively, when Alpha was the dominant variant. The associations between county-level vaccine rates and COVID-19 incidence diminished during the Delta and Omicron predominance. However, each percent increase in people receiving a booster shot was associated with reduction of 6% (RR 0.9356 (95% CI: 0.9235, 0.9479)) and 4% (RR 0.9595 (95% CI: 0.9431, 0.9761)) in COVID-19 incidence and mortality in the community, respectively, during the Omicron predominance. CONCLUSIONS Associations between complete vaccination rates and COVID-19 incidence and mortality appeared to vary with shifts in the dominant variant, perhaps due to variations in vaccine efficacy by variant or to waning vaccine immunity over time. Vaccine boosters were associated with notable protection against Omicron disease and mortality.
Collapse
Affiliation(s)
- Fang Fang
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - John David Clemens
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, United States of America
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
- International Vaccination Institute (IVI), Seoul, the Republic of Korea
| | - Zuo-Feng Zhang
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, United States of America
- Department of Medicine, Center for Human Nutrition, UCLA David Geffen School of Medicine, University of California at Los Angeles (UCLA), Los Angeles, CA, United States of America
| | - Timothy F. Brewer
- Department of Epidemiology, Fielding School of Public Health, University of California at Los Angeles (UCLA), Los Angeles, CA, United States of America
- Division of Infectious Diseases, UCLA David Geffen School of Medicine, Los Angeles, CA, United States of America
| |
Collapse
|
202
|
Bianco A, Di Sante G, Colò F, De Arcangelis V, Cicia A, Del Giacomo P, De Bonis M, Morganti TG, Carlomagno V, Lucchini M, Minucci A, Calabresi P, Mirabella M. Multiple Sclerosis Onset before and after COVID-19 Vaccination: Can HLA Haplotype Be Determinant? Int J Mol Sci 2024; 25:4556. [PMID: 38674141 PMCID: PMC11050425 DOI: 10.3390/ijms25084556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
A few cases of multiple sclerosis (MS) onset after COVID-19 vaccination have been reported, although the evidence is insufficient to establish causality. The aim of this study is to compare cases of newly diagnosed relapsing-remitting MS before and after the outbreak of the COVID-19 pandemic and the impact of COVID-19 vaccination. Potential environmental and genetic predisposing factors were also investigated, as well as clinical patterns. This is a single-centre retrospective cohort study including all patients who presented with relapsing-remitting MS onset between January 2018 and July 2022. Data on COVID-19 vaccination administration, dose, and type were collected. HLA-DRB1 genotyping was performed in three subgroups. A total of 266 patients received a new diagnosis of relapsing-remitting MS in our centre, 143 before the COVID-19 pandemic (until and including March 2020), and 123 during the COVID-19 era (from April 2020). The mean number of new MS onset cases per year was not different before and during the COVID-19 era and neither were baseline patients' characteristics, type of onset, clinical recovery, or radiological patterns. Fourteen (11.4%) patients who subsequently received a new diagnosis of MS had a history of COVID-19 vaccination within one month before symptoms onset. Patients' characteristics, type of onset, clinical recovery, and radiological patterns did not differ from those of patients with non-vaccine-related new diagnoses of MS. The allele frequencies of HLA-DRB1*15 were 17.6% and 22.2% in patients with non-vaccine-related disease onset before and during the COVID-19 era, respectively, while no case of HLA-DRB1*15 was identified among patients with a new diagnosis of MS post-COVID-19 vaccine. In contrast, HLA-DRB1*08+ or HLA-DRB1*10+ MS patients were present only in this subgroup. Although a causal link between COVID-19 vaccination and relapsing-remitting MS cannot be detected, it is interesting to note and speculate about the peculiarities and heterogeneities underlying disease mechanisms of MS, where the interactions of genetics and the environment could be crucial also for the follow-up and the evaluation of therapeutic options.
Collapse
Affiliation(s)
- Assunta Bianco
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Gabriele Di Sante
- Department of Medicine and Surgery, Section of Human, Clinical and Forensic Anatomy, University of Perugia, 06123 Perugia, Italy
| | - Francesca Colò
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Valeria De Arcangelis
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Alessandra Cicia
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Paola Del Giacomo
- Department of Laboratory and Infectious Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Maria De Bonis
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Tommaso Giuseppe Morganti
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Vincenzo Carlomagno
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Matteo Lucchini
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Angelo Minucci
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Genomics Core Facility, Gemelli Science and Technology Park (G-STeP), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Paolo Calabresi
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Massimiliano Mirabella
- Division of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Department of Neurosciences, Centro di Ricerca per la Sclerosi Multipla “Anna Paola Batocchi”, Catholic University of Sacred Heart, 00168 Rome, Italy
| |
Collapse
|
203
|
Files JK, Goepfert PA. Messenger RNA Vaccine Technology: Success for SARS-CoV-2 and Prospects for an HIV-1 Vaccine. TOPICS IN ANTIVIRAL MEDICINE 2024; 32:420-430. [PMID: 39141920 PMCID: PMC11293605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
Over the past several years, messenger RNA (mRNA) vaccine has evolved from a term familiar only to vaccine scientists into one easily recognized by much of the general population. This change occurred because of the remarkable success of effective and safe mRNA vaccines during the COVID-19 pandemic that saved countless lives. Although mRNA vaccine technology has a clear use for combating future emerging diseases, its role in fighting currently known pathogens, such as HIV-1, is not well defined. This review summarizes mRNA vaccine technology, highlighting its success during the COVID-19 pandemic. It then addresses past and current efforts to develop a vaccine for HIV-1, including how mRNA vaccine technology has created opportunities in the ongoing search for an effective HIV-1 vaccine.
Collapse
|
204
|
Iketani S, Ho DD. SARS-CoV-2 resistance to monoclonal antibodies and small-molecule drugs. Cell Chem Biol 2024; 31:632-657. [PMID: 38640902 PMCID: PMC11084874 DOI: 10.1016/j.chembiol.2024.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/21/2024]
Abstract
Over four years have passed since the beginning of the COVID-19 pandemic. The scientific response has been rapid and effective, with many therapeutic monoclonal antibodies and small molecules developed for clinical use. However, given the ability for viruses to become resistant to antivirals, it is perhaps no surprise that the field has identified resistance to nearly all of these compounds. Here, we provide a comprehensive review of the resistance profile for each of these therapeutics. We hope that this resource provides an atlas for mutations to be aware of for each agent, particularly as a springboard for considerations for the next generation of antivirals. Finally, we discuss the outlook and thoughts for moving forward in how we continue to manage this, and the next, pandemic.
Collapse
Affiliation(s)
- Sho Iketani
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - David D Ho
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA; Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA.
| |
Collapse
|
205
|
Warner BM, Chan M, Tailor N, Vendramelli R, Audet J, Meilleur C, Truong T, Garnett L, Willman M, Soule G, Tierney K, Albietz A, Moffat E, Higgins R, Santry LA, Leacy A, Pham PH, Yates JGE, Pei Y, Safronetz D, Strong JE, Susta L, Embury-Hyatt C, Wootton SK, Kobasa D. Mucosal Vaccination with a Newcastle Disease Virus-Vectored Vaccine Reduces Viral Loads in SARS-CoV-2-Infected Cynomolgus Macaques. Vaccines (Basel) 2024; 12:404. [PMID: 38675786 PMCID: PMC11054841 DOI: 10.3390/vaccines12040404] [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: 03/01/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/28/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged following an outbreak of unexplained viral illness in China in late 2019. Since then, it has spread globally causing a pandemic that has resulted in millions of deaths and has had enormous economic and social consequences. The emergence of SARS-CoV-2 saw the rapid and widespread development of a number of vaccine candidates worldwide, and this never-before-seen pace of vaccine development led to several candidates progressing immediately through clinical trials. Many countries have now approved vaccines for emergency use, with large-scale vaccination programs ongoing. Despite these successes, there remains a need for ongoing pre-clinical and clinical development of vaccine candidates against SARS-CoV-2, as well as vaccines that can elicit strong mucosal immune responses. Here, we report on the efficacy of a Newcastle disease virus-vectored vaccine candidate expressing SARS-CoV-2 spike protein (NDV-FLS) administered to cynomolgus macaques. Macaques given two doses of the vaccine via respiratory immunization developed robust immune responses and had reduced viral RNA levels in nasal swabs and in the lower airway. Our data indicate that NDV-FLS administered mucosally provides significant protection against SARS-CoV-2 infection, resulting in reduced viral burden and disease manifestation, and should be considered as a viable candidate for clinical development.
Collapse
Affiliation(s)
- Bryce M. Warner
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Mable Chan
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Nikesh Tailor
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Robert Vendramelli
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Jonathan Audet
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Courtney Meilleur
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Thang Truong
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Lauren Garnett
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Marnie Willman
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Geoff Soule
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Kevin Tierney
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Alixandra Albietz
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
| | - Estella Moffat
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3R2, Canada; (E.M.); (C.E.-H.)
| | - Rick Higgins
- Department of Radiology, Health Sciences Center, Winnipeg, MB R3A 1S1, Canada;
| | - Lisa A. Santry
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Alexander Leacy
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Phuc H. Pham
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Jacob G. E. Yates
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Yanlong Pei
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - David Safronetz
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - James E. Strong
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Leonardo Susta
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Carissa Embury-Hyatt
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB R3E 3R2, Canada; (E.M.); (C.E.-H.)
| | - Sarah K. Wootton
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.A.S.); (A.L.); (P.H.P.); (J.G.E.Y.); (Y.P.); (L.S.)
| | - Darwyn Kobasa
- Special Pathogens, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB R3E 3R2, Canada; (M.C.); (N.T.); (R.V.); (J.A.); (C.M.); (T.T.); (L.G.); (M.W.); (G.S.); (K.T.); (A.A.); (D.S.); (J.E.S.); (D.K.)
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
206
|
Audley G, Raubenheimer P, Symons G, Mendelson M, Meintjes G, Ntusi NAB, Wasserman S, Dlamini S, Dheda K, van Zyl-Smit R, Calligaro G. High-flow nasal oxygen in resource-constrained, non-intensive, high-care wards for COVID-19 acute hypoxaemic respiratory failure: Comparing outcomes of the first v. third waves at a tertiary centre in South Africa. Afr J Thorac Crit Care Med 2024; 30:e1151. [PMID: 38756391 PMCID: PMC11094705 DOI: 10.7196/ajtccm.2024.v30i1.1151] [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: 06/05/2023] [Accepted: 01/08/2024] [Indexed: 05/18/2024] Open
Abstract
Background High-flow nasal oxygen (HFNO) is an accepted treatment for severe COVID-19-related acute hypoxaemic respiratory failure (AHRF). Objectives To determine whether treatment outcomes at Groote Schuur Hospital, Cape Town, South Africa, during the third COVID-19 wave would be affected by increased institutional experience and capacity for HNFO and more restrictive admission criteria for respiratory high-care wards and intensive care units. Methods We included consecutive patients with COVID-19-related AHRF treated with HFNO during the first and third COVID-19 waves. The primary endpoint was comparison of HFNO failure (composite of the need for intubation or death while on HFNO) between waves. Results A total of 744 patients were included: 343 in the first COVID-19 wave and 401 in the third. Patients treated with HFNO in the first wave were older (median (interquartile range) age 53 (46 - 61) years v. 47 (40 - 56) years; p<0.001), and had higher prevalences of diabetes (46.9% v. 36.9%; p=0.006), hypertension (51.0% v. 35.2%; p<0.001), obesity (33.5% v. 26.2%; p=0.029) and HIV infection (12.5% v. 5.5%; p<0.001). The partial pressure of arterial oxygen to fraction of inspired oxygen (PaO2 /FiO2 ) ratio at HFNO initiation and the ratio of oxygen saturation/FiO2 to respiratory rate within 6 hours (ROX-6 score) after HFNO commencement were lower in the first wave compared with the third (median 57.9 (47.3 - 74.3) mmHg v. 64.3 (51.2 - 79.0) mmHg; p=0.005 and 3.19 (2.37 - 3.77) v. 3.43 (2.93 - 4.00); p<0.001, respectively). The likelihood of HFNO failure (57.1% v. 59.6%; p=0.498) and mortality (46.9% v. 52.1%; p=0.159) did not differ significantly between the first and third waves. Conclusion Despite differences in patient characteristics, circulating viral variant and institutional experience with HFNO, treatment outcomes were very similar in the first and third COVID-19 waves. We conclude that once AHRF is established in COVID-19 pneumonia, the comorbidity profile and HFNO provider experience do not appear to affect outcome. Study synopsis What the study adds. This study adds to the body of evidence demonstrating the utility of high-flow nasal oxygen (HFNO) in avoiding invasive mechanical ventilation (IMV) in patients with severe COVID-19 hypoxaemic respiratory failure, and shows that this utility remained consistent across different waves of the COVID-19 pandemic.Implications of the study. In resource-constrained settings, HFNO is a feasible non-invasive alternative to IMV and can be employed with favourable and consistent outcomes outside traditional critical care wards. It also confirms that the degree of gas exchange abnormality, and not pre-existing patient-related factors, circulating wave variant or provider experience, is the main predictor of HFNO failure.
Collapse
Affiliation(s)
- G Audley
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
| | - P Raubenheimer
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
| | - G Symons
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
- Division of Pulmonology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - M Mendelson
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote
Schuur Hospital, Cape Town, South Africa
| | - G Meintjes
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town,
South Africa
| | - N A B Ntusi
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
- Division of Cardiology, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, Cape Town,
South Africa
- South African Medical Research Council/University of Cape Town Extramural Research Unit on the Intersection of Noncommunicable Diseases
and Infectious Diseases, University of Cape Town, South Africa
| | - S Wasserman
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote
Schuur Hospital, Cape Town, South Africa
| | - S Dlamini
- Division of General Internal Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur
Hospital, Cape Town, South Africa
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote
Schuur Hospital, Cape Town, South Africa
| | - K Dheda
- Division of Pulmonology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
- South African Medical Research Council/University of Cape Town Extramural Research Unit on the Intersection of Noncommunicable Diseases
and Infectious Diseases, University of Cape Town, South Africa
- Faculty of Infectious and Tropical Diseases, Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute, University of Cape Town,
South Africa; South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, South Africa
| | - R van Zyl-Smit
- Division of Pulmonology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - G Calligaro
- Division of Pulmonology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine and UCT Lung Institute, University of Cape Town,
South Africa; South African MRC/UCT Centre for the Study of Antimicrobial Resistance, University of Cape Town, South Africa
| |
Collapse
|
207
|
Campos GRF, Almeida NBF, Filgueiras PS, Corsini CA, Gomes SVC, de Miranda DAP, de Assis JV, Silva TBDS, Alves PA, Fernandes GDR, de Oliveira JG, Rahal P, Grenfell RFQ, Nogueira ML. Second booster dose improves antibody neutralization against BA.1, BA.5 and BQ.1.1 in individuals previously immunized with CoronaVac plus BNT162B2 booster protocol. Front Cell Infect Microbiol 2024; 14:1371695. [PMID: 38638823 PMCID: PMC11024236 DOI: 10.3389/fcimb.2024.1371695] [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: 01/16/2024] [Accepted: 03/14/2024] [Indexed: 04/20/2024] Open
Abstract
Introduction SARS-CoV-2 vaccines production and distribution enabled the return to normalcy worldwide, but it was not fast enough to avoid the emergence of variants capable of evading immune response induced by prior infections and vaccination. This study evaluated, against Omicron sublineages BA.1, BA.5 and BQ.1.1, the antibody response of a cohort vaccinated with a two doses CoronaVac protocol and followed by two heterologous booster doses. Methods To assess vaccination effectiveness, serum samples were collected from 160 individuals, in 3 different time points (9, 12 and 18 months after CoronaVac protocol). For each time point, individuals were divided into 3 subgroups, based on the number of additional doses received (No booster, 1 booster and 2 boosters), and a viral microneutralization assay was performed to evaluate neutralization titers and seroconvertion rate. Results The findings presented here show that, despite the first booster, at 9m time point, improved neutralization level against omicron ancestor BA.1 (133.1 to 663.3), this trend was significantly lower for BQ.1.1 and BA.5 (132.4 to 199.1, 63.2 to 100.2, respectively). However, at 18m time point, the administration of a second booster dose considerably improved the antibody neutralization, and this was observed not only against BA.1 (2361.5), but also against subvariants BQ.1.1 (726.1) and BA.5 (659.1). Additionally, our data showed that, after first booster, seroconvertion rate for BA.5 decayed over time (93.3% at 12m to 68.4% at 18m), but after the second booster, seroconvertion was completely recovered (95% at 18m). Discussion Our study reinforces the concerns about immunity evasion of the SARS-CoV-2 omicron subvariants, where BA.5 and BQ.1.1 were less neutralized by vaccine induced antibodies than BA.1. On the other hand, the administration of a second booster significantly enhanced antibody neutralization capacity against these subvariants. It is likely that, as new SARS-CoV-2 subvariants continue to emerge, additional immunizations will be needed over time.
Collapse
Affiliation(s)
- Guilherme R. F. Campos
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
| | | | - Priscilla Soares Filgueiras
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | - Camila Amormino Corsini
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | - Sarah Vieira Contin Gomes
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | - Daniel Alvim Pena de Miranda
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | - Jéssica Vieira de Assis
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | - Thaís Bárbara de Souza Silva
- Laboratório de Imunologia de Doenças Virais, Instituto Rene Rachou - Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Pedro Augusto Alves
- Laboratório de Imunologia de Doenças Virais, Instituto Rene Rachou - Fundação Oswaldo Cruz, Belo Horizonte, Brazil
| | - Gabriel da Rocha Fernandes
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
| | | | - Paula Rahal
- Laboratório de Estudos Genômicos, Departamento de Biologia, Instituto de Biociências Letras e Ciências Exatas (IBILCE), Universidade Estadual Paulista (Unesp), São José do Rio Preto, Brazil
| | - Rafaella Fortini Queiroz Grenfell
- Diagnosis and Therapy of Infectious Diseases and Cancer, Oswaldo Cruz Foundation (Fiocruz), Belo Horizonte, Brazil
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Maurício L. Nogueira
- Laboratório de Pesquisas em Virologia (LPV), Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, Brazil
- Hospital de Base, São José do Rio Preto, Brazil
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
208
|
Ezzikouri S, Tajudeen R, Majidi H, Redwane S, Aqillouch S, Abdulaziz M, Aragaw M, Papa Fallah M, Sembuche S, Batcho S, Kabwe P, Gonese E, Laazaazia O, Elmessaoudi-Idrissi M, Meziane N, Ainahi A, Sarih M, Ogwell Ouma AE, Maaroufi A. Seroepidemiological assessment of SARS-CoV-2 vaccine responsiveness and associated factors in the vaccinated community of the Casablanca-Settat Region, Morocco. Sci Rep 2024; 14:7817. [PMID: 38570577 PMCID: PMC10991243 DOI: 10.1038/s41598-024-58498-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 03/29/2024] [Indexed: 04/05/2024] Open
Abstract
Assessing the prevalence of SARS-CoV-2 IgG positivity through population-based serological surveys is crucial for monitoring COVID-19 vaccination efforts. In this study, we evaluated SARS-CoV-2 IgG positivity within a provincial cohort to understand the magnitude of the humoral response against the SARS-CoV-2 vaccine and to inform evidence-based public health decisions. A community-based cross-sectional seroprevalence study was conducted, involving 10,669 participants who received various vaccines (two doses for BBIBP-CorV/Sinopharm, Covishield vaccine, and Pfizer/BioNTech, and one dose for Johnson & Johnson's Janssen COVID-19 vaccine). The study spanned 16 provinces in the Casablanca-Settat region from February to June 2022, during which comprehensive demographic and comorbidity data were collected. We screened samples for the presence of IgG antibodies using the SARS-CoV-2 IgG II Quant assay, which quantifies antibodies against the receptor-binding domain (RBD) of the spike (S) protein, measured on the Abbott Architect i2000SR. The overall crude seroprevalence was 96% (95% CI: 95.6-96.3%), and after adjustment for assay performance, it was estimated as 96.2% (95% CI: 95.7-96.6). The adjusted overall seroprevalences according to vaccine brands showed no significant difference (96% for BBIBP-CorV/Sinopharm, 97% for ChAdOx1 nCoV-19/Oxford/AstraZeneca, 98.5% for BNT162b2/Pfizer-BioNTech, and 98% for Janssen) (p = 0.099). Participants of older age, female sex, those with a history of previous COVID-19 infection, and those with certain chronic diseases were more likely to be seropositive among ChAdOx1 nCoV-19/Oxford/AstraZeneca and BBIBP-CorV/Sinopharm vaccinee groups. Median RBD antibody concentrations were 2355 AU/mL, 3714 AU/mL, 5838 AU/mL, and 2495 AU/mL, respectively, after two doses of BBIBP-CorV/Sinopharm, ChAdOx1 nCoV-19/Oxford/AstraZeneca, BNT162b2/Pfizer-BioNTech, and after one dose of Janssen (p < 0.0001). Furthermore, we observed that participants vaccinated with ChAdOx1 nCoV-19/Oxford/AstraZeneca and BBIBP-CorV/Sinopharm with comorbid chronic diseases exhibited a more pronounced response to vaccination compared to those without comorbidities. In contrast, no significant differences were observed among Pfizer-vaccinated participants (p > 0.05). In conclusion, our serosurvey findings indicate that all four investigated vaccines provide a robust humoral immune response in the majority of participants (more than 96% of participants had antibodies against SARS-CoV-2). The BNT162b2 vaccine was found to be effective in eliciting a strong humoral response compared to the other three vaccines. However, challenges still remain in examining the dynamics and durability of immunoprotection in the Moroccan context.
Collapse
Affiliation(s)
- Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco.
| | - Raji Tajudeen
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Hind Majidi
- Ministry of Health and Social Protection, Rabat, Morocco
| | - Soad Redwane
- Direction Régionale de la santé Casablanca-Settat, Observatoire régional de santé, Casablanca, Morocco
| | - Safaa Aqillouch
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco
| | - Mohammed Abdulaziz
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Merawi Aragaw
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Mosoka Papa Fallah
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Senga Sembuche
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Serge Batcho
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Patrick Kabwe
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Elizabeth Gonese
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Oumaima Laazaazia
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco
| | - Mohcine Elmessaoudi-Idrissi
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco
| | - Nadia Meziane
- Centre Régional de Transfusion Sanguine, Casablanca, Morocco
| | - Abdelhakim Ainahi
- Hormonology and Tumor Markers Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - M'hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Ahmed E Ogwell Ouma
- Africa Centres for Disease Control and Prevention, African Union, Addis Ababa, Ethiopia
| | - Abderrahmane Maaroufi
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360, Casablanca, Morocco
| |
Collapse
|
209
|
Galhaut M, Lundberg U, Marlin R, Schlegl R, Seidel S, Bartuschka U, Heindl-Wruss J, Relouzat F, Langlois S, Dereuddre-Bosquet N, Morin J, Galpin-Lebreau M, Gallouët AS, Gros W, Naninck T, Pascal Q, Chapon C, Mouchain K, Fichet G, Lemaitre J, Cavarelli M, Contreras V, Legrand N, Meinke A, Le Grand R. Immunogenicity and efficacy of VLA2001 vaccine against SARS-CoV-2 infection in male cynomolgus macaques. COMMUNICATIONS MEDICINE 2024; 4:62. [PMID: 38570605 PMCID: PMC10991505 DOI: 10.1038/s43856-024-00488-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND The fight against COVID-19 requires mass vaccination strategies, and vaccines inducing durable cross-protective responses are still needed. Inactivated vaccines have proven lasting efficacy against many pathogens and good safety records. They contain multiple protein antigens that may improve response breadth and can be easily adapted every year to maintain preparedness for future seasonally emerging variants. METHODS The vaccine dose was determined using ELISA and pseudoviral particle-based neutralization assay in the mice. The immunogenicity was assessed in the non-human primates with multiplex ELISA, neutralization assays, ELISpot and intracellular staining. The efficacy was demonstrated by viral quantification in fluids using RT-qPCR and respiratory tissue lesions evaluation. RESULTS Here we report the immunogenicity and efficacy of VLA2001 in animal models. VLA2001 formulated with alum and the TLR9 agonist CpG 1018™ adjuvant generate a Th1-biased immune response and serum neutralizing antibodies in female BALB/c mice. In male cynomolgus macaques, two injections of VLA2001 are sufficient to induce specific and polyfunctional CD4+ T cell responses, predominantly Th1-biased, and high levels of antibodies neutralizing SARS-CoV-2 infection in cell culture. These antibodies also inhibit the binding of the Spike protein to human ACE2 receptor of several variants of concern most resistant to neutralization. After exposure to a high dose of homologous SARS-CoV-2, vaccinated groups exhibit significant levels of protection from viral replication in the upper and lower respiratory tracts and from lung tissue inflammation. CONCLUSIONS We demonstrate that the VLA2001 adjuvanted vaccine is immunogenic both in mouse and NHP models and prevent cynomolgus macaques from the viruses responsible of COVID-19.
Collapse
Affiliation(s)
- Mathilde Galhaut
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | | | - Romain Marlin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | | | | | | | | | - Francis Relouzat
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Sébastien Langlois
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Nathalie Dereuddre-Bosquet
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Julie Morin
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Maxence Galpin-Lebreau
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Anne-Sophie Gallouët
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Wesley Gros
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Thibaut Naninck
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Quentin Pascal
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Catherine Chapon
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Karine Mouchain
- ONCODESIGN SERVICES, François Hyafil Research Center, Villebon-sur-Yvette, France
| | - Guillaume Fichet
- ONCODESIGN SERVICES, François Hyafil Research Center, Villebon-sur-Yvette, France
| | - Julien Lemaitre
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Mariangela Cavarelli
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Vanessa Contreras
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France
| | - Nicolas Legrand
- ONCODESIGN SERVICES, François Hyafil Research Center, Villebon-sur-Yvette, France
| | | | - Roger Le Grand
- Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Université Paris-Saclay, Inserm, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
210
|
Garcia‐Moure M, Gillard AG, Alonso MM, Fueyo J, Gomez‐Manzano C. Oncolytic adenoviruses and immunopeptidomics: a convenient marriage. Mol Oncol 2024; 18:781-784. [PMID: 38561242 PMCID: PMC10994240 DOI: 10.1002/1878-0261.13648] [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: 03/19/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Oncolytic viruses (OVs) are biological therapeutic agents that selectively destroy cancer cells while sparing normal healthy cells. Besides direct oncolysis, OV infection induces a proinflammatory shift in the tumor microenvironment and the release of tumor-associated antigens (TAAs) that might induce an anti-tumor immunity. Due to their immunostimulatory effect, OVs have been explored for cancer vaccination against specific TAAs. However, this approach usually requires genetic modification of the virus and the production of a new viral vector for each target, which is difficult to implement for low prevalent antigens. In a recent study, Chiaro et al. presented an elegant proof of concept on how to implement the PeptiCRAd vaccination platform to overcome this limitation for the treatment of mesothelioma. Authors showed the feasibility of identifying immunogenic TAAs in human mesothelioma and using them to coat oncolytic adenovirus particles. The result was a customized virus-based cancer vaccine that circumvents time and resource-consuming steps incurred from genetically engineering viruses. Although some questions remain to be addressed, this interesting approach suggests novel strategies for personalized cancer medicine using oncolytic virotherapy.
Collapse
Affiliation(s)
- Marc Garcia‐Moure
- Department of Neuro‐OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Andrew G. Gillard
- Department of Neuro‐OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Marta M. Alonso
- Department of PediatricsClinica Universidad de NavarraPamplonaSpain
- Program of Solid TumorsFoundation for the Applied Medical ResearchPamplonaSpain
| | - Juan Fueyo
- Department of Neuro‐OncologyThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | | |
Collapse
|
211
|
Hoyois A, Gulkilik C, Mekkaoui L, Dahma H, Wambacq V, Minsart C, Rosewick N, Liefferinckx C, Amininejad L, Van Gossum A, Cremer A, Vandenberg O, Franchimont D. SARS-CoV-2 antibody vaccine response in Inflammatory Bowel Disease patients with positive anti-nucleocapsid serology or history of COVID-19 infection. Acta Gastroenterol Belg 2024; 87:263-273. [PMID: 39210758 DOI: 10.51821/87.2.12805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Previous history of COVID-19 infection is a natural booster of the vaccine response in the general population. The response to COVID-19 vaccines is lessened in Inflammatory Bowel Disease patients on selected class of immunosuppressive treatments. Aims The study was to assess anti-SARS-CoV-2 spike-specific IgG antibody response in Inflammatory Bowel Disease patients with a history of COVID-19 infection. Patients and methods This single-center prospective study involved 504 Inflammatory Bowel Disease patients. Demographic data and clinical data were gathered through questionnaires and patient charts. Anti-SARS-CoV-2 spike-specific and antinucleocapsid antibody levels were measured at T1, T2 (after the 2-dose series), and T3 or T4 (booster vaccine). Results This study included 504 Inflammatory Bowel Disease patients, and 234 completed one year follow-up with blood tests. Positive anti-nucleocapsid serology or history of COVID-19 infection was significantly associated with increased median anti- SARS-CoV-2 spike-specific IgG titers after the 2-dose series (1930 BAU/mL vs. 521 BAU/mL p < 0.0001) and the booster vaccine (4390 BAU/mL vs. 2160 BAU/mL, p = 0.0156). Multivariate analysis showed that higher anti-SARS-CoV-2 spike-specific IgG levels were independently associated with anti-nucleocapsid antibodies at T2 (OR=2.23, p < 0.0001) and T3 (OR=1.72, p = 0.00011). Immunosuppressive treatments did not impact the antibody response or levels in patients with a history of COVID-19 infection or positive anti-nucleocapsid serology. Conclusions In Inflammatory Bowel Disease, prior COVID-19 infection or positive anti-nucleocapsid serology leads to increased anti-SARS-CoV-2 spike-specific IgG levels after vaccination, regardless of immunosuppressive treatments. This emphasizes the significance of accounting for previous infection in vaccination approaches.
Collapse
Affiliation(s)
- A Hoyois
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
- Department of Hepato-Gastroenterology, CHU Saint-Pierre, Université Libre de Bruxelles, Brussels, Belgium
| | - C Gulkilik
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - L Mekkaoui
- Department of Microbiology, Laboratoire Hospitalier Universitaire de Bruxelles, Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles, Brussels, Belgium
| | - H Dahma
- Department of Microbiology, Laboratoire Hospitalier Universitaire de Bruxelles, Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles, Brussels, Belgium
| | - V Wambacq
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - C Minsart
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - N Rosewick
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - C Liefferinckx
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - L Amininejad
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - A Van Gossum
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - A Cremer
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| | - O Vandenberg
- Innovation and Business Development Unit, Laboratoire Hospitalier Universitaire de Bruxelles-Universitair Laboratorium Brussel (LHUB-ULB), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - D Franchimont
- Department of Gastroenterology, Hepatopancreatology, and Digestive Oncology, HUB Hôpital Erasme, Université Libre de Bruxelles, Brussels Belgium
| |
Collapse
|
212
|
Yasmin S, Ansari MY, Pandey K, Dikhit MR. Identification of potential vaccine targets for elicitation of host immune cells against SARS-CoV-2 by reverse vaccinology approach. Int J Biol Macromol 2024; 265:130754. [PMID: 38508555 DOI: 10.1016/j.ijbiomac.2024.130754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024]
Abstract
The COVID-19 pandemic has emerged as a critical global health crisis, demanding urgent and effective strategies for containment. While some knowledge exists about epitope sequences recognized by human immune cells and their activation of CD8+ T cells within the HLA context, comprehensive information remains limited. This study employs reverse vaccinology to explore antigenic HLA-restricted T-cell epitopes capable of eliciting durable immunity. Screening reveals 187 consensus epitopes, with 23 offering broad population coverage worldwide, spanning over 5000 HLA alleles. Sequence alignment analysis highlights the genetic distinctiveness of these peptides from Homo sapiens and their intermediate to high TAP binding efficiency. Notably, these epitopes share 100 % sequence identity across strains from nine countries, indicating potential for a uniform protective immune response among diverse ethnic populations. Docking simulations further confirm their binding capacity with the HLA allele, validating them as promising targets for SARS-CoV-2 immune recognition. The anticipated epitopes are connected with suitable linkers and adjuvant, and then assessed for its translational efficacy within a bacterial expression vector through computational cloning. Through docking, it is observed that the chimeric vaccine construct forms lasting hydrogen bonds with Toll-like receptor (TLR4), while immune simulation illustrates an increased cytotoxic response aimed at CD8+ T cells. This comprehensive computational analysis suggests the chimeric vaccine construct's potential to provoke a robust immune response against SARS-CoV-2. By delineating these antigenic fragments, our study offers valuable insights into effective vaccine and immunotherapy development against COVID-19, contributing significantly to global efforts in combating this infectious threat.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University (KKU), Abha 62529, Saudi Arabia
| | - Mohammad Yousuf Ansari
- Department of Pharmaceutical Chemistry, M.M. College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, India.
| | - Krishna Pandey
- Department of Clinical Medicine, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India
| | - Manas Ranjan Dikhit
- Department of Bioinformatics, ICMR-Rajendra Memorial Research Institute of Medical Sciences, Agamkuan, Patna 800007, India.
| |
Collapse
|
213
|
Qiu Y, Mo C, Chen L, Ye W, Chen G, Zhu T. Alterations in microbiota of patients with COVID-19: implications for therapeutic interventions. MedComm (Beijing) 2024; 5:e513. [PMID: 38495122 PMCID: PMC10943180 DOI: 10.1002/mco2.513] [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: 08/28/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/19/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) recently caused a global pandemic, resulting in more than 702 million people being infected and over 6.9 million deaths. Patients with coronavirus disease (COVID-19) may suffer from diarrhea, sleep disorders, depression, and even cognitive impairment, which is associated with long COVID during recovery. However, there remains no consensus on effective treatment methods. Studies have found that patients with COVID-19 have alterations in microbiota and their metabolites, particularly in the gut, which may be involved in the regulation of immune responses. Consumption of probiotics may alleviate the discomfort caused by inflammation and oxidative stress. However, the pathophysiological process underlying the alleviation of COVID-19-related symptoms and complications by targeting the microbiota remains unclear. In the current study, we summarize the latest research and evidence on the COVID-19 pandemic, together with symptoms of SARS-CoV-2 and vaccine use, with a focus on the relationship between microbiota alterations and COVID-19-related symptoms and vaccine use. This work provides evidence that probiotic-based interventions may improve COVID-19 symptoms by regulating gut microbiota and systemic immunity. Probiotics may also be used as adjuvants to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yong Qiu
- Department of AnesthesiologyNational Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012)West China HospitalSichuan UniversityChengduChina
- Laboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Center of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduChina
| | - Chunheng Mo
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOEState Key Laboratory of BiotherapyWest China Second University HospitalSichuan UniversityChengduChina
| | - Lu Chen
- Department of AnesthesiologyNational Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012)West China HospitalSichuan UniversityChengduChina
- Laboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Center of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduChina
| | - Wanlin Ye
- Department of AnesthesiologyNational Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012)West China HospitalSichuan UniversityChengduChina
- Laboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Center of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduChina
| | - Guo Chen
- Department of AnesthesiologyNational Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012)West China HospitalSichuan UniversityChengduChina
- Laboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Center of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduChina
| | - Tao Zhu
- Department of AnesthesiologyNational Clinical Research Center for Geriatrics and The Research Units of West China (2018RU012)West China HospitalSichuan UniversityChengduChina
- Laboratory of Anesthesia and Critical Care MedicineNational‐Local Joint Engineering Research Center of Translational Medicine of AnesthesiologyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
214
|
Zhu C, Pang S, Liu J, Duan Q. Current Progress, Challenges and Prospects in the Development of COVID-19 Vaccines. Drugs 2024; 84:403-423. [PMID: 38652356 DOI: 10.1007/s40265-024-02013-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 04/25/2024]
Abstract
The COVID-19 pandemic has resulted in over 772 million confirmed cases, including nearly 7 million deaths, according to the World Health Organization (WHO). Leveraging rapid development, accelerated vaccine approval processes, and large-scale production of various COVID-19 vaccines using different technical platforms, the WHO declared an end to the global health emergency of COVID-19 on May 5, 2023. Current COVID-19 vaccines encompass inactivated, live attenuated, viral vector, protein subunit, nucleic acid (DNA and RNA), and virus-like particle (VLP) vaccines. However, the efficacy of these vaccines is diminishing due to the constant mutation of SARS-CoV-2 and the heightened immune evasion abilities of emerging variants. This review examines the impact of the COVID-19 pandemic, the biological characteristics of the virus, and its diverse variants. Moreover, the review underscores the effectiveness, advantages, and disadvantages of authorized COVID-19 vaccines. Additionally, it analyzes the challenges, strategies, and future prospects of developing a safe, broad-spectrum vaccine that confers sufficient and sustainable immune protection against new variants of SARS-CoV-2. These discussions not only offer insight for the development of next-generation COVID-19 vaccines but also summarize experiences for combating future emerging viruses.
Collapse
Affiliation(s)
- Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510000, China
| | - Shengmei Pang
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jiaqi Liu
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Qiangde Duan
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
215
|
Tatsi EB, Filippatos F, Bello T, Syriopoulou V, Michos A. Comparative Study of T-Cell Repertoires after COVID-19 Immunization with Homologous or Heterologous Vaccine Booster. Pathogens 2024; 13:284. [PMID: 38668239 PMCID: PMC11054887 DOI: 10.3390/pathogens13040284] [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: 01/24/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/29/2024] Open
Abstract
Sequencing of the T-cell repertoire is an innovative method to assess the cellular responses after immunization. The purpose of this study was to compare T-cell repertoires after COVID-19 immunization with homologous (HOB) and heterologous (HEB) boosting. The study included 20 participants with a median age of 27.5 (IQR:23) years, who were vaccinated with one dose of the Ad26.COV2.S vaccine and were boosted with either Ad26.COV2.S (n = 10) or BNT162b2 (n = 10) vaccine. Analysis of the T-cell receptor beta locus (TCRβ) sequencing one month after the booster dose identified that the HEB compared to the HOB group exhibited a higher number of both total and COVID-19-related functional T-cell rearrangements [mean of total productive rearrangements (TPRs): 63151.8 (SD ± 18441.5) vs. 34915.4 (SD ± 11121.6), p = 0.001 and COVID-19-TPRs: 522.5 (SD ± 178.0) vs. 298.3 (SD ± 101.1), p = 0.003]. A comparison between the HOB and HEB groups detected no statistically significant differences regarding T-cell Simpson clonality [0.021 (IQR:0.014) vs. 0.019 (IQR:0.007)], richness [8734.5 (IQR:973.3) vs. 8724 (IQR:383.7)] and T-cell fraction [0.19 (IQR:0.08) vs. 0.18 (IQR:0.08)]. HEB also exhibited a substantially elevated humoral immune response one month after the booster dose compared to HOB [median antibody titer (IQR): 10115.0 U/mL (6993.0) vs. 1781.0 U/mL (1314.0), p = 0.001]. T-cell repertoire sequencing indicated that HEB had increased SARS-CoV-2-related T-cell rearrangements, which was in accordance with higher humoral responses and possibly conferring longer protection. Data from the present study indicate that the administration of different COVID-19 vaccines as a booster may provide better protection.
Collapse
Affiliation(s)
- Elizabeth-Barbara Tatsi
- Infectious Diseases and Chemotherapy Research Laboratory, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-B.T.); (F.F.)
| | - Filippos Filippatos
- Infectious Diseases and Chemotherapy Research Laboratory, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-B.T.); (F.F.)
| | - Thomas Bello
- Adaptive Biotechnologies, Seattle 98109, WA, USA;
| | - Vasiliki Syriopoulou
- Infectious Diseases and Chemotherapy Research Laboratory, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-B.T.); (F.F.)
| | - Athanasios Michos
- Infectious Diseases and Chemotherapy Research Laboratory, First Department of Pediatrics, Medical School, “Aghia Sophia” Children’s Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.-B.T.); (F.F.)
| |
Collapse
|
216
|
Zhang Q, Pavlinov I, Ye Y, Zheng W. Therapeutic development targeting host heparan sulfate proteoglycan in SARS-CoV-2 infection. Front Med (Lausanne) 2024; 11:1364657. [PMID: 38618194 PMCID: PMC11014733 DOI: 10.3389/fmed.2024.1364657] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/18/2024] [Indexed: 04/16/2024] Open
Abstract
The global pandemic caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has led to an urgent need for effective therapeutic options. SARS-CoV-2 is a novel coronavirus responsible for the COVID-19 pandemic that has resulted in significant morbidity and mortality worldwide. The virus is known to enter host cells by binding to the angiotensin-converting enzyme 2 (ACE2) receptor, and emerging evidence suggests that heparan sulfate proteoglycans (HSPGs) play a crucial role in facilitating this process. HSPGs are abundant cell surface proteoglycan present in many tissues, including the lung, and have been shown to interact directly with the spike protein of SARS-CoV-2. This review aims to summarize the current understanding of the role of HSPGs in SARS-CoV-2 infection and the potential of developing new therapies targeting HSPGs.
Collapse
Affiliation(s)
- Qi Zhang
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Ivan Pavlinov
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| | - Yihong Ye
- Laboratory of Molecular Biology, National Institute of Diabetes, Digestive, and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Wei Zheng
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
217
|
Gardner BJ, Kilpatrick AM. Predicting Vaccine Effectiveness for Hospitalization and Symptomatic Disease for Novel SARS-CoV-2 Variants Using Neutralizing Antibody Titers. Viruses 2024; 16:479. [PMID: 38543844 PMCID: PMC10975673 DOI: 10.3390/v16030479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 05/23/2024] Open
Abstract
The emergence of new virus variants, including the Omicron variant (B.1.1.529) of SARS-CoV-2, can lead to reduced vaccine effectiveness (VE) and the need for new vaccines or vaccine doses if the extent of immune evasion is severe. Neutralizing antibody titers have been shown to be a correlate of protection for SARS-CoV-2 and other pathogens, and could be used to quickly estimate vaccine effectiveness for new variants. However, no model currently exists to provide precise VE estimates for a new variant against severe disease for SARS-CoV-2 using robust datasets from several populations. We developed predictive models for VE against COVID-19 symptomatic disease and hospitalization across a 54-fold range of mean neutralizing antibody titers. For two mRNA vaccines (mRNA-1273, BNT162b2), models fit without Omicron data predicted that infection with the BA.1 Omicron variant increased the risk of hospitalization 2.8-4.4-fold and increased the risk of symptomatic disease 1.7-4.2-fold compared to the Delta variant. Out-of-sample validation showed that model predictions were accurate; all predictions were within 10% of observed VE estimates and fell within the model prediction intervals. Predictive models using neutralizing antibody titers can provide rapid VE estimates, which can inform vaccine booster timing, vaccine design, and vaccine selection for new virus variants.
Collapse
Affiliation(s)
- Billy J. Gardner
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| | - A. Marm Kilpatrick
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| |
Collapse
|
218
|
Dhillon A, Persson BD, Volkov AN, Sülzen H, Kádek A, Pompach P, Kereïche S, Lepšík M, Danskog K, Uetrecht C, Arnberg N, Zoll S. Structural insights into the interaction between adenovirus C5 hexon and human lactoferrin. J Virol 2024; 98:e0157623. [PMID: 38323814 PMCID: PMC10949841 DOI: 10.1128/jvi.01576-23] [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: 10/10/2023] [Accepted: 01/13/2024] [Indexed: 02/08/2024] Open
Abstract
Adenovirus (AdV) infection of the respiratory epithelium is common but poorly understood. Human AdV species C types, such as HAdV-C5, utilize the Coxsackie-adenovirus receptor (CAR) for attachment and subsequently integrins for entry. CAR and integrins are however located deep within the tight junctions in the mucosa where they would not be easily accessible. Recently, a model for CAR-independent AdV entry was proposed. In this model, human lactoferrin (hLF), an innate immune protein, aids the viral uptake into epithelial cells by mediating interactions between the major capsid protein, hexon, and yet unknown host cellular receptor(s). However, a detailed understanding of the molecular interactions driving this mechanism is lacking. Here, we present a new cryo-EM structure of HAdV-5C hexon at high resolution alongside a hybrid structure of HAdV-5C hexon complexed with human lactoferrin (hLF). These structures reveal the molecular determinants of the interaction between hLF and HAdV-C5 hexon. hLF engages hexon primarily via its N-terminal lactoferricin (Lfcin) region, interacting with hexon's hypervariable region 1 (HVR-1). Mutational analyses pinpoint critical Lfcin contacts and also identify additional regions within hLF that critically contribute to hexon binding. Our study sheds more light on the intricate mechanism by which HAdV-C5 utilizes soluble hLF/Lfcin for cellular entry. These findings hold promise for advancing gene therapy applications and inform vaccine development. IMPORTANCE Our study delves into the structural aspects of adenovirus (AdV) infections, specifically HAdV-C5 in the respiratory epithelium. It uncovers the molecular details of a novel pathway where human lactoferrin (hLF) interacts with the major capsid protein, hexon, facilitating viral entry, and bypassing traditional receptors such as CAR and integrins. The study's cryo-EM structures reveal how hLF engages hexon, primarily through its N-terminal lactoferricin (Lfcin) region and hexon's hypervariable region 1 (HVR-1). Mutational analyses identify critical Lfcin contacts and other regions within hLF vital for hexon binding. This structural insight sheds light on HAdV-C5's mechanism of utilizing soluble hLF/Lfcin for cellular entry, holding promise for gene therapy and vaccine development advancements in adenovirus research.
Collapse
Affiliation(s)
- Arun Dhillon
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Alexander N. Volkov
- VIB-VUB Center for Structural Biology, Flemish Institute of Biotechnology (VIB), Brussels, Belgium
- Jean Jeener NMR Centre, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Hagen Sülzen
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
- Faculty of Science, Charles University, Prague, Czech Republic
| | - Alan Kádek
- Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
- Leibniz Institute of Virology (LIV), Hamburg, Germany
| | - Petr Pompach
- Biotechnology and Biomedical Center of the Academy of Sciences and Charles University in Vestec, Vestec, Czech Republic
| | - Sami Kereïche
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
- First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martin Lepšík
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| | - Katarina Danskog
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Charlotte Uetrecht
- Department of Health Sciences and Biomedicine, Faculty V: School of Life Sciences, CSSB Centre for Structural Systems Biology, Deutsches Elektronen Synchrotron DESY and Leibniz Institute of Virology, Hamburg, University of Siegen, Siegen, Germany
| | - Niklas Arnberg
- Department of Clinical Microbiology, Umeå University, Umeå, Sweden
| | - Sebastian Zoll
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czech Republic
| |
Collapse
|
219
|
Silva-Pilipich N, Beloki U, Salaberry L, Smerdou C. Self-Amplifying RNA: A Second Revolution of mRNA Vaccines against COVID-19. Vaccines (Basel) 2024; 12:318. [PMID: 38543952 PMCID: PMC10974399 DOI: 10.3390/vaccines12030318] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 11/12/2024] Open
Abstract
SARS-CoV-2 virus, the causative agent of COVID-19, has produced the largest pandemic in the 21st century, becoming a very serious health problem worldwide. To prevent COVID-19 disease and infection, a large number of vaccines have been developed and approved in record time, including new vaccines based on mRNA encapsulated in lipid nanoparticles. While mRNA-based vaccines have proven to be safe and effective, they are more expensive to produce compared to conventional vaccines. A special type of mRNA vaccine is based on self-amplifying RNA (saRNA) derived from the genome of RNA viruses, mainly alphaviruses. These saRNAs encode a viral replicase in addition to the antigen, usually the SARS-CoV-2 spike protein. The replicase can amplify the saRNA in transfected cells, potentially reducing the amount of RNA needed for vaccination and promoting interferon I responses that can enhance adaptive immunity. Preclinical studies with saRNA-based COVID-19 vaccines in diverse animal models have demonstrated the induction of robust protective immune responses, similar to conventional mRNA but at lower doses. Initial clinical trials have confirmed the safety and immunogenicity of saRNA-based vaccines in individuals that had previously received authorized COVID-19 vaccines. These findings have led to the recent approval of two of these vaccines by the national drug agencies of India and Japan, underscoring the promising potential of this technology.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Uxue Beloki
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| | - Laura Salaberry
- Facultad de Ingeniería, Universidad ORT Uruguay, Montevideo 11100, Uruguay;
- Nanogrow Biotech, Montevideo 11500, Uruguay
| | - Cristian Smerdou
- Division of DNA and RNA Medicine, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- Instituto de Investigación Sanitaria de Navarra (IdISNA) and CCUN, 31008 Pamplona, Spain
| |
Collapse
|
220
|
Ogando-Rivas E, Castillo P, Yang C, Trivedi V, Zhang D, Pohl-Guimarães F, Liu R, Barpujari A, Candelario KM, Mendez-Gomez H, Sayour EJ, Mitchell DA. Expanded specific T cells to hypomutated regions of the SARS-CoV-2 using mRNA electroporated antigen-presenting cells. Mol Ther Methods Clin Dev 2024; 32:101192. [PMID: 38327807 PMCID: PMC10847775 DOI: 10.1016/j.omtm.2024.101192] [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: 04/10/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
The COVID-19 pandemic has caused about seven million deaths worldwide. Preventative vaccines have been developed including Spike gp mRNA-based vaccines that provide protection to immunocompetent patients. However, patients with primary immunodeficiencies, patients with cancer, or hematopoietic stem cell transplant recipients are not able to mount robust immune responses against current vaccine approaches. We propose to target structural SARS-CoV-2 antigens (i.e., Spike gp, Membrane, Nucleocapsid, and Envelope) using circulating human antigen-presenting cells electroporated with full length SARS-CoV-2 structural protein-encoding mRNAs to activate and expand specific T cells. Based on the Th1-type cytokine and cytolytic enzyme secretion upon antigen rechallenge, we were able to generate SARS-CoV-2 specific T cells in up to 70% of unexposed unvaccinated healthy donors (HDs) after 3 subsequent stimulations and in 100% of recovered patients (RPs) after 2 stimulations. By means of SARS-CoV-2 specific TCRβ repertoire analysis, T cells specific to Spike gp-derived hypomutated regions were identified in HDs and RPs despite viral genomic evolution. Hence, we demonstrated that SARS-CoV-2 mRNA-loaded antigen-presenting cells are effective activating and expanding COVID19-specific T cells. This approach represents an alternative to patients who are not able to mount adaptive immune responses to current COVID-19 vaccines with potential protection across new variants that have conserved genetic regions.
Collapse
Affiliation(s)
- Elizabeth Ogando-Rivas
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Paul Castillo
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Changlin Yang
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Vrunda Trivedi
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Dingpeng Zhang
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Fernanda Pohl-Guimarães
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Ruixuan Liu
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Arnav Barpujari
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kate M. Candelario
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Hector Mendez-Gomez
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| | - Elias J. Sayour
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
- UF Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Duane A. Mitchell
- UF Brain Tumor Immunotherapy Program, Preston A. Wells Center for Brain Tumor Therapy, Lillian S. Wells Department of Neurosurgery, University of Florida, Gainesville, FL, USA
| |
Collapse
|
221
|
Magaret CA, Li L, deCamp AC, Rolland M, Juraska M, Williamson BD, Ludwig J, Molitor C, Benkeser D, Luedtke A, Simpkins B, Heng F, Sun Y, Carpp LN, Bai H, Dearlove BL, Giorgi EE, Jongeneelen M, Brandenburg B, McCallum M, Bowen JE, Veesler D, Sadoff J, Gray GE, Roels S, Vandebosch A, Stieh DJ, Le Gars M, Vingerhoets J, Grinsztejn B, Goepfert PA, de Sousa LP, Silva MST, Casapia M, Losso MH, Little SJ, Gaur A, Bekker LG, Garrett N, Truyers C, Van Dromme I, Swann E, Marovich MA, Follmann D, Neuzil KM, Corey L, Greninger AL, Roychoudhury P, Hyrien O, Gilbert PB. Quantifying how single dose Ad26.COV2.S vaccine efficacy depends on Spike sequence features. Nat Commun 2024; 15:2175. [PMID: 38467646 PMCID: PMC10928100 DOI: 10.1038/s41467-024-46536-w] [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: 05/16/2023] [Accepted: 02/29/2024] [Indexed: 03/13/2024] Open
Abstract
In the ENSEMBLE randomized, placebo-controlled phase 3 trial (NCT04505722), estimated single-dose Ad26.COV2.S vaccine efficacy (VE) was 56% against moderate to severe-critical COVID-19. SARS-CoV-2 Spike sequences were determined from 484 vaccine and 1,067 placebo recipients who acquired COVID-19. In this set of prespecified analyses, we show that in Latin America, VE was significantly lower against Lambda vs. Reference and against Lambda vs. non-Lambda [family-wise error rate (FWER) p < 0.05]. VE differed by residue match vs. mismatch to the vaccine-insert at 16 amino acid positions (4 FWER p < 0.05; 12 q-value ≤ 0.20); significantly decreased with physicochemical-weighted Hamming distance to the vaccine-strain sequence for Spike, receptor-binding domain, N-terminal domain, and S1 (FWER p < 0.001); differed (FWER ≤ 0.05) by distance to the vaccine strain measured by 9 antibody-epitope escape scores and 4 NTD neutralization-impacting features; and decreased (p = 0.011) with neutralization resistance level to vaccinee sera. VE against severe-critical COVID-19 was stable across most sequence features but lower against the most distant viruses.
Collapse
Affiliation(s)
- Craig A Magaret
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Li Li
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Allan C deCamp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Morgane Rolland
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Michal Juraska
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Brian D Williamson
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Biostatistics Division, Kaiser Permanente Washington Health Research Institute, Seattle, WA, USA
| | - James Ludwig
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Cindy Molitor
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David Benkeser
- Departments of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - Alex Luedtke
- Department of Statistics, University of Washington, Seattle, WA, USA
| | - Brian Simpkins
- Department of Computer Science, Pitzer College, Claremont, CA, USA
| | - Fei Heng
- University of North Florida, Jacksonville, FL, USA
| | - Yanqing Sun
- University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Lindsay N Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Hongjun Bai
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Bethany L Dearlove
- US Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD, USA
| | - Elena E Giorgi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Mandy Jongeneelen
- Johnson & Johnson Innovative Medicine, Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Boerries Brandenburg
- Johnson & Johnson Innovative Medicine, Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - John E Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Jerald Sadoff
- Johnson & Johnson Innovative Medicine, Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Glenda E Gray
- Perinatal HIV Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - Sanne Roels
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - An Vandebosch
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Daniel J Stieh
- Johnson & Johnson Innovative Medicine, Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Mathieu Le Gars
- Johnson & Johnson Innovative Medicine, Janssen Vaccines & Prevention B.V, Leiden, The Netherlands
| | - Johan Vingerhoets
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Beatriz Grinsztejn
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Leonardo Paiva de Sousa
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Mayara Secco Torres Silva
- Evandro Chagas National Institute of Infectious Diseases-Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Martin Casapia
- Facultad de Medicina Humana, Universidad Nacional de la Amazonia Peru, Iquitos, Peru
| | - Marcelo H Losso
- Hospital General de Agudos José María Ramos Mejia, Buenos Aires, Argentina
| | - Susan J Little
- Division of Infectious Diseases, University of California San Diego, La Jolla, CA, USA
| | - Aditya Gaur
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Linda-Gail Bekker
- The Desmond Tutu HIV Centre, University of Cape Town, Observatory, Cape Town, South Africa
| | - Nigel Garrett
- Centre for the AIDS Programme of Research in South Africa, University of KwaZulu-Natal, Durban, South Africa
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
| | - Carla Truyers
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Ilse Van Dromme
- Janssen R&D, a division of Janssen Pharmaceutica NV, Beerse, Belgium
| | - Edith Swann
- Vaccine Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mary A Marovich
- Vaccine Research Program, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dean Follmann
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen M Neuzil
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Alexander L Greninger
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Pavitra Roychoudhury
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Peter B Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Biostatistics, University of Washington School of Public Health, Seattle, WA, USA.
| |
Collapse
|
222
|
Luo R, Wan Y, Liu G, Chen J, Luo X, Li Z, Su D, Lu N, Luo Z. Engineering Self-Assembling Peptide Hydrogel to Enhance the Capacity of Dendritic Cells to Activate In Vivo T-Cell Immunity. Biomacromolecules 2024; 25:1408-1428. [PMID: 38236703 DOI: 10.1021/acs.biomac.3c00511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
The efficacy of the dendritic cell (DC) has failed to meet expectations thus far, and crucial problems such as the immature state of DCs, low targeting efficiency, insufficient number of dendritic cells, and microenvironment are still the current focus. To address these problems, we developed two self-assembling peptides, RLDI and RQDT, that mimic extracellular matrix (ECM). These peptides can be self-assembled into highly ordered three-dimensional nanofiber scaffold structures, where RLDI can form gelation immediately. In addition, we found that RLDI and RQDT enhance the biological function of DCs, including releasing antigens sustainably, adhering to DCs, promoting the maturation of DCs, and increasing the ability of DC antigen presentation. Moreover, peptide hydrogel-based DC treatment significantly achieved prophylactic and treatment effects on colon cancer. These results have certain implications for the design of new broad-spectrum vaccines in the future.
Collapse
Affiliation(s)
- Ruyue Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Yuan Wan
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
- Roy J. Carver Department of Biomedical Engineering, College of Engineering, University of Iowa, Iowa City, Iowa 52242, United States
| | - Guicen Liu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Jialei Chen
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Xinyi Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhaoxu Li
- Department of Materials Science and Engineering, University of California, Irvine, Irvine, California 92697, United States
| | - Di Su
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Na Lu
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhongli Luo
- College of Basic Medical Sciences, Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
223
|
Luo H, Zhou Q, Feng J, Wu Y, Chen H, Mao M, Qi R. Global Prevalence of Preexisting Antibodies against Human Adenoviruses, Surveyed from 1962 to 2021. Intervirology 2024; 67:19-39. [PMID: 38452738 PMCID: PMC11006277 DOI: 10.1159/000538233] [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: 01/15/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Human adenoviruses (HAdVs) are extensively used as vectors for vaccines development and cancer therapy. People who already have antibodies against HAdVs, on the other hand, would have an impact on the preventative or therapeutic effect. This review focuses primarily on the prevalence of pre-existing antibodies against HAdVs in distinct geographical populations. SUMMARY After screening, 64 studies from 31 countries between 1962 and 2021 were selected, totaling 39,427 samples. The total prevalence of preexisting antibodies to HAdVs varied by country or location, ranging from 2.00 to 95.70%. Southeast Asia had the highest prevalence (54.57%) while Europe had the lowest (18.17%). The prevalence in practically all developing nations was higher than in developed nations. Adults have a greater frequency than children and newborns in most nations. The primary HAdV antibody types varied by country. Adults in China, the USA, the United Kingdom, and Belgium had the lowest prevalence of preexisting antibodies against HAdV55, HAdV37, HAdV8, and HAdV36, respectively. Children in the USA, China, the United Kingdom, and Japan had the lowest rates of HAdV48, HAdV11, HAdV8, and HAdV40. The frequency of antibodies differed significantly between military and civilian groups. KEY MESSAGES Preexisting antibodies against various types of HAdVs differed greatly throughout worldwide populations. Future development of HAdV-vector vaccines and medicines should focus on preexisting antibodies in target groups rather than a "one-size-fits-all" strategy. It might be advantageous in selecting HAdV vectors for studying the prevalence of preexisting antibodies against HAdVs in different locations and people throughout the world.
Collapse
Affiliation(s)
- Hui Luo
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Qian Zhou
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Jinqi Feng
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Yi Wu
- School of Public Health, Lanzhou University, Lanzhou, China
| | | | - Meihan Mao
- School of Public Health, Lanzhou University, Lanzhou, China
| | - Rui Qi
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
224
|
Toback S, Marchese AM, Warren B, Ayman S, Zarkovic S, ElTantawy I, Mallory RM, Rousculp M, Almarzooqi F, Piechowski-Jozwiak B, Bonilla MF, Bakkour AE, Hussein SE, Al Kaabi N. Safety and immunogenicity of the NVX-CoV2373 vaccine as a booster in adults previously vaccinated with the BBIBP-CorV vaccine. Vaccine 2024; 42:1777-1784. [PMID: 38365482 DOI: 10.1016/j.vaccine.2024.02.037] [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: 09/26/2023] [Revised: 02/01/2024] [Accepted: 02/10/2024] [Indexed: 02/18/2024]
Abstract
This phase 3 observer-blind, randomized, controlled study was conducted in adults ≥18 years of age to assess the safety and immunogenicity of NVX-CoV2373 as a heterologous booster compared to BBIBP-CorV when utilized as a homologous booster. Approximately 1000 participants were randomly assigned in a 1:1 ratio to receive a single dose of NVX-CoV2373 or BBIBP-CorV after prior vaccination with 2 or 3 doses of BBIBP-CorV. Solicited adverse events (AEs) were collected for 7 days after vaccination. Unsolicited AEs were collected for 28 days following the booster dose and serious adverse and adverse events of special interest (AESI) were collected throughout the study. Anti-spike IgG and neutralizing antibodies against SARS-CoV-2 were measured at baseline, day 14, day 28, and day 180. The study achieved its primary non-inferiority endpoint and also demonstrated statistically higher neutralization responses when NVX-CoV2373 was utilized as a heterologous booster compared with BBIBP-CorV as a homologous booster. Both vaccines had an acceptably low reactogenicity profile, and no new safety concerns were found. Heterologous boosting with NVX-CoV2373 was a highly immunogenic and safe vaccine regimen in those previously vaccinated with BBIBP-CorV.
Collapse
Affiliation(s)
- Seth Toback
- Novavax Inc., 700 Quince Orchard Rd, Gaithersburg, MD 20878, United States.
| | - Anthony M Marchese
- Novavax Inc., 700 Quince Orchard Rd, Gaithersburg, MD 20878, United States.
| | - Brandy Warren
- Novavax Inc., 700 Quince Orchard Rd, Gaithersburg, MD 20878, United States.
| | - Sondos Ayman
- Insights Research Organization & Solutions (IROS), Building of Masdar M13 T Limited, SE 45_05, Plot C16, Khalifa City, Abu Dhabi, United Arab Emirates.
| | - Senka Zarkovic
- Insights Research Organization & Solutions (IROS), Building of Masdar M13 T Limited, SE 45_05, Plot C16, Khalifa City, Abu Dhabi, United Arab Emirates.
| | - Islam ElTantawy
- Insights Research Organization & Solutions (IROS), Building of Masdar M13 T Limited, SE 45_05, Plot C16, Khalifa City, Abu Dhabi, United Arab Emirates.
| | - Raburn M Mallory
- Novavax Inc., 700 Quince Orchard Rd, Gaithersburg, MD 20878, United States.
| | - Matthew Rousculp
- Novavax Inc., 700 Quince Orchard Rd, Gaithersburg, MD 20878, United States.
| | - Fahed Almarzooqi
- G42 Healthcare, 3(rd) Floor, 1B Building, Mohamed bin Zayed University of Artificial Intelligence, Masdar City, Abu Dhabi, United Arab Emirates.
| | - Bartlomiej Piechowski-Jozwiak
- Cleveland Clinic Abu Dhabi, 59 Hamouda Bin Ali Al Dhaheri St - Al Maryah Island - Abu Dhabi Global Market Square, Abu Dhabi, United Arab Emirates.
| | - Maria-Fernanda Bonilla
- Cleveland Clinic Abu Dhabi, 59 Hamouda Bin Ali Al Dhaheri St - Al Maryah Island - Abu Dhabi Global Market Square, Abu Dhabi, United Arab Emirates.
| | - Agyad Ebrahim Bakkour
- Sheikh Khalifa Medical City, SEHA, Al Karamah St - Al Manhal - Al Tibbiya, Abu Dhabi, United Arab Emirates.
| | - Salah Eldin Hussein
- Sheikh Khalifa Medical City, SEHA, Al Karamah St - Al Manhal - Al Tibbiya, Abu Dhabi, United Arab Emirates.
| | - Nawal Al Kaabi
- Sheikh Khalifa Medical City, SEHA, Al Karamah St - Al Manhal - Al Tibbiya, Abu Dhabi, United Arab Emirates; College of Medicine and Health Sciences, Khalifa University, Shakhbout Bin Sultan St - Hadbat Al Za'faranah - Zone 1, Abu Dhabi, United Arab Emirates
| |
Collapse
|
225
|
Roederer AL, Cao Y, Denis KS, Sheehan ML, Li CJ, Lam EC, Gregory DJ, Poznansky MC, Iafrate AJ, Canaday DH, Gravenstein S, Garcia-Beltran WF, Balazs AB. Ongoing evolution of SARS-CoV-2 drives escape from mRNA vaccine-induced humoral immunity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.05.24303815. [PMID: 38496628 PMCID: PMC10942518 DOI: 10.1101/2024.03.05.24303815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Since the COVID-19 pandemic began in 2020, viral sequencing has documented 131 individual mutations in the viral spike protein across 48 named variants. To determine the ability of vaccine-mediated humoral immunity to keep pace with continued SARS-CoV-2 evolution, we assessed the neutralization potency of sera from 76 vaccine recipients collected after 2 to 6 immunizations against a comprehensive panel of mutations observed during the pandemic. Remarkably, while many individual mutations that emerged between 2020 and 2022 exhibit escape from sera following primary vaccination, few escape boosted sera. However, progressive loss of neutralization was observed across newer variants, irrespective of vaccine doses. Importantly, an updated XBB.1.5 booster significantly increased titers against newer variants but not JN.1. These findings demonstrate that seasonal boosters improve titers against contemporaneous strains, but novel variants continue to evade updated mRNA vaccines, demonstrating the need for novel approaches to adequately control SARS-CoV-2 transmission.
Collapse
Affiliation(s)
- Alex L. Roederer
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Yi Cao
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Kerri St. Denis
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | | | - Chia Jung Li
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - Evan C. Lam
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - David J. Gregory
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, 02129, USA
- Pediatric Infectious Disease, Massachusetts General Hospital for Children, Boston, MA 02114, USA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, MA, 02129, USA
- Massachusetts General Hospital Cancer Center, Boston, MA, 02114, USA
| | - A. John Iafrate
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - David H. Canaday
- Case Western Reserve University School of Medicine, Cleveland, OH
- Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Department of Veterans Affairs Medical Center, Cleveland, Ohio
| | - Stefan Gravenstein
- Center of Innovation in Long-Term Services and Supports, Veterans Administration Medical Center, Providence, Rhode Island
- Division of Geriatrics and Palliative Medicine, Alpert Medical School of Brown University, Providence, Rhode Island, USA
- Brown University School of Public Health Center for Gerontology and Healthcare Research, Providence, Rhode Island
| | - Wilfredo F. Garcia-Beltran
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, 02114, USA
| | | |
Collapse
|
226
|
Vitti JN, Vitti R, Chu K, Mellis S. The ethics of clinical research in the era of COVID-19. Front Public Health 2024; 12:1359654. [PMID: 38510356 PMCID: PMC10950982 DOI: 10.3389/fpubh.2024.1359654] [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: 12/21/2023] [Accepted: 02/20/2024] [Indexed: 03/22/2024] Open
Abstract
There is an urgent need for increased understanding of COVID-19 and strategies for its prevention, treatment, and mitigation. All participants in the research enterprise, including institutional review boards, have an ethical duty to protect participants and ensure that the benefits gained from such research do not conflict with the core principles that guided researchers prior to the pandemic. In this review, we discuss the ethical issues surrounding initiation and conduct of clinical trials, focusing on novel COVID-19 therapeutic, vaccine, or biospecimen research, using the principles of autonomy, beneficence, and justice. We discuss strategies to manage the practical challenges associated with the conduct of clinical trials, with an emphasis on maintaining the rights and welfare of research participants.
Collapse
Affiliation(s)
| | - Robert Vitti
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, United States
| | - Karen Chu
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, United States
| | - Scott Mellis
- Regeneron Pharmaceuticals Inc., Tarrytown, NY, United States
| |
Collapse
|
227
|
O'Regan E, Svalgaard IB, Sørensen AIV, Spiliopoulos L, Bager P, Nielsen NM, Hansen JV, Koch A, Meder IK, Videbech P, Ethelberg S, Hviid A. A register and questionnaire study of long-term general health symptoms following SARS-CoV-2 vaccination in Denmark. NPJ Vaccines 2024; 9:52. [PMID: 38438399 PMCID: PMC10912726 DOI: 10.1038/s41541-024-00844-w] [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: 07/27/2023] [Accepted: 02/21/2024] [Indexed: 03/06/2024] Open
Abstract
Many individuals who refuse COVID-19 vaccination have concerns about long-term side effects. Here, we report findings on self-reported symptoms from a Danish survey- and register study. The study included 34,868 vaccinated primary course recipients, 95.8% of whom received mRNA vaccines, and 1,568 unvaccinated individuals. Participants had no known history of SARS-CoV-2 infection. Using g-computation on logistic regression, risk differences (RDs) for symptoms between vaccinated and unvaccinated persons were estimated with adjustments for possible confounders. Within six weeks after vaccination, higher risks were observed for physical exhaustion (RD 4.9%, 95% CI 1.1% to 8.4%), fever or chills (RD 4.4%, 95% CI 2.1% to 6.7%), and muscle/joint pain (RD 7.0%, 95% CI 3.1% to 10.7%), compared to unvaccinated individuals. Beyond twenty-six weeks, risks were higher among the vaccinated for sleeping problems (RD 3.0, 95% 0.2 to 5.8), fever or chills (RD 2.0, 95% CI 0.4 to 3.6), reduced/altered taste (RD 1.2, 95% CI 0.2 to 2.3) and shortness of breath (RD 2.6, 95% CI 0.9 to 4.0). However, when examining pre-omicron responses only, the difference for reduced/altered taste was significant. As expected, the risk of experiencing physical exhaustion, fever or chills, and muscle/joint pain was higher among persons who responded within six weeks of completing the primary course. No significant differences were observed for the 7-25-week period after vaccination. Associations for the period beyond 26 weeks must be interpreted with caution and in the context of undetected SARS-CoV-2 infection, wide confidence intervals, and multiple testing. Overall, we observe no concerning signs of long-term self-reported physical, cognitive, or fatigue symptoms after vaccination.
Collapse
Affiliation(s)
- Elisabeth O'Regan
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark.
| | - Ingrid Bech Svalgaard
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
| | | | - Lampros Spiliopoulos
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
| | - Peter Bager
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
- Infectious Disease Epidemiology and Prevention, Statens Serum Institut, 2300, Copenhagen, Denmark
| | - Nete Munk Nielsen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
- Focused Research Unit in Neurology, Department of Neurology, Hospital of Southern Jutland, University of Southern Denmark, 6200, Aabenraa, Denmark
| | - Jørgen Vinsløv Hansen
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
| | - Anders Koch
- Infectious Disease Epidemiology and Prevention, Statens Serum Institut, 2300, Copenhagen, Denmark
- Department of Public Health, Global Health Section, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Rigshospitalet University Hospital, 2100, Copenhagen, Denmark
| | - Inger Kristine Meder
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
| | - Poul Videbech
- Center for Neuropsychiatric Depression Research, Mental Health Center Glostrup, 2600 Glostrup, Denmark and University of Copenhagen, Copenhagen, Denmark
| | - Steen Ethelberg
- Infectious Disease Epidemiology and Prevention, Statens Serum Institut, 2300, Copenhagen, Denmark
- Department of Public Health, Global Health Section, University of Copenhagen, Copenhagen, Denmark
| | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, 2300, Copenhagen, Denmark
- Pharmacovigilance Research Centre, Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| |
Collapse
|
228
|
Qin L, Sun Y, Gao N, Ling G, Zhang P. Nanotechnology of inhalable vaccines for enhancing mucosal immunity. Drug Deliv Transl Res 2024; 14:597-620. [PMID: 37747597 DOI: 10.1007/s13346-023-01431-7] [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] [Accepted: 09/05/2023] [Indexed: 09/26/2023]
Abstract
Vaccines are the cornerstone of world health. The majority of vaccines are formulated as injectable products, facing the drawbacks of cold chain transportation, needle-stick injuries, and primary systemic immunity. Inhalable vaccines exhibited unique advantages due to their small dose, easy to use, quick effect, and simultaneous induction of mucosal and systemic responses. Facing global pandemics, especially the coronavirus disease 2019 (COVID-19), a majority of inhalable vaccines are in preclinical or clinical trials. A better understanding of advanced delivery technologies of inhalable vaccines may provide new scientific insights for developing inhalable vaccines. In this review article, detailed immune mechanisms involving mucosal, cellular, and humoral immunity were described. The preparation methods of inhalable vaccines were then introduced. Advanced nanotechnologies of inhalable vaccines containing inhalable nucleic acid vaccines, inhalable adenovirus vector vaccines, novel adjuvant-assisted inhalable vaccines, and biomaterials for inhalable vaccine delivery were emphatically discussed. Meanwhile, the latest clinical progress in inhalable vaccines for COVID-19 and tuberculosis was discussed.
Collapse
Affiliation(s)
- Li Qin
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Yanhua Sun
- Shandong Provincial Key Laboratory of Microparticles Drug Delivery Technology, Qilu Pharmaceutical Co. Ltd., No. 243, Gongyebei Road, Jinan, 250100, China
| | - Nan Gao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, China.
| |
Collapse
|
229
|
Kruglov AA, Bondareva MA, Gogoleva VS, Semin IK, Astrakhantseva IV, Zvartsev R, Lunin AS, Apolokhov VD, Shustova EY, Volok VP, Ustyugov AA, Ishmukhametov AA, Nedospasov SA, Kozlovskaya LI, Drutskaya MS. Inactivated whole virion vaccine protects K18-hACE2 Tg mice against the Omicron SARS-CoV-2 variant via cross-reactive T cells and nonneutralizing antibody responses. Eur J Immunol 2024; 54:e2350664. [PMID: 38088236 DOI: 10.1002/eji.202350664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 01/02/2024]
Abstract
COVID-19 is a systemic inflammatory disease initiated by SARS-CoV-2 virus infection. Multiple vaccines against the Wuhan variant of SARS-CoV-2 have been developed including a whole virion beta-propiolactone-inactivated vaccine based on the B.1.1 strain (CoviVac). Since most of the population has been vaccinated by targeting the original or early variants of SARS-CoV-2, the emergence of novel mutant variants raises concern over possible evasion of vaccine-induced immune responses. Here, we report on the mechanism of protection by CoviVac, a whole virion-based vaccine, against the Omicron variant. CoviVac-immunized K18-hACE2 Tg mice were protected against both prototype B.1.1 and BA.1-like (Omicron) variants. Subsequently, vaccinated K18-hACE2 Tg mice rapidly cleared the infection via cross-reactive T-cell responses and cross-reactive, non-neutralizing antibodies recognizing the Omicron variant Spike protein. Thus, our data indicate that efficient protection from SARS-CoV-2 variants can be achieved by the orchestrated action of cross-reactive T cells and non-neutralizing antibodies.
Collapse
Affiliation(s)
- Andrey A Kruglov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Systems Rheumatology, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Marina A Bondareva
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Systems Rheumatology, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Violetta S Gogoleva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Iaroslav K Semin
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Department of Systems Rheumatology, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Irina V Astrakhantseva
- Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Russia
| | - Ruslan Zvartsev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr S Lunin
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Vasiliy D Apolokhov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Elena Yu Shustova
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Viktor P Volok
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
| | - Aleksey A Ustyugov
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medical Chemistry, Russian Academy of Sciences, Chernogolovka, Russia
| | - Aydar A Ishmukhametov
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Moskva, Moscow, Russia
| | - Sergei A Nedospasov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology and Biological Faculty, M.V. Lomonosov Moscow State University, Moscow, Russia
- Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Russia
- Institute of Cell Biology and Neurobiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Liubov I Kozlovskaya
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences (Institute of Poliomyelitis), Moscow, Russia
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), Moskva, Moscow, Russia
| | - Marina S Drutskaya
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Sirius University of Science and Technology, Federal Territory Sirius, Krasnodarsky Krai, Russia
| |
Collapse
|
230
|
Baxter RM, Cabrera-Martinez B, Ghosh T, Rester C, Moreno MG, Borko TL, Selva S, Fleischer CL, Haakonsen N, Mayher A, Bowhay E, Evans C, Miller TM, Huey L, McWilliams J, van Bokhoven A, Deane KD, Knight V, Jordan KR, Ghosh D, Klarquist J, Kedl RM, Piquet AL, Hsieh EWY. SARS-CoV-2 Vaccine-Elicited Immunity after B Cell Depletion in Multiple Sclerosis. Immunohorizons 2024; 8:254-268. [PMID: 38483384 PMCID: PMC10985059 DOI: 10.4049/immunohorizons.2300108] [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: 11/27/2023] [Accepted: 02/15/2024] [Indexed: 04/04/2024] Open
Abstract
The impact of B cell deficiency on the humoral and cellular responses to SARS-CoV2 mRNA vaccination remains a challenging and significant clinical management question. We evaluated vaccine-elicited serological and cellular responses in 1) healthy individuals who were pre-exposed to SARS-CoV-2 (n = 21), 2) healthy individuals who received a homologous booster (mRNA, n = 19; or Novavax, n = 19), and 3) persons with multiple sclerosis on B cell depletion therapy (MS-αCD20) receiving mRNA homologous boosting (n = 36). Pre-exposure increased humoral and CD4 T cellular responses in immunocompetent individuals. Novavax homologous boosting induced a significantly more robust serological response than mRNA boosting. MS-α CD20 had an intact IgA mucosal response and an enhanced CD8 T cell response to mRNA boosting compared with immunocompetent individuals. This enhanced cellular response was characterized by the expansion of only effector, not memory, T cells. The enhancement of CD8 T cells in the setting of B cell depletion suggests a regulatory mechanism between B and CD8 T cell vaccine responses.
Collapse
Affiliation(s)
- Ryan M. Baxter
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | | | - Tusharkanti Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Cody Rester
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Miguel Guerrero Moreno
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Tyler L. Borko
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Sean Selva
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Chelsie L. Fleischer
- Department of Medicine, Division of Rheumatology, University of Colorado, School of Medicine, Aurora, CO
| | - Nicola Haakonsen
- Department of Medicine, Division of Infectious Diseases, University of Colorado, School of Medicine, Aurora, CO
| | - Ariana Mayher
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Emily Bowhay
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Courtney Evans
- Allergy and Immunology Research, Research Institute, Children’s Hospital Colorado, Aurora, CO
| | - Todd M. Miller
- Analytics Resource Center, Children’s Hospital Colorado, Aurora, CO
| | - Leah Huey
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| | - Jennifer McWilliams
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Adrie van Bokhoven
- Department of Pathology, Section of Pathology Shared Resource, University of Colorado, Aurora, CO
| | - Kevin D. Deane
- Department of Medicine, Division of Rheumatology, University of Colorado, School of Medicine, Aurora, CO
| | - Vijaya Knight
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| | - Kimberly R. Jordan
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Debashis Ghosh
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Jared Klarquist
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Ross M. Kedl
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
| | - Amanda L. Piquet
- Department of Neurology, Sections of Neuroimmunology, Neuroinfectious Disease, and Neurohospitalist, University of Colorado School of Medicine, Aurora, CO
| | - Elena W. Y. Hsieh
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO
- Department of Pediatrics, Section of Allergy and Immunology, University of Colorado, School of Medicine, Aurora, CO
| |
Collapse
|
231
|
Lasrado N, Collier ARY, Miller J, Hachmann NP, Liu J, Anand T, A. Bondzie E, Fisher JL, Mazurek CR, Patio RC, Rodrigues SL, Rowe M, Surve N, Ty DM, Wu C, Chicz TM, Tong X, Korber B, McNamara RP, Barouch DH. Waning immunity and IgG4 responses following bivalent mRNA boosting. SCIENCE ADVANCES 2024; 10:eadj9945. [PMID: 38394195 PMCID: PMC10889350 DOI: 10.1126/sciadv.adj9945] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 01/22/2024] [Indexed: 02/25/2024]
Abstract
Messenger RNA (mRNA) vaccines were highly effective against the ancestral SARS-CoV-2 strain, but the efficacy of bivalent mRNA boosters against XBB variants was substantially lower. Here, we show limited durability of neutralizing antibody (NAb) responses against XBB variants and isotype switching to immunoglobulin G4 (IgG4) responses following bivalent mRNA boosting. Bivalent mRNA boosting elicited modest XBB.1-, XBB.1.5-, and XBB.1.16-specific NAbs that waned rapidly within 3 months. In contrast, bivalent mRNA boosting induced more robust and sustained NAbs against the ancestral WA1/2020 strain, suggesting immune imprinting. Following bivalent mRNA boosting, serum antibody responses were primarily IgG2 and IgG4 responses with poor Fc functional activity. In contrast, a third monovalent mRNA immunization boosted all isotypes including IgG1 and IgG3 with robust Fc functional activity. These data show substantial immune imprinting for the ancestral spike and isotype switching to IgG4 responses following bivalent mRNA boosting, with important implications for future booster designs and boosting strategies.
Collapse
Affiliation(s)
- Ninaad Lasrado
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ai-ris Y. Collier
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jessica Miller
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nicole P. Hachmann
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jinyan Liu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Trisha Anand
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Esther A. Bondzie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jana L. Fisher
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Camille R. Mazurek
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Robert C. Patio
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | | | - Marjorie Rowe
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Nehalee Surve
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Darren M. Ty
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Cindy Wu
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Taras M. Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Xin Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Bette Korber
- Los Alamos National Laboratory and New Mexico Consortium, Los Alamos, NM, USA
| | | | - Dan H. Barouch
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| |
Collapse
|
232
|
Altincekic N, Jores N, Löhr F, Richter C, Ehrhardt C, Blommers MJJ, Berg H, Öztürk S, Gande SL, Linhard V, Orts J, Abi Saad MJ, Bütikofer M, Kaderli J, Karlsson BG, Brath U, Hedenström M, Gröbner G, Sauer UH, Perrakis A, Langer J, Banci L, Cantini F, Fragai M, Grifagni D, Barthel T, Wollenhaupt J, Weiss MS, Robertson A, Bax A, Sreeramulu S, Schwalbe H. Targeting the Main Protease (M pro, nsp5) by Growth of Fragment Scaffolds Exploiting Structure-Based Methodologies. ACS Chem Biol 2024; 19:563-574. [PMID: 38232960 PMCID: PMC10877576 DOI: 10.1021/acschembio.3c00720] [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: 11/27/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 01/19/2024]
Abstract
The main protease Mpro, nsp5, of SARS-CoV-2 (SCoV2) is one of its most attractive drug targets. Here, we report primary screening data using nuclear magnetic resonance spectroscopy (NMR) of four different libraries and detailed follow-up synthesis on the promising uracil-containing fragment Z604 derived from these libraries. Z604 shows time-dependent binding. Its inhibitory effect is sensitive to reducing conditions. Starting with Z604, we synthesized and characterized 13 compounds designed by fragment growth strategies. Each compound was characterized by NMR and/or activity assays to investigate their interaction with Mpro. These investigations resulted in the four-armed compound 35b that binds directly to Mpro. 35b could be cocrystallized with Mpro revealing its noncovalent binding mode, which fills all four active site subpockets. Herein, we describe the NMR-derived fragment-to-hit pipeline and its application for the development of promising starting points for inhibitors of the main protease of SCoV2.
Collapse
Affiliation(s)
- Nadide Altincekic
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Nathalie Jores
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Frank Löhr
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Institute
of Biophysical Chemistry, Goethe University
Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Christian Richter
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Claus Ehrhardt
- Department
of Biochemistry, University of Zurich, 8093 Zurich, Switzerland
| | | | - Hannes Berg
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Sare Öztürk
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Santosh L. Gande
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Verena Linhard
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Julien Orts
- Department
of Pharmaceutical Sciences, University of
Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Marie Jose Abi Saad
- Department
of Pharmaceutical Sciences, University of
Vienna, Josef-Holaubek-Platz 2, 1090 Vienna, Austria
| | - Matthias Bütikofer
- Swiss
Federal Institute of Technology, Laboratory of Physical Chemistry, ETH Zurich, 8093 Zürich, Switzerland
| | - Janina Kaderli
- Swiss
Federal Institute of Technology, Laboratory of Physical Chemistry, ETH Zurich, 8093 Zürich, Switzerland
| | - B. Göran Karlsson
- Swedish
NMR Centre, Department of Chemistry and Molecular Biology, University of Gothenburg, SE40530 Göteborg, Sweden
- SciLifeLab, University of Gothenburg, SE40530 Göteborg, Sweden
| | - Ulrika Brath
- Swedish
NMR Centre, Department of Chemistry and Molecular Biology, University of Gothenburg, SE40530 Göteborg, Sweden
| | - Mattias Hedenström
- Swedish
NMR Centre, Department of Chemistry, University
of Umeå, SE-90187 Umeå, Sweden
| | - Gerhard Gröbner
- Swedish
NMR Centre, Department of Chemistry, University
of Umeå, SE-90187 Umeå, Sweden
| | - Uwe H. Sauer
- Protein
Production Sweden, Department of Chemistry, University of Umeå, SE-90187 Umeå, Sweden
| | - Anastassis Perrakis
- Oncode
Institute and Division of Biochemistry, The Netherlands Cancer Institute, 1066CX Amsterdam, The Netherlands
| | - Julian Langer
- Max Planck Institute of
Biophysics, D-60438 Frankfurt am Main, Germany
| | - Lucia Banci
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
- Consorzio
Interuniversitario Risonanze Magnetiche Metalloproteine, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Francesca Cantini
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
- Consorzio
Interuniversitario Risonanze Magnetiche Metalloproteine, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Marco Fragai
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
- Consorzio
Interuniversitario Risonanze Magnetiche Metalloproteine, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Deborah Grifagni
- Magnetic
Resonance Center and Department of Chemistry, University of Florence, Via L. Sacconi 6, 50019 Sesto Fiorentino, Italy
| | - Tatjana Barthel
- Macromolecular
Crystallography, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, D-12489 Berlin, Germany
| | - Jan Wollenhaupt
- Macromolecular
Crystallography, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, D-12489 Berlin, Germany
| | - Manfred S. Weiss
- Macromolecular
Crystallography, Helmholtz-Zentrum Berlin, Albert-Einstein-Str. 15, D-12489 Berlin, Germany
| | | | - Adriaan Bax
- NIH, LCP NIDDK, Bethesda, Maryland 20892, United States
| | - Sridhar Sreeramulu
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| | - Harald Schwalbe
- Institute
for Organic Chemistry and Chemical Biology, Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
- Center
of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt am Main, D-60438 Frankfurt, Germany
| |
Collapse
|
233
|
Halfmann PJ, Loeffler K, Duffy A, Kuroda M, Yang JE, Wright ER, Kawaoka Y, Kane RS. Broad protection against clade 1 sarbecoviruses after a single immunization with cocktail spike-protein-nanoparticle vaccine. Nat Commun 2024; 15:1284. [PMID: 38346966 PMCID: PMC10861510 DOI: 10.1038/s41467-024-45495-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/25/2024] [Indexed: 02/15/2024] Open
Abstract
The 2002 SARS outbreak, the 2019 emergence of COVID-19, and the continuing evolution of immune-evading SARS-CoV-2 variants together highlight the need for a broadly protective vaccine against ACE2-utilizing sarbecoviruses. While updated variant-matched formulations are a step in the right direction, protection needs to extend beyond SARS-CoV-2 and its variants to include SARS-like viruses. Here, we introduce bivalent and trivalent vaccine formulations using our spike protein nanoparticle platform that completely protect female hamsters against BA.5 and XBB.1 challenges with no detectable virus in the lungs. The trivalent cocktails elicit highly neutralizing responses against all tested Omicron variants and the bat sarbecoviruses SHC014 and WIV1. Finally, our 614D/SHC014/XBB trivalent spike formulation completely protects human ACE2-transgenic female hamsters against challenges with WIV1 and SHC014 with no detectable virus in the lungs. Collectively, these results illustrate that our trivalent protein-nanoparticle cocktail can provide broad protection against SARS-CoV-2-like and SARS-CoV-1-like sarbecoviruses.
Collapse
Affiliation(s)
- Peter J Halfmann
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Kathryn Loeffler
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Augustine Duffy
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Makoto Kuroda
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA
| | - Jie E Yang
- Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biochemistry, Cryo-EM Research Center, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biochemistry, Midwest Center for Cryo-Electron Tomography, University of Wisconsin, Madison, WI, 53706, USA
| | - Elizabeth R Wright
- Department of Biochemistry, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biochemistry, Cryo-EM Research Center, University of Wisconsin, Madison, WI, 53706, USA
- Department of Biochemistry, Midwest Center for Cryo-Electron Tomography, University of Wisconsin, Madison, WI, 53706, USA
| | - Yoshihiro Kawaoka
- Department of Pathobiological Sciences, Influenza Research Institute, School of Veterinary Medicine, University of Wisconsin, Madison, WI, 53711, USA.
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, 108-8639, Japan.
- The Research Center for Global Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, 162-8655, Japan.
- Pandemic Preparedness, Infection and Advanced Research Center (UTOPIA), University of Tokyo, Tokyo, 162-8655, Japan.
| | - Ravi S Kane
- School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
| |
Collapse
|
234
|
Abera BT, Teka H, Berhe E, Gebru MA, Zenebe D, Abraha HE, Hailu A. Assessment of adverse events among healthcare workers following the Janssen COVID-19 vaccine in Tigray, Ethiopia. Sci Rep 2024; 14:3255. [PMID: 38332047 PMCID: PMC10853211 DOI: 10.1038/s41598-024-53561-8] [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: 01/23/2023] [Accepted: 02/01/2024] [Indexed: 02/10/2024] Open
Abstract
Apart from the inequality in vaccination, war zones and areas where communication is disrupted are affected by myths and misconceptions about COVID-19 vaccines, heightening vaccine hesitancy. Local data on adverse events of the vaccines and their mildness can increase confidence and acceptance of the vaccines in the respective population. In areas of conflict and communication blackouts, the perception of the vaccines by health workers is of paramount importance as public health recommendations may not reach the public. Therefore, the scientific evaluation of adverse events following COVID-19 vaccination in such areas is invaluable. This cross-sectional, facility-based study was conducted using a structured, interviewer-administered questionnaire to assess the adverse events experienced by healthcare workers who received the Janssen COVID-19 vaccine. The sample was divided proportionally to the number of vaccinated healthcare workers for the different healthcare professions, and participants were then randomly selected from each profession. Prior to data collection, a pilot test was conducted with 5% of the sample size outside the selected hospital. The study was conducted using a structured questionnaire completed by an interviewer to assess adverse events in 442 healthcare workers who had received the Janssen COVID-19 vaccine between July 11 and 25, 2022. The study period was from August 15 to September 15, 2022. A significant number of healthcare workers [366 (83.3%); 95% CI 79.5%, 86.5%] experienced at least one adverse event. Nearly 90% of participants reported that the adverse events were mild to moderate. Pain at the injection site [307 (69.5%); 95% CI 65.0%, 73.6%] and headache [247 (55.9%); 95% CI 51.2%, 60.4%] were the most common local and systemic adverse events, respectively. Two HCWs experienced anaphylactic reaction. Younger age was significantly associated with the occurrence of adverse events. We deciphered that the adverse events reported by the study participants were not different from the typically occurring vaccine-related adverse reactions, and therefore concluded that post-vaccination reactions in healthcare workers were minor. Although vaccination in Tigray is currently stalled due to the siege, responsible stakeholders should develop a mechanism to track population-wide adverse events once the vaccines start to rollout.
Collapse
Affiliation(s)
- Bisrat Tesfay Abera
- Department of Internal Medicine, School of Medicine, Mekelle University, Mekelle, Tigray, Ethiopia.
| | - Hale Teka
- Department of Obstetrics and Gynecology, School of Medicine, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Ephrem Berhe
- Department of Internal Medicine, School of Medicine, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Marta Abrha Gebru
- Department of Internal Medicine, School of Medicine, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Dawit Zenebe
- Department of Epidemiology, School of Public Health, Mekelle University, Mekelle, Tigray, Ethiopia
| | - Hiluf Ebuy Abraha
- Department of Biostatistics, School of Public Health, Mekelle University, Mekelle, Tigray, Ethiopia
- Arnold School of Public Health, University of South Carolina, Columbia, USA
| | - Abraha Hailu
- Department of Internal Medicine, School of Medicine, Mekelle University, Mekelle, Tigray, Ethiopia
| |
Collapse
|
235
|
Peng ZY, Yang S, Lu HZ, Wang LM, Li N, Zhang HT, Xing SY, Du YN, Deng SQ. A review on Zika vaccine development. Pathog Dis 2024; 82:ftad036. [PMID: 38192053 PMCID: PMC10901608 DOI: 10.1093/femspd/ftad036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 12/15/2023] [Accepted: 01/05/2024] [Indexed: 01/10/2024] Open
Abstract
Zika virus (ZIKV), which belongs to the Flavivirus family, is mainly transmitted via the bite of Aedes mosquitoes. In newborns, ZIKV infection can cause severe symptoms such as microcephaly, while in adults, it can lead to Guillain‒Barré syndrome (GBS). Due to the lack of specific therapeutic methods against ZIKV, the development of a safe and effective vaccine is extremely important. Several potential ZIKV vaccines, such as live attenuated, inactivated, nucleic acid, viral vector, and recombinant subunit vaccines, have demonstrated promising outcomes in clinical trials involving human participants. Therefore, in this review, the recent developmental progress, advantages and disadvantages of these five vaccine types are examined, and practical recommendations for future development are provided.
Collapse
Affiliation(s)
- Zhe-Yu Peng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Song Yang
- Institute of Agro-products Processing, Anhui Academy of Agricultural Sciences, Hefei 230031, Anhui, China
| | - Hong-Zheng Lu
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Lin-Min Wang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Ni Li
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hai-Ting Zhang
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Si-Yu Xing
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Yi-Nan Du
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Sheng-Qun Deng
- The Key Laboratory of Microbiology and Parasitology of Anhui Province, the Key Laboratory of Zoon-oses of High Institutions in Anhui, Department of Pathogen Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
236
|
Samanta S, Banerjee J, Das A, Das S, Ahmed R, Das S, Pal A, Ali KM, Mukhopadhyay R, Giri B, Dash SK. Enhancing Immunological Memory: Unveiling Booster Doses to Bolster Vaccine Efficacy Against Evolving SARS-CoV-2 Mutant Variants. Curr Microbiol 2024; 81:91. [PMID: 38311669 DOI: 10.1007/s00284-023-03597-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 12/19/2023] [Indexed: 02/06/2024]
Abstract
A growing number of re-infections with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in previously immunized individuals has sparked discussions about the potential need for a booster vaccine dosage to counteract declining antibody levels and new strains. The protective immunity produced by vaccinations, and past illnesses relies on immunological memory. CD4 + T cells, CD8 + T cells, B cells, and long-lasting antibody responses are all components of the adaptive immune system that can generate and maintain this immunological memory. Since novel mutant variants have emerged one after the other, the world has been hit by repeated waves. Various vaccine formulations against SARS-CoV-2 have been administered across the globe. Thus, estimating the efficacy of those vaccines against gradually developed mutant stains is the essential parameter regarding the fate of those vaccine formulations and the necessity of booster doses and their frequency. In this review, focus has also been given to how vaccination stacks up against moderate and severe acute infections in terms of the longevity of the immune cells, neutralizing antibody responses, etc. However, hybrid immunity shows a greater accuracy of re-infection of variants of concern (VOCs) of SARS-CoV-2 than infection and immunization. The review conveys knowledge of detailed information about several marketed vaccines and the status of their efficacy against specific mutant strains of SARS-CoV-2. Furthermore, this review discusses the status of immunological memory after infection, mixed infection, and vaccination.
Collapse
Affiliation(s)
- Sovan Samanta
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Jhimli Banerjee
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Aparna Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sourav Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Rubai Ahmed
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Swarnali Das
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Amitava Pal
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Kazi Monjur Ali
- Department of Nutrition, Maharajadhiraj Uday Chand Women's College, B.C. Road, Bardhaman, 713104, West Bengal, India
| | - Rupanjan Mukhopadhyay
- Department of Physiology, City College, 102/1, Raja Rammohan Sarani, Kolkata, 700009, West Bengal, India
| | - Biplab Giri
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India
| | - Sandeep Kumar Dash
- Department of Physiology, University of Gour Banga, Malda, 732103, West Bengal, India.
| |
Collapse
|
237
|
Fischer C, Willscher E, Paschold L, Gottschick C, Klee B, Diexer S, Bosurgi L, Dutzmann J, Sedding D, Frese T, Girndt M, Hoell JI, Gekle M, Addo MM, Schulze Zur Wiesch J, Mikolajczyk R, Binder M, Schultheiß C. SARS-CoV-2 vaccination may mitigate dysregulation of IL-1/IL-18 and gastrointestinal symptoms of the post-COVID-19 condition. NPJ Vaccines 2024; 9:23. [PMID: 38316833 PMCID: PMC10844289 DOI: 10.1038/s41541-024-00815-1] [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: 09/07/2023] [Accepted: 01/24/2024] [Indexed: 02/07/2024] Open
Abstract
The rapid development of safe and effective vaccines helped to prevent severe disease courses after SARS-CoV-2 infection and to mitigate the progression of the COVID-19 pandemic. While there is evidence that vaccination may reduce the risk of developing post-COVID-19 conditions (PCC), this effect may depend on the viral variant. Therapeutic effects of post-infection vaccination have been discussed but the data for individuals with PCC remains inconclusive. In addition, extremely rare side effects after SARS-CoV-2 vaccination may resemble the heterogeneous PCC phenotype. Here, we analyze the plasma levels of 25 cytokines and SARS-CoV-2 directed antibodies in 540 individuals with or without PCC relative to one or two mRNA-based COVID-19 vaccinations as well as in 20 uninfected individuals one month after their initial mRNA-based COVID-19 vaccination. While none of the SARS-CoV-2 naïve individuals reported any persisting sequelae or exhibited PCC-like dysregulation of plasma cytokines, we detected lower levels of IL-1β and IL-18 in patients with ongoing PCC who received one or two vaccinations at a median of six months after infection as compared to unvaccinated PCC patients. This reduction correlated with less frequent reporting of persisting gastrointestinal symptoms. These data suggest that post-infection vaccination in patients with PCC might be beneficial in a subgroup of individuals displaying gastrointestinal symptoms.
Collapse
Affiliation(s)
- Claudia Fischer
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland
| | - Edith Willscher
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Lisa Paschold
- Department of Internal Medicine IV, Oncology/Hematology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Cornelia Gottschick
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Bianca Klee
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Sophie Diexer
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jochen Dutzmann
- Mid-German Heart Center, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Cardiology and Intensive Care Medicine, University Hospital, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Thomas Frese
- Institute of General Practice and Family Medicine, Martin-Luther-University Halle-Wittenberg, Halle, (Saale), Germany
| | - Matthias Girndt
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Jessica I Hoell
- Pediatric Hematology and Oncology, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Michael Gekle
- Julius Bernstein-Institute of Physiology, Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Marylyn M Addo
- I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Braunschweig, Germany
- University Medical Center Hamburg-Eppendorf, Institute for Infection Research and Vaccine Development (IIRVD), Hamburg, Germany
| | | | - Rafael Mikolajczyk
- Institute for Medical Epidemiology, Biometrics, and Informatics (IMEBI), Interdisciplinary Center for Health Sciences, Medical School of the Martin Luther University Halle-Wittenberg, Halle, (Saale), Germany
| | - Mascha Binder
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland
| | - Christoph Schultheiß
- Division of Medical Oncology, University Hospital Basel, Basel, Switzerland.
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University, and University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
238
|
Tandler C, Heitmann JS, Michel TM, Marconato M, Jaeger SU, Tegeler CM, Denk M, Richter M, Oezbek MT, Maringer Y, Schroeder SM, Schneiderhan-Marra N, Wiesmüller KH, Bitzer M, Ruetalo N, Schindler M, Meisner C, Fischer I, Rammensee HG, Salih HR, Walz JS. Long-term efficacy of the peptide-based COVID-19 T cell activator CoVac-1 in healthy adults. Int J Infect Dis 2024; 139:69-77. [PMID: 38016500 DOI: 10.1016/j.ijid.2023.11.009] [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: 08/28/2023] [Revised: 11/04/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
OBJECTIVES T cell immunity is key for the control of viral infections including SARS-CoV-2, in particular with regard to immune memory and protection against arising genetic variants. METHODS We recently evaluated a peptide-based SARS-CoV-2 T cell activator termed CoVac-1 in a first-in-human trial in healthy adults. Here, we report on long-term safety and efficacy data of CoVac-1 until month 12. RESULTS CoVac-1 is well tolerated without long-term immune-related side effects and induces long-lasting anti-viral T cell responses in 100% of study participants, with potent expandability of clusters of differentiation (CD4+) and CD8+ T cells targeting multiple different CoVac-1 T cell epitopes. T cell responses were associated with stronger injection site reaction. Beyond induction of T cell immunity, 89% of subjects developed CoVac-1-specific immunoglobulin G antibodies which associated with the intensity of the T cell response, indicating that CoVac-1-specific CD4+ T cells support the induction of B-cell responses. Vaccination with approved COVID-19 vaccines boosted CoVac-1-specific T cell responses. Overall, a low SARS-CoV-2 infection rate (8.3%) was observed. CONCLUSION Together, a single application of CoVac-1 elicits long-lived and broad SARS-CoV-2-specific T cell immunity, which further supports the current evaluation of our T cell activator in patients with congenital or acquired B-cell defects.
Collapse
Affiliation(s)
- Claudia Tandler
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Jonas S Heitmann
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Tanja M Michel
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Maddalena Marconato
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Simon U Jaeger
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Stuttgart, Germany; Department of Clinical Pharmacology, University Hospital Tübingen, Tübingen, Germany
| | - Christian M Tegeler
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany; Department of Obstetrics and Gynecology, University Hospital Tübingen, Tübingen, Germany
| | - Monika Denk
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Marion Richter
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Melek Tutku Oezbek
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Yacine Maringer
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany
| | - Sarah M Schroeder
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Department of Otorhinolaryngology, Head and Neck Surgery, University of Hospital Tübingen, Tübingen, Germany
| | | | | | - Michael Bitzer
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Natalia Ruetalo
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Michael Schindler
- Institute for Medical Virology and Epidemiology of Viral Diseases, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Meisner
- Robert Bosch Hospital, Robert Bosch Society for Medical Research, Stuttgart, Germany
| | - Imma Fischer
- Institute for Clinical Epidemiology and Applied Biometry, University Hospital Tübingen, Tübingen, Germany
| | - Hans-Georg Rammensee
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), partner site Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Juliane S Walz
- Department of Peptide-based Immunotherapy, Institute of Immunology, University and University Hospital Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC2180) "Image-Guided and Functionally Instructed Tumor Therapies", University of Tübingen, Tübingen, Germany; Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany.
| |
Collapse
|
239
|
Pastor Pueyo P, Gambó Ruberte E, Gayán Ordás J, Matute Blanco L, Pascual Figal D, Larrañaga Moreira JM, Gómez Barrado JJ, González Calle D, Almenar Bonet L, Alonso Salinas GL, Corbí Pascual MJ, Plaza Martín M, Pons Llinares J, Durante López A, Barreiro Pérez M, Candanedo Ocaña F, Bautista García J, Merchán Ortega G, Domínguez Rodríguez F, Martínez Mateo V, Campreciós Crespo M, Quintás Guzmán M, Jordán Martínez L, Aboal Viñas J, Rodríguez López J, Fernández Santos S, Revilla Martí P, Álvarez Roy L, Gómez Polo JC, García Pinilla JM, Ferré Vallverdú M, García Bueno L, Soriano Colomé T, Worner Diz F. Vaccine-carditis study: Spanish multicenter registry of inflammatory heart disease after COVID-19 vaccination. Clin Res Cardiol 2024; 113:223-234. [PMID: 37368015 DOI: 10.1007/s00392-023-02225-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/08/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION AND OBJECTIVES Vaccines against SARS-CoV-2 have been a major scientific and medical achievement in the control of the COVID-19 pandemic. However, very infrequent cases of inflammatory heart disease have been described as adverse events, leading to uncertainty in the scientific community and in the general population. METHODS The Vaccine-Carditis Registry has included all cases of myocarditis and pericarditis diagnosed within 30 days after COVID-19 vaccination since August 1, 2021 in 29 centers throughout the Spanish territory. The definitions of myocarditis (probable or confirmed) and pericarditis followed the consensus of the Centers for Disease Control and the Clinical Practice Guidelines of the European Society of Cardiology. A comprehensive analysis of clinical characteristics and 3-month evolution is presented. RESULTS From August 1, 2021, to March 10, 2022, 139 cases of myocarditis or pericarditis were recorded (81.3% male, median age 28 years). Most cases were detected in the 1st week after administration of an mRNA vaccine, the majority after the second dose. The most common presentation was mixed inflammatory disease (myocarditis and pericarditis). 11% had left ventricular systolic dysfunction, 4% had right ventricular systolic dysfunction, and 21% had pericardial effusion. In cardiac magnetic resonance studies, left ventricular inferolateral involvement was the most frequent pattern (58%). More than 90% of cases had a benign clinical course. After a 3-month follow-up, the incidence of adverse events was 12.78% (1.44% mortality). CONCLUSIONS In our setting, inflammatory heart disease after vaccination against SARS-CoV-2 predominantly affects young men in the 1st week after the second dose of RNA-m vaccine and presents a favorable clinical course in most cases.
Collapse
Affiliation(s)
- Pablo Pastor Pueyo
- Cardiology Department, Hospital Universitario Arnau de Vilanova, Lleida, Institut de Reserça Biomèdica (IRB) Lleida, Lleida, Spain.
| | - Elena Gambó Ruberte
- Cardiology Department, Hospital Universitario Arnau de Vilanova, Lleida, Institut de Reserça Biomèdica (IRB) Lleida, Lleida, Spain
| | - Jara Gayán Ordás
- Cardiology Department, Hospital Universitario Arnau de Vilanova, Lleida, Institut de Reserça Biomèdica (IRB) Lleida, Lleida, Spain
| | - Lucía Matute Blanco
- Cardiology Department, Hospital Universitario Arnau de Vilanova, Lleida, Institut de Reserça Biomèdica (IRB) Lleida, Lleida, Spain
| | - Domingo Pascual Figal
- Cardiology Department, Hospital Clínico Universitario Virgen de La Arrixaca, Murcia, Spain
| | | | | | - David González Calle
- Cardiology Department, Complejo Asistencial Universitario de Salamanca, Salamanca, Spain
| | | | | | | | - María Plaza Martín
- Cardiology Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | | | | | | | - Javier Bautista García
- Cardiology Department, Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Spain
| | | | | | - Virgilio Martínez Mateo
- Cardiology Department, Complejo Hospitalario La Mancha Centro, Alcázar de San Juan, Ciudad Real, Spain
| | | | - Martín Quintás Guzmán
- Cardiology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | | | - Jaime Aboal Viñas
- Cardiology Department, Hospital Universitario Dr. Josep Trueta, Girona, Spain
| | | | | | - Pablo Revilla Martí
- Cardiology Department, Hospital Clínico Universitario Lozano Blesa, Saragossa, Spain
| | - Laura Álvarez Roy
- Cardiology Department, Hospital Universitario Miguel Servet, Saragossa, Spain
| | | | | | - María Ferré Vallverdú
- Cardiology Department, Servicio de Cardiología, Hospital Sant Joan, Reus, Tarragona, Spain
| | | | - Toni Soriano Colomé
- Cardiology Department, Hospital Universitario Vall d´Hebron, Barcelona, Spain
| | - Fernando Worner Diz
- Cardiology Department, Hospital Universitario Arnau de Vilanova, Lleida, Institut de Reserça Biomèdica (IRB) Lleida, Lleida, Spain
| |
Collapse
|
240
|
Ahmed N, Athavale A, Tripathi AH, Subramaniam A, Upadhyay SK, Pandey AK, Rai RC, Awasthi A. To be remembered: B cell memory response against SARS-CoV-2 and its variants in vaccinated and unvaccinated individuals. Scand J Immunol 2024; 99:e13345. [PMID: 38441373 DOI: 10.1111/sji.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 03/07/2024]
Abstract
COVID-19 disease has plagued the world economy and affected the overall well-being and life of most of the people. Natural infection as well as vaccination leads to the development of an immune response against the pathogen. This involves the production of antibodies, which can neutralize the virus during future challenges. In addition, the development of cellular immune memory with memory B and T cells provides long-lasting protection. The longevity of the immune response has been a subject of intensive research in this field. The extent of immunity conferred by different forms of vaccination or natural infections remained debatable for long. Hence, understanding the effectiveness of these responses among different groups of people can assist government organizations in making informed policy decisions. In this article, based on the publicly available data, we have reviewed the memory response generated by some of the vaccines against SARS-CoV-2 and its variants, particularly B cell memory in different groups of individuals.
Collapse
Affiliation(s)
- Nafees Ahmed
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Atharv Athavale
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ankita H Tripathi
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Adarsh Subramaniam
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Santosh K Upadhyay
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | | | - Ramesh Chandra Rai
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
241
|
Artarini A, Hadianti T, Giri-Rachman EA, Tan MI, Safitri IA, Hidayat NA, Retnoningrum DS, Natalia D. Development of Adenovirus-Based Covid-19 Vaccine Candidate in Indonesia. Mol Biotechnol 2024; 66:222-232. [PMID: 37076664 PMCID: PMC10115376 DOI: 10.1007/s12033-023-00749-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 04/05/2023] [Indexed: 04/21/2023]
Abstract
Covid-19 pandemic has struck worldwide by end of 2019 and the use of various vaccine platforms was one of the main strategies to end this. To meet the needs for vaccine technology equality among many countries, we developed adenovirus-based Covid-19 vaccine candidate in Indonesia. SARS-CoV-2 Spike gene (S) was constructed into pAdEasy vector. The recombinant serotype 5 Adenovirus (AdV_S) genome was transfected into AD293 cells to produce recombinant adenovirus. Characterization using PCR confirmed the presence of spike gene. Transgene expression analysis showed the expression of S protein in AdV_S infected AD293 and A549 cells. Optimization of viral production showed the highest titer was obtained at MOI of 0.1 and 1 at 4 days. The in vivo study was performed by injecting Balb/c mice with 3.5 × 107 ifu of purified adenovirus. The result showed that S1-specific IgG was increased up to 56 days after single-dose administration of AdV_S. Interestingly, significant increase of S1 glycoprotein-specific IFN-γ ELISpot was observed in AdV_S treated Balb/c mice. In conclusion, the AdV_S vaccine candidate was successfully produced at laboratory scale, immunogenic, and did not cause severe inflammation in Balb/c mice. This study serves as initial step towards manufacturing of adenovirus-based vaccine in Indonesia.
Collapse
Affiliation(s)
- Anita Artarini
- School of Pharmacy, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia.
| | - Tia Hadianti
- School of Pharmacy, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| | | | - Marselina Irasonia Tan
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| | - Intan A Safitri
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| | - Nurhamidah A Hidayat
- School of Life Sciences and Technology, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| | - Debbie S Retnoningrum
- School of Pharmacy, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| | - Dessy Natalia
- Faculty of Mathematics and Natural Sciences, Institut Teknologi Bandung, Jln. Ganesha 10, Bandung, 40132, Indonesia
| |
Collapse
|
242
|
Cankat S, Demael MU, Swadling L. In search of a pan-coronavirus vaccine: next-generation vaccine design and immune mechanisms. Cell Mol Immunol 2024; 21:103-118. [PMID: 38148330 PMCID: PMC10805787 DOI: 10.1038/s41423-023-01116-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/21/2023] [Indexed: 12/28/2023] Open
Abstract
Members of the coronaviridae family are endemic to human populations and have caused several epidemics and pandemics in recent history. In this review, we will discuss the feasibility of and progress toward the ultimate goal of creating a pan-coronavirus vaccine that can protect against infection and disease by all members of the coronavirus family. We will detail the unmet clinical need associated with the continued transmission of SARS-CoV-2, MERS-CoV and the four seasonal coronaviruses (HCoV-OC43, NL63, HKU1 and 229E) in humans and the potential for future zoonotic coronaviruses. We will highlight how first-generation SARS-CoV-2 vaccines and natural history studies have greatly increased our understanding of effective antiviral immunity to coronaviruses and have informed next-generation vaccine design. We will then consider the ideal properties of a pan-coronavirus vaccine and propose a blueprint for the type of immunity that may offer cross-protection. Finally, we will describe a subset of the diverse technologies and novel approaches being pursued with the goal of developing broadly or universally protective vaccines for coronaviruses.
Collapse
Affiliation(s)
- S Cankat
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - M U Demael
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK
| | - L Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London, NW3 2PP, UK.
| |
Collapse
|
243
|
Yu R, Zhang L, Zhou P, Zhang Z, Liu X, Wang Y, Guo H, Pan L, Liu X. Evaluation of the immunoprotective effects of porcine deltacoronavirus subunit vaccines. Virology 2024; 590:109955. [PMID: 38070302 DOI: 10.1016/j.virol.2023.109955] [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: 08/29/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024]
Abstract
Porcine deltacoronavirus (PDCoV), a new porcine enteric coronavirus, has seriously endangered the pig breeding industry and caused great economic losses. However, a PDCoV vaccine is not commercially available. Therefore, new and efficient PDCoV vaccines must be developed without delay. In this study, we used the ExpiCHO eukaryotic expression system to express and purify the following 3 structural proteins of PDCoV: S, N and M. Subsequently, the level of humoral and cellular immunity induced by the S protein (immunization with the S protein alone) and a protein mixture (immunization with a mixture of S, N and M proteins) were evaluated in mice and piglets, respectively, and the performances of the 2 immunizations in a challenge protection test were assessed in piglets. The results showed that both the S protein and the protein mixture induced the production of high levels of specific IgG antibodies and neutralizing antibodies and effectively neutralized PDCoV-infected LLC-PK cells in vitro. Furthermore, compared with the S protein, the N and M proteins in the protein mixture promoted the expression of CD8+ T cells and IFN-γ, induced a stronger cellular immune response, and effectively protected 4/5 of the piglets from PDCoV infection. In conclusion, the results of this study showed that the N and M proteins play important roles in inducing an immunoprotective response. Using N and M antigens as effective antigenic components in the development of PDCoV vaccines in the future will effectively increase the immune efficacy of the vaccines.
Collapse
Affiliation(s)
- Ruiming Yu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; National Center of Technology Innovation for Pigs, China
| | - Liping Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; National Center of Technology Innovation for Pigs, China
| | - Peng Zhou
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhongwang Zhang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiaoqing Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yonglu Wang
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Huichen Guo
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Li Pan
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China.
| | - Xinsheng Liu
- State Key Laboratory for Animal Disease Control and Prevention, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China; National Center of Technology Innovation for Pigs, China.
| |
Collapse
|
244
|
Le Bert N, Samandari T. Silent battles: immune responses in asymptomatic SARS-CoV-2 infection. Cell Mol Immunol 2024; 21:159-170. [PMID: 38221577 PMCID: PMC10805869 DOI: 10.1038/s41423-024-01127-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 01/03/2024] [Accepted: 01/03/2024] [Indexed: 01/16/2024] Open
Abstract
SARS-CoV-2 infections manifest with a broad spectrum of presentations, ranging from asymptomatic infections to severe pneumonia and fatal outcomes. This review centers on asymptomatic infections, a widely reported phenomenon that has substantially contributed to the rapid spread of the pandemic. In such asymptomatic infections, we focus on the role of innate, humoral, and cellular immunity. Notably, asymptomatic infections are characterized by an early and robust innate immune response, particularly a swift type 1 IFN reaction, alongside a rapid and broad induction of SARS-CoV-2-specific T cells. Often, antibody levels tend to be lower or undetectable after asymptomatic infections, suggesting that the rapid control of viral replication by innate and cellular responses might impede the full triggering of humoral immunity. Even if antibody levels are present in the early convalescent phase, they wane rapidly below serological detection limits, particularly following asymptomatic infection. Consequently, prevalence studies reliant solely on serological assays likely underestimate the extent of community exposure to the virus.
Collapse
Affiliation(s)
- Nina Le Bert
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore.
| | - Taraz Samandari
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
245
|
Schell TL, Caldera F. A Practical Update on COVID-19 and Inflammatory Bowel Disease: COVID-19 Disease Risk and Vaccine Safety and Efficacy. Gastroenterol Hepatol (N Y) 2024; 20:88-97. [PMID: 38414911 PMCID: PMC10895915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
The COVID-19 pandemic introduced significant uncertainty regarding the care of patients with inflammatory bowel disease (IBD). Substantial research efforts have made progress in answering many of the questions that arose, but the constantly shifting paradigm of COVID-19-related research and recommendations has made it challenging for IBD clinicians to remain up-to-date. The goal of this article is to provide a concise and practical summary of the literature evaluating COVID-19 disease risk in addition to COVID-19 vaccine safety, immunogenicity, real-world effectiveness, and uptake among patients with IBD.
Collapse
Affiliation(s)
- Trevor L. Schell
- Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Freddy Caldera
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
246
|
McMahan K, Wegmann F, Aid M, Sciacca M, Liu J, Hachmann NP, Miller J, Jacob-Dolan C, Powers O, Hope D, Wu C, Pereira J, Murdza T, Mazurek CR, Hoyt A, Boon ACM, Davis-Gardner M, Suthar MS, Martinot AJ, Boursiquot M, Cook A, Pessaint L, Lewis MG, Andersen H, Tolboom J, Serroyen J, Solforosi L, Costes LMM, Zahn RC, Barouch DH. Mucosal boosting enhances vaccine protection against SARS-CoV-2 in macaques. Nature 2024; 626:385-391. [PMID: 38096903 PMCID: PMC10849944 DOI: 10.1038/s41586-023-06951-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/07/2023] [Indexed: 01/26/2024]
Abstract
A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.
Collapse
Affiliation(s)
- Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Frank Wegmann
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michaela Sciacca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicole P Hachmann
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jessica Miller
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Olivia Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Cindy Wu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Juliana Pereira
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tetyana Murdza
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Camille R Mazurek
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Amelia Hoyt
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | - Amanda J Martinot
- Tufts University Cummings School of Veterinary Medicine, Grafton, MA, USA
| | | | | | | | | | | | | | - Jan Serroyen
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | | | - Roland C Zahn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
247
|
Gengiah TN, Naidoo DC, Maduma N, Govender S, Dhindayal S, Lewis L. COVID-19 vaccine hesitancy in KwaZulu-Natal, South Africa: A survey of unvaccinated adults. Health SA 2024; 29:2468. [PMID: 38445041 PMCID: PMC10913108 DOI: 10.4102/hsag.v29i0.2468] [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: 08/22/2023] [Accepted: 11/29/2023] [Indexed: 03/07/2024] Open
Abstract
Background Concerns and misconceptions surrounding coronavirus disease 2019 (COVID-19) vaccines may account for vaccine hesitancy and low uptake. Aim To determine prevalence of COVID-19 vaccine hesitancy, vaccine-related misconceptions, and predictors of vaccine hesitancy among South Africans. Setting Community setting in five districts in KwaZulu- Natal province. Methods Between August 20, 2021, and September 27, 2021, we conducted a cross-sectional survey, interviewing 300 unvaccinated adults amid the national vaccination campaign. Predictors of hesitancy were identified through multivariable logistic regression analysis. Results Participants had a median age of 29 years (IQR: 23-39), 86.7% were Black African, 63.2% were male, 53.3% resided in rural communities, and 59.3% (95% CI: 53.8% - 64.9%) were classified as vaccine hesitant. The primary reason for not vaccinating was a lack of trust in the vaccine (62.1%). Factors associated with reduced vaccine hesitancy included age (participants aged 35-49 years: OR: 0.28, 95% CI: 0.18-0.64, p = 0.003; participants over 50 years: OR: 0.18, 95% CI: 0.07-0.47, p = 0.0004), previous COVID-19 infection (OR: 0.31, 95% CI: 0.11-0.87, p = 0.03), and receiving vaccine information from healthcare workers (OR: 0.32, 95% CI: 0.10-1.0, p = 0.05). Unemployed (OR: 2.14, 95% CI: 1.1-4.2, p = 0.03) and self-employed individuals (OR: 2.98, 95% CI: 1.27-7.02, p = 0.01) were more likely to be vaccine hesitant. Conclusion COVID-19 vaccine hesitancy rates are high in KwaZulu-Natal. Uptake could be enhanced by healthcare workers leading information campaigns with messages targeting younger individuals, the unemployed, and the self-employed. Contribution This survey provides evidence to improve COVID-19 vaccination uptake in South Africa.
Collapse
Affiliation(s)
- Tanuja N Gengiah
- Center for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Donavan C Naidoo
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nomcebo Maduma
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Saien Govender
- Center for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Sherishka Dhindayal
- Center for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| | - Lara Lewis
- Center for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa
| |
Collapse
|
248
|
Lapuente D, Winkler TH, Tenbusch M. B-cell and antibody responses to SARS-CoV-2: infection, vaccination, and hybrid immunity. Cell Mol Immunol 2024; 21:144-158. [PMID: 37945737 PMCID: PMC10805925 DOI: 10.1038/s41423-023-01095-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019 prompted scientific, medical, and biotech communities to investigate infection- and vaccine-induced immune responses in the context of this pathogen. B-cell and antibody responses are at the center of these investigations, as neutralizing antibodies (nAbs) are an important correlate of protection (COP) from infection and the primary target of SARS-CoV-2 vaccine modalities. In addition to absolute levels, nAb longevity, neutralization breadth, immunoglobulin isotype and subtype composition, and presence at mucosal sites have become important topics for scientists and health policy makers. The recent pandemic was and still is a unique setting in which to study de novo and memory B-cell (MBC) and antibody responses in the dynamic interplay of infection- and vaccine-induced immunity. It also provided an opportunity to explore new vaccine platforms, such as mRNA or adenoviral vector vaccines, in unprecedented cohort sizes. Combined with the technological advances of recent years, this situation has provided detailed mechanistic insights into the development of B-cell and antibody responses but also revealed some unexpected findings. In this review, we summarize the key findings of the last 2.5 years regarding infection- and vaccine-induced B-cell immunity, which we believe are of significant value not only in the context of SARS-CoV-2 but also for future vaccination approaches in endemic and pandemic settings.
Collapse
Affiliation(s)
- Dennis Lapuente
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
| | - Thomas H Winkler
- Department of Biology, Division of Genetics, Nikolaus-Fiebiger-Center for Molecular Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany.
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany.
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossgarten 4, 91054, Erlangen, Germany
- Medical Immunology Campus Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Schlossplatz 1, 91054, Erlangen, Germany
| |
Collapse
|
249
|
Nkolola JP, Barouch DH. Prophylactic HIV-1 vaccine trials: past, present, and future. Lancet HIV 2024; 11:e117-e124. [PMID: 38141639 PMCID: PMC11736820 DOI: 10.1016/s2352-3018(23)00264-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/24/2023] [Accepted: 10/11/2023] [Indexed: 12/25/2023]
Abstract
An effective HIV-1 vaccine is a global health priority but has remained elusive for more than 40 years. Key scientific hurdles that have hampered vaccine development are the unprecedented genetic variability of the virus, the rapid establishment of persistent viral latency, and the challenges associated with induction of broadly neutralising antibodies. Clinical trials have been instrumental in evaluating scientific concepts and testing vaccine strategies. This Review discusses lessons learned from clinical trials of HIV-1 vaccines, current technologies that are being explored, and future considerations in the development of a safe and effective HIV-1 vaccine.
Collapse
Affiliation(s)
- Joseph P Nkolola
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
250
|
Goryaynov S, Gurova O. Effect of Platform Type on Clinical Efficacy of SARS-CoV-2 Vaccines in Prime Vaccination Settings: A Systematic Review and Meta-Regression of Randomized Controlled Trials. Vaccines (Basel) 2024; 12:130. [PMID: 38400114 PMCID: PMC10892687 DOI: 10.3390/vaccines12020130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/14/2024] [Accepted: 01/24/2024] [Indexed: 02/25/2024] Open
Abstract
This systematic review investigated the association between platform type and the clinical efficacy of SARS-CoV-2 vaccines using the meta-regression of randomized controlled trials to compare the rates of the first appearance of symptomatic COVID-19 on the platforms. The trial search was conducted using PubMed, ClinicalTrials.gov, and the EU Clinical Trials Register. The main selection criteria included: non-active control, immunocompetent individuals without previous vaccination, and a low risk of bias. The platform effect was summarized with an incidence rate ratio (IRR) and a 95% confidence interval for every platform category against the reference. IRR was obtained by random-effect meta-regression with adjustment for confounding by effect modifiers. The analysis was conducted in per-protocol (PP) and modified intention-to-treat (mITT) sets. Six vaccine types with 35 trials were included. Vector vaccines were a reference category. In the PP set, rates of symptomatic COVID-19 on mRNA and protein subunit vaccines were significantly lower than on the vector: IRR = 0.30 [0.19; 0.46], p = 0.001 and 0.63 [0.46; 0.86], p = 0.012, respectively. There was no difference for inactivated and virus-like particle vaccines compared to the vector: IRR = 0.98 [0.71; 1.36], p = 0.913 and 0.70 [0.41; 1.20], p = 0.197, respectively. The rate of cases on DNA vaccines was significantly higher than that on the vector: IRR = 2.58 [1.17; 5.68], p = 0.034. Results for the mITT set were consistent. Platform type is an effect modifier of the clinical efficacy of SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
| | - Olesya Gurova
- Department of Endocrinology No. 1, N.V. Sklifosovsky Institute of Clinical Medicine, Sechenov First Moscow State Medical University, 119435 Moscow, Russia;
| |
Collapse
|