1
|
Murayama G, Kusaoi M, Horiuchi Y, Tabe Y, Naito T, Ito S, Yamaji K, Tamura N. Effects of the induction of humoral and cellular immunity by third vaccination for SARS-CoV-2. J Infect Chemother 2024; 30:1021-1027. [PMID: 38570139 DOI: 10.1016/j.jiac.2024.03.021] [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: 12/12/2023] [Revised: 03/08/2024] [Accepted: 03/31/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION To control the spread of severe disease caused by mutant strains of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), it is necessary to determine whether continued vaccination enhances humoral and cellular immunity. AIM In this study, we examined the changes in humoral and cellular immunity to SARS-CoV-2 after administration of the third vaccination in Japanese adults who had received the second dose of messenger ribonucleic acid (mRNA)-1273 vaccine and the third vaccination (BNT162b2 or mRNA-1273). METHODS We measured anti-spike antibodies in immunoglobulin G (IgG) and anti-nucleocapsid IgG titers in the serum of the vaccinated subjects. To evaluate cellular immunity, the peripheral blood mononuclear cells of inoculated individuals were cultured with spiked proteins, including those of the SARS-CoV-2 conventional strain and Omicron strain, and then subjected to enzyme-linked immunospot (ELISPOT). RESULTS The results revealed that the anti-SARS-CoV-2 spike protein antibody titer increased after the third vaccination and was maintained; however, a decrease was observed at 6 months after vaccination. SARS-CoV-2 antigen-specific T helper (Th)1 and Th2 cell responses were also induced after the third vaccination and were maintained for 6 months after vaccination. Furthermore, induction of cellular immunity against Omicron strains by the omicron non-compliant vaccines, BNT162b2 or mRNA-1273, was observed. CONCLUSION These findings demonstrate the effectiveness of vaccination against unknown mutant strains that may occur in the future and provide important insights into vaccination strategies.
Collapse
Affiliation(s)
- Goh Murayama
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan.
| | - Makio Kusaoi
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Yoko Tabe
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, Tokyo, 113-8421, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, 113-8421, Japan
| | - Suminobu Ito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, 113-8421, Japan; Medical Technology Innovation Centre, Juntendo University, Tokyo, 113-8421, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, 113-8421, Japan
| |
Collapse
|
2
|
Díaz-Dinamarca DA, Cárdenas-Cáceres S, Muena NA, Díaz P, Barra G, Puentes R, Escobar DF, Díaz-Samirin M, Santis-Alay NT, Canales C, Díaz J, García-Escorza HE, Grifoni A, Sette A, Tischler ND, Vasquez AE. Booster Vaccination with BNT162b2 Improves Cellular and Humoral Immune Response in the Pediatric Population Immunized with CoronaVac. Vaccines (Basel) 2024; 12:919. [PMID: 39204043 PMCID: PMC11359105 DOI: 10.3390/vaccines12080919] [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: 02/08/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 09/03/2024] Open
Abstract
The SARS-CoV-2 Omicron variant and its sublineages continue to cause COVID-19-associated pediatric hospitalizations, severe disease, and death globally. BNT162b2 and CoronaVac are the main vaccines used in Chile. Much less is known about the Wuhan-Hu-1 strain-based vaccines in the pediatric population compared to adults. Given the worldwide need for booster vaccinations to stimulate the immune response against new Omicron variants of SARS-CoV-2, we characterized the humoral and cellular immune response against Omicron variant BA.1 in a pediatric cohort aged 10 to 16 years who received heterologous vaccination based on two doses of CoronaVac, two doses of CoronaVac (2x) plus one booster dose of BNT162b2 [CoronaVac(2x) + BNT162b2 (1x)], two doses of CoronaVac plus two booster doses of BNT162b2 [CoronaVac(2x) + BNT162b2 (2x)], and three doses of BNT162b2. We observed that the [CoronaVac(2x) + BNT162b2 (2x)] vaccination showed higher anti-S1 and neutralizing antibody titers and CD4 and CD8 T cell immunity specific to the Omicron variant compared to immunization with two doses of CoronaVac alone. Furthermore, from all groups tested, immunity against Omicron was highest in individuals who received three doses of BNT162b2. We conclude that booster vaccination with BNT162b2, compared to two doses of CoronaVac alone, induces a greater protective immunity.
Collapse
Affiliation(s)
- Diego A. Díaz-Dinamarca
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Simone Cárdenas-Cáceres
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
| | - Nicolás A. Muena
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
| | - Pablo Díaz
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Gisselle Barra
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Rodrigo Puentes
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Daniel F. Escobar
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Michal Díaz-Samirin
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| | - Natalia T. Santis-Alay
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Cecilia Canales
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | - Janepsy Díaz
- Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (R.P.); (N.T.S.-A.); (C.C.); (J.D.)
| | | | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (A.G.); (A.S.)
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; (A.G.); (A.S.)
- Department of Pathology, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicole D. Tischler
- Laboratorio de Virología Molecular, Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8581151, Chile; (S.C.-C.); (N.A.M.)
- Escuela de Bioquímica, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Abel E. Vasquez
- Subdepartamento Innovación y Desarrollo, Departamento Agencia Nacional de Dispositivos Médicos, Innovación y Desarrollo, Instituto de Salud Pública de Chile, Santiago 7780050, Chile; (D.A.D.-D.); (P.D.); (G.B.); (D.F.E.)
| |
Collapse
|
3
|
Nhu LNT, Chambers M, Chantratita N, Cheah PY, Day NP, Dejnirattisai W, Dunachie SJ, Grifoni A, Hamers RL, Hill J, Jones EY, Klenerman P, Mongkolsapaya J, Screaton G, Sette A, Stuart DI, Tan CW, Thwaites G, Thanh VD, Wang LF, Tan LV. Southeast Asia initiative to combat SARS-CoV-2 variants (SEACOVARIANTS) consortium. Wellcome Open Res 2024; 9:181. [PMID: 39022321 PMCID: PMC11252647 DOI: 10.12688/wellcomeopenres.20742.1] [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] [Accepted: 03/26/2024] [Indexed: 07/20/2024] Open
Abstract
A strong and effective COVID-19 and future pandemic responses rely on global efforts to carry out surveillance of infections and emerging SARS-CoV-2 variants and to act accordingly in real time. Many countries in Southeast Asia lack capacity to determine the potential threat of new variants, or other emerging infections. Funded by Wellcome, the Southeast Asia initiative to combat SARS-CoV-2 variants (SEACOVARIANTS) consortium aims to develop and apply a multidisciplinary research platform in Southeast Asia (SEA) for rapid assessment of the biological significance of SARS-CoV-2 variants, thereby informing coordinated local, regional and global responses to the COVID-19 pandemic. Our proposal is delivered by the Vietnam and Thailand Wellcome Africa Asia Programmes, bringing together a multidisciplinary team in Indonesia, Thailand and Vietnam with partners in Singapore, the UK and the USA. Herein we outline five work packages to deliver strengthened regional scientific capacity that can be rapidly deployed for future outbreak responses.
Collapse
Affiliation(s)
| | - Mary Chambers
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Narisara Chantratita
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phaik Yeong Cheah
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Nicholas P.J. Day
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanwisa Dejnirattisai
- Division of Emerging Infectious Disease, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Susanna J. Dunachie
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Alba Grifoni
- La Jolla Institute for Immunology, San Diego, California, USA
| | - Raph L. Hamers
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Oxford University Clinical Research Unit Indonesia, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
| | - Jennifer Hill
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - E. Yvonne Jones
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Paul Klenerman
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Juthathip Mongkolsapaya
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Gavin Screaton
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | | | - David I. Stuart
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Guy Thwaites
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
| | - Vu Duy Thanh
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Le Van Tan
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
| | - SEACOVARIANTS Consortium
- Oxford University Clinical Research Unit, Ho Chi Minh city, Vietnam
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Department of Microbiology and Immunology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Division of Emerging Infectious Disease, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- La Jolla Institute for Immunology, San Diego, California, USA
- Oxford University Clinical Research Unit Indonesia, Faculty of Medicine Universitas Indonesia, Jakarta, Indonesia
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, England, UK
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, England, UK
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
4
|
Bohmwald K, Diethelm-Varela B, Rodríguez-Guilarte L, Rivera T, Riedel CA, González PA, Kalergis AM. Pathophysiological, immunological, and inflammatory features of long COVID. Front Immunol 2024; 15:1341600. [PMID: 38482000 PMCID: PMC10932978 DOI: 10.3389/fimmu.2024.1341600] [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: 11/20/2023] [Accepted: 02/09/2024] [Indexed: 04/12/2024] Open
Abstract
The COVID-19 pandemic continues to cause severe global disruption, resulting in significant excess mortality, overwhelming healthcare systems, and imposing substantial social and economic burdens on nations. While most of the attention and therapeutic efforts have concentrated on the acute phase of the disease, a notable proportion of survivors experience persistent symptoms post-infection clearance. This diverse set of symptoms, loosely categorized as long COVID, presents a potential additional public health crisis. It is estimated that 1 in 5 COVID-19 survivors exhibit clinical manifestations consistent with long COVID. Despite this prevalence, the mechanisms and pathophysiology of long COVID remain poorly understood. Alarmingly, evidence suggests that a significant proportion of cases within this clinical condition develop debilitating or disabling symptoms. Hence, urgent priority should be given to further studies on this condition to equip global public health systems for its management. This review provides an overview of available information on this emerging clinical condition, focusing on the affected individuals' epidemiology, pathophysiological mechanisms, and immunological and inflammatory profiles.
Collapse
Affiliation(s)
- Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Thomas Rivera
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy. Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
5
|
Amaral LDF, Lana RM, Bastos LS. Was the COVID-19 epidemic synchronous in space? An analysis in the health regions of the Rio de Janeiro state, 2020-2022. REVISTA BRASILEIRA DE EPIDEMIOLOGIA 2024; 27:e240010. [PMID: 38422234 PMCID: PMC10896236 DOI: 10.1590/1980-549720240010] [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: 09/01/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024] Open
Abstract
OBJECTIVE To analyze the spatio-temporal dynamics of COVID-19 in the Rio de Janeiro state within the nine health regions, between March 2020 and December 2022. METHODS The Poisson model with random effects was used to smooth and estimate the incidence of COVID-19 hospitalizations reported in the Influenza Epidemiological Surveillance Information System (SIVEP-Gripe) to verify the synchronicity of the epidemic in the state. RESULTS The COVID-19 epidemic in the state is characterized by the presence of seven peaks during the analyzed period corresponding to seven found. An asynchrony in hospitalizations was identified, varying according to the different virus variants in the nine health regions of the state. The incidence peaks of hospitalizations ranged from 1 to 12 cases per 100,000 inhabitants during the pandemic. CONCLUSION This spatio-temporal analysis is applicable to other scenarios, enabling monitoring and decision-making for the control of epidemic diseases in different areas.
Collapse
Affiliation(s)
- Léa de Freitas Amaral
- Fundação Oswaldo Cruz, National School of Public Health - Rio de Janeiro (RJ), Brazil
| | | | | |
Collapse
|
6
|
Binayke A, Zaheer A, Vishwakarma S, Singh S, Sharma P, Chandwaskar R, Gosain M, Raghavan S, Murugesan DR, Kshetrapal P, Thiruvengadam R, Bhatnagar S, Pandey AK, Garg PK, Awasthi A. A quest for universal anti-SARS-CoV-2 T cell assay: systematic review, meta-analysis, and experimental validation. NPJ Vaccines 2024; 9:3. [PMID: 38167915 PMCID: PMC10762233 DOI: 10.1038/s41541-023-00794-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024] Open
Abstract
Measuring SARS-CoV-2-specific T cell responses is crucial to understanding an individual's immunity to COVID-19. However, high inter- and intra-assay variability make it difficult to define T cells as a correlate of protection against COVID-19. To address this, we performed systematic review and meta-analysis of 495 datasets from 94 original articles evaluating SARS-CoV-2-specific T cell responses using three assays - Activation Induced Marker (AIM), Intracellular Cytokine Staining (ICS), and Enzyme-Linked Immunospot (ELISPOT), and defined each assay's quantitative range. We validated these ranges using samples from 193 SARS-CoV-2-exposed individuals. Although IFNγ ELISPOT was the preferred assay, our experimental validation suggested that it under-represented the SARS-CoV-2-specific T cell repertoire. Our data indicate that a combination of AIM and ICS or FluoroSpot assay would better represent the frequency, polyfunctionality, and compartmentalization of the antigen-specific T cell responses. Taken together, our results contribute to defining the ranges of antigen-specific T cell assays and propose a choice of assay that can be employed to better understand the cellular immune response against viral diseases.
Collapse
Affiliation(s)
- Akshay Binayke
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India
- Jawaharlal Nehru University, New Delhi, India
| | - Aymaan Zaheer
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Siddhesh Vishwakarma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Savita Singh
- Translational Health Science and Technology Institute, Faridabad, India
| | - Priyanka Sharma
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India
| | - Rucha Chandwaskar
- Department of Microbiology, AMITY University Rajasthan, Jaipur, India
| | - Mudita Gosain
- Translational Health Science and Technology Institute, Faridabad, India
| | | | | | | | - Ramachandran Thiruvengadam
- Translational Health Science and Technology Institute, Faridabad, India
- Pondicherry Institute of Medical Sciences, Puducherry, India
| | | | | | - Pramod Kumar Garg
- Translational Health Science and Technology Institute, Faridabad, India
- All India Institute of Medical Sciences, New Delhi, India
| | - Amit Awasthi
- Immunology Core Laboratory, Translational Health Science and Technology Institute, Faridabad, India.
- Centre for Immunobiology and Immunotherapy, Translational Health Science and Technology Institute, Faridabad, India.
| |
Collapse
|
7
|
Allen NE, Zhang J, McGhee CNJ. COVID-19 vaccination and corneal allograft rejection- a review. Front Cell Infect Microbiol 2023; 13:1307655. [PMID: 38162575 PMCID: PMC10757323 DOI: 10.3389/fcimb.2023.1307655] [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: 10/05/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Aim To provide a comprehensive literature review on the perceived correlation between COVID-19 vaccination and corneal allograft rejection, and to characterize risk factors, time course, graft outcomes and proposed immunological basis. Methods A literature review was conducted in August 2023 using 4 electronic databases: PubMed, EMBASE, MEDLINE and Scopus. Articles were sourced using key words associated with COVID-19 vaccination and corneal graft. All articles were screened for relevance by abstract review. Duplicates and articles related to COVID-19 infection were excluded. No time limits were set. Additional literature searches regarding cause of corneal graft rejection, rates of graft rejection associated with other vaccines and the cellular mechanism of rejection were also performed. Results 262 articles were identified from the literature search. 37 papers were included in the analysis based on defined inclusion criteria. This consisted of systematic reviews (n=6), review articles (n=5), retrospective studies (n=3), case series (n=8), letter to the editor (n=1) and case reports (n= 14). The majority of reported allograft rejections were in penetrating keratoplasties. Risk factors for COVID-19 vaccination associated rejection were previous allograft rejection episodes, repeat grafts and penetrating keratoplasty. Most reported rejection episodes were mild and resolved with treatment. Notably, several studies reported nil increase in corneal allograft rejection episodes over the COVID-19 vaccination period. Rejection episodes are associated with a broad spectrum of other vaccines and the complete pathophysiology is undetermined. Conclusion Corneal allograft rejection appears to be a rare complication of COVID-19 vaccination most frequently observed in high-risk corneal transplants. The true extent of this correlation remains controversial; however, clinician awareness of this risk is essential to its mitigation. Patient counselling around symptom monitoring following vaccination and discussion around topical steroid prophylaxis may be prudent.
Collapse
Affiliation(s)
| | | | - Charles N. J. McGhee
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
8
|
da Silva Antunes R, Weiskopf D, Sidney J, Rubiro P, Peters B, Arlehamn CSL, Grifoni A, Sette A. The MegaPool Approach to Characterize Adaptive CD4+ and CD8+ T Cell Responses. Curr Protoc 2023; 3:e934. [PMID: 37966108 PMCID: PMC10662678 DOI: 10.1002/cpz1.934] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
Epitopes recognized by T cells are a collection of short peptide fragments derived from specific antigens or proteins. Immunological research to study T cell responses is hindered by the extreme degree of heterogeneity of epitope targets, which are usually derived from multiple antigens; within a given antigen, hundreds of different T cell epitopes can be recognized, differing from one individual to the next because T cell epitope recognition is restricted by the epitopes' ability to bind to MHC molecules, which are extremely polymorphic in different individuals. Testing large pools encompassing hundreds of peptides is technically challenging because of logistical considerations regarding solvent-induced toxicity. To address this issue, we developed the MegaPool (MP) approach based on sequential lyophilization of large numbers of peptides that can be used in a variety of assays to measure T cell responses, including ELISPOT, intracellular cytokine staining, and activation-induced marker assays, and that has been validated in the study of infectious diseases, allergies, and autoimmunity. Here, we describe the procedures for generating and testing MPs, starting with peptide synthesis and lyophilization, as well as a step-by-step guide and recommendations for their handling and experimental usage. Overall, the MP approach is a powerful strategy for studying T cell responses and understanding the immune system's role in health and disease. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Generation of peptide pools ("MegaPools") Basic Protocol 2: MegaPool testing and quantitation of antigen-specific T cell responses.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Paul Rubiro
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | | | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI); La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| |
Collapse
|
9
|
Totolian AA, Smirnov VS, Krasnov AA, Ramsay ES, Dedkov VG, Popova AY. COVID-19 Incidence Proportion as a Function of Regional Testing Strategy, Vaccination Coverage, and Vaccine Type. Viruses 2023; 15:2181. [PMID: 38005859 PMCID: PMC10675075 DOI: 10.3390/v15112181] [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: 09/23/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Introduction: The COVID-19 pandemic has become a serious challenge for humanity almost everywhere globally. Despite active vaccination around the world, the incidence proportion in different countries varies significantly as of May 2022. The reason may be a combination of demographic, immunological, and epidemiological factors. The purpose of this study was to analyze possible relationships between COVID-19 incidence proportion in the population and the types of SARS-CoV-2 vaccines used in different countries globally, taking into account demographic and epidemiological factors. Materials and methods: An initial database was created of demographic and immunoepidemiological information about the COVID-19 situation in 104 countries collected from published official sources and repository data. The baseline included, for each country, population size and density; SARS-CoV-2 testing coverage; vaccination coverage; incidence proportion; and a list of vaccines that were used, including their relative share among all vaccinations. Subsequently, the initial data set was stratified by population and vaccination coverage. The final data set was subjected to statistical processing both in general and taking into account population testing coverage. Results: After formation of the final data set (including 53 countries), it turned out that reported COVID-19 case numbers correlated most strongly with testing coverage and the proportions of vaccine types used, specifically, mRNA (V1); vector (V2); peptide/protein (V3); and whole-virion/inactivated (V4). Due to the fact that an inverse correlation was found between 'reported COVID-19 case numbers' with V2, V3, and V4, these three vaccine types were also combined into one analytic group, 'non-mRNA group' vaccines (Vnmg). When the relationship between vaccine type and incidence proportion was examined, minimum incidence proportion was noted at V1:Vnmg ratios (%:%) from 0:100 to 30:70. Maximum incidence proportion was seen with V1:Vnmg from 80:20 to 100:0. On the other hand, we have shown that the number of reported COVID-19 cases in different countries largely depends on testing coverage. To offset this factor, countries with low and extremely high levels of testing were excluded from the data set; it was then confirmed that the largest number of reported COVID-19 cases occurred in countries with a dominance of V1 vaccines. The fewest reported cases were seen in countries with a dominance of Vnmg vaccines. Conclusion: In this paper, we have shown for the first time that the level of reported COVID-19 incidence proportion depends not only on SARS-CoV-2 testing and vaccination coverage, which is quite logical, but probably also on the vaccine types used. With the same vaccination level and testing coverage, those countries that predominantly use vector and whole-virion vaccines feature incidence proportion that is significantly lower than countries that predominantly use mRNA vaccines.
Collapse
Affiliation(s)
- Areg A. Totolian
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (A.A.T.); (V.S.S.); (A.A.K.); (E.S.R.)
| | - Viacheslav S. Smirnov
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (A.A.T.); (V.S.S.); (A.A.K.); (E.S.R.)
| | - Alexei A. Krasnov
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (A.A.T.); (V.S.S.); (A.A.K.); (E.S.R.)
| | - Edward S. Ramsay
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (A.A.T.); (V.S.S.); (A.A.K.); (E.S.R.)
| | - Vladimir G. Dedkov
- Saint Petersburg Pasteur Institute, 197101 St. Petersburg, Russia; (A.A.T.); (V.S.S.); (A.A.K.); (E.S.R.)
| | - Anna Y. Popova
- Federal Service for Supervision of Consumer Rights Protection and Human Welfare, 127994 Moscow, Russia;
| |
Collapse
|
10
|
Reyes H, Méndez C, Kalergis AM. Statistical explanation of the protective effect of four COVID-19 vaccine doses in the general population. Front Public Health 2023; 11:1253762. [PMID: 37808972 PMCID: PMC10556658 DOI: 10.3389/fpubh.2023.1253762] [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: 07/06/2023] [Accepted: 09/06/2023] [Indexed: 10/10/2023] Open
Abstract
Objectives To assess the effectiveness of four doses of the vaccine against SARS-CoV-2 in the general population and the impact of this on the severity of the disease by age group. Methods By using data from the health authority public data base, we build statistical models using R and the GAMLSS library to explain the behavior of new SARS-CoV-2 infections, active COVID-19 cases, ICU bed requirement total and by age group, and deaths at the national level. Results The four doses of vaccine and at least the interaction between the first and second doses were important explanatory factors for the protective effect against COVID-19. The R2 for new cases per day was 0.5644 and for occupied ICU beds the R2 is 0.9487. For occupied ICU beds for >70 years R2 is 0.9195 and with the interaction between 4 doses as the main factor. Conclusions Although the increase in the number of vaccine doses did not adequately explain the decrease in the number of COVID-19 cases, it explained the decrease in ICU admissions and deaths nationwide and by age group.
Collapse
Affiliation(s)
- Humberto Reyes
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Constanza Méndez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
Salvetat ML, Musa M, Pellegrini F, Salati C, Spadea L, Zeppieri M. Considerations of COVID-19 in Ophthalmology. Microorganisms 2023; 11:2220. [PMID: 37764064 PMCID: PMC10538084 DOI: 10.3390/microorganisms11092220] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/30/2023] [Accepted: 08/30/2023] [Indexed: 09/15/2023] Open
Abstract
Since its emergence in early 2020, the SARS-CoV-2 infection has had a significant impact on the entire eye care system. Ophthalmologists have been categorized as a high-risk group for contracting the virus due to the belief that the eye may be a site of inoculation and transmission of the SARS-CoV-2 infection. As a result, clinical ophthalmologists, optometrists, and eyecare professionals have had to familiarize themselves with the ocular manifestations of COVID-19, as well as its treatments and vaccines. The implementation of measures to prevent the transmission of the virus, such as restrictions, lockdowns, telemedicine, and artificial intelligence (AI), have led to substantial and potentially irreversible changes in routine clinical practice, education, and research. This has resulted in the emergence of a new mode of managing patients in a routine clinical setting. This brief review aims to provide an overview of various aspects of COVID-19 in ophthalmology, including the ocular manifestations related to the disease, the modes of transmission of SARS-CoV-2 infection, precautions taken in ophthalmic practice to prevent the spread of the virus, drugs, and vaccines used in the treatment of COVID-19, the impact of the pandemic on patients, clinicians, and the eye care system as a whole, and the future of ophthalmology conditioned by this global pandemic experience.
Collapse
Affiliation(s)
- Maria Letizia Salvetat
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Mutali Musa
- Department of Optometry, University of Benin, Benin City 300238, Edo State, Nigeria
| | - Francesco Pellegrini
- Department of Ophthalmology, Azienda Sanitaria Friuli Occidentale, 33170 Pordenone, Italy
| | - Carlo Salati
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| | - Leopoldo Spadea
- Eye Clinic, Policlinico Umberto I, “Sapienza” University of Rome, 00142 Rome, Italy
| | - Marco Zeppieri
- Department of Ophthalmology, University Hospital of Udine, 33100 Udine, Italy
| |
Collapse
|
12
|
Pacheco GA, Andrade CA, Gálvez NM, Vázquez Y, Rodríguez-Guilarte L, Abarca K, González PA, Bueno SM, Kalergis AM. Characterization of the humoral and cellular immunity induced by a recombinant BCG vaccine for the respiratory syncytial virus in healthy adults. Front Immunol 2023; 14:1215893. [PMID: 37533867 PMCID: PMC10390696 DOI: 10.3389/fimmu.2023.1215893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/27/2023] [Indexed: 08/04/2023] Open
Abstract
Introduction The human respiratory syncytial virus (hRSV) is responsible for most respiratory tract infections in infants. Even though currently there are no approved hRSV vaccines for newborns or infants, several candidates are being developed. rBCG-N-hRSV is a vaccine candidate previously shown to be safe in a phase I clinical trial in adults (clinicaltrials.gov identifier #NCT03213405). Here, secondary immunogenicity analyses were performed on these samples. Methods PBMCs isolated from immunized volunteers were stimulated with hRSV or mycobacterial antigens to evaluate cytokines and cytotoxic T cell-derived molecules and the expansion of memory T cell subsets. Complement C1q binding and IgG subclass composition of serum antibodies were assessed. Results Compared to levels detected prior to vaccination, perforin-, granzyme B-, and IFN-γ-producing PBMCs responding to stimulus increased after immunization, along with their effector memory response. N-hRSV- and mycobacterial-specific antibodies from rBCG-N-hRSV-immunized subjects bound C1q. Conclusion Immunization with rBCG-N-hRSV induces cellular and humoral immune responses, supporting that rBCG-N-hRSV is immunogenic and safe in healthy individuals. Clinical trial registration https://classic.clinicaltrials.gov/ct2/show/, identifier NCT03213405.
Collapse
Affiliation(s)
- Gaspar A. Pacheco
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M.S. Gálvez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Linmar Rodríguez-Guilarte
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katia Abarca
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediá trica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
13
|
Petrone L, Sette A, de Vries RD, Goletti D. The Importance of Measuring SARS-CoV-2-Specific T-Cell Responses in an Ongoing Pandemic. Pathogens 2023; 12:862. [PMID: 37513709 PMCID: PMC10385870 DOI: 10.3390/pathogens12070862] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Neutralizing antibodies are considered a correlate of protection against SARS-CoV-2 infection and severe COVID-19, although they are not the only contributing factor to immunity: T-cell responses are considered important in protecting against severe COVID-19 and contributing to the success of vaccination effort. T-cell responses after vaccination largely mirror those of natural infection in magnitude and functional capacity, but not in breadth, as T-cells induced by vaccination exclusively target the surface spike glycoprotein. T-cell responses offer a long-lived line of defense and, unlike humoral responses, largely retain reactivity against the SARS-CoV-2 variants. Given the increasingly recognized role of T-cell responses in protection against severe COVID-19, the circulation of SARS-CoV-2 variants, and the potential implementation of novel vaccines, it becomes imperative to continuously monitor T-cell responses. In addition to "classical" T-cell assays requiring the isolation of peripheral blood mononuclear cells, simple whole-blood-based interferon-γ release assays have a potential role in routine T-cell response monitoring. These assays could be particularly useful for immunocompromised people and other clinically vulnerable populations, where interactions between cellular and humoral immunity are complex. As we continue to live alongside COVID-19, the importance of considering immunity as a whole, incorporating both humoral and cellular responses, is crucial.
Collapse
Affiliation(s)
- Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA;
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Rory D. de Vries
- Department Viroscience, Erasmus University Medical Center, 3015CN Rotterdam, The Netherlands;
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases “Lazzaro Spallanzani”-IRCCS, 00149 Rome, Italy;
| |
Collapse
|
14
|
Tarke A, Zhang Y, Methot N, Narowski TM, Phillips E, Mallal S, Frazier A, Filaci G, Weiskopf D, Dan JM, Premkumar L, Scheuermann RH, Sette A, Grifoni A. Targets and cross-reactivity of human T cell recognition of common cold coronaviruses. Cell Rep Med 2023; 4:101088. [PMID: 37295422 PMCID: PMC10242702 DOI: 10.1016/j.xcrm.2023.101088] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 03/17/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023]
Abstract
The coronavirus (CoV) family includes several viruses infecting humans, highlighting the importance of exploring pan-CoV vaccine strategies to provide broad adaptive immune protection. We analyze T cell reactivity against representative Alpha (NL63) and Beta (OC43) common cold CoVs (CCCs) in pre-pandemic samples. S, N, M, and nsp3 antigens are immunodominant, as shown for severe acute respiratory syndrome 2 (SARS2), while nsp2 and nsp12 are Alpha or Beta specific. We further identify 78 OC43- and 87 NL63-specific epitopes, and, for a subset of those, we assess the T cell capability to cross-recognize sequences from representative viruses belonging to AlphaCoV, sarbecoCoV, and Beta-non-sarbecoCoV groups. We find T cell cross-reactivity within the Alpha and Beta groups, in 89% of the instances associated with sequence conservation >67%. However, despite conservation, limited cross-reactivity is observed for sarbecoCoV, indicating that previous CoV exposure is a contributing factor in determining cross-reactivity. Overall, these results provide critical insights in developing future pan-CoV vaccines.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Experimental Medicine and Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genoa, Italy
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Nils Methot
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Tara M Narowski
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Elizabeth Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA 6150, Australia
| | - Simon Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA 6150, Australia
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Gilberto Filaci
- Center of Excellence for Biomedical Research, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Richard H Scheuermann
- J. Craig Venter Institute, La Jolla, CA 92037, USA; Department of Pathology, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| |
Collapse
|
15
|
Luan N, Cao H, Wang Y, Lin K, Hu J, Liu C. Comparison of Immune Responses between Inactivated and mRNA SARS-CoV-2 Vaccines Used for a Booster Dose in Mice. Viruses 2023; 15:1351. [PMID: 37376650 DOI: 10.3390/v15061351] [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: 05/16/2023] [Revised: 06/08/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
A large amount of real-world data suggests that the emergence of variants of concern (VOCs) has brought new challenges to the fight against SARS-CoV-2 because the immune protection elicited by the existing coronavirus disease 2019 (COVID-19) vaccines was weakened. In response to the VOCs, it is necessary to advocate for the administration of booster vaccine doses to extend the effectiveness of vaccines and enhance neutralization titers. In this study, the immune effects of mRNA vaccines based on the WT (prototypic strain) and omicron (B1.1.529) strains for use as booster vaccines were investigated in mice. It was determined that with two-dose inactivated vaccine priming, boosting with mRNA vaccines could elevate IgG titers, enhance cell-mediated immunity, and provide immune protection against the corresponding variants, but cross-protection against distinct strains was inferior. This study comprehensively describes the differences in the mice boosted with mRNA vaccines based on the WT strain and the omicron strain, a harmful VOC that has resulted in a sharp rise in the number of infections, and reveals the most efficacious vaccination strategy against omicron and future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Ning Luan
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Han Cao
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Yunfei Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Kangyang Lin
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Jingping Hu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| | - Cunbao Liu
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming 650118, China
| |
Collapse
|
16
|
de Souza Nogueira J, Santos-Rebouças CB, Piergiorge RM, Valente AP, Gama-Almeida MC, El-Bacha T, Lopes Moreira ML, Baptista Marques BS, de Siqueira JR, de Carvalho EM, da Costa Ferreira O, Porto LC, Kelly da Silva Fidalgo T, Costa Dos Santos G. Metabolic Adaptations Correlated with Antibody Response after Immunization with Inactivated SARS-CoV-2 in Brazilian Subjects. J Proteome Res 2023. [PMID: 37167433 DOI: 10.1021/acs.jproteome.3c00014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The adsorbed vaccine SARS-CoV-2 (inactivated) produced by Sinovac (SV) was the first vaccine against COVID-19 to be used in Brazil. To understand the metabolic effects of SV in Brazilian subjects, NMR-based metabolomics was used, and the immune response was studied in Brazilian subjects. Forty adults without (group-, n = 23) and with previous COVID-19 infection (group+, n = 17) were followed-up for 90 days postcompletion of the vaccine regimen. After 90 days, our results showed that subjects had increased levels of lipoproteins, lipids, and N-acetylation of glycoproteins (NAG) as well as decreased levels of amino acids, lactate, citrate, and 3-hydroxypropionate. NAG and threonine were the highest correlated metabolites with N and S proteins, and neutralizing Ab levels. This study sheds light on the immunometabolism associated with the use of SV in Brazilian subjects from Rio de Janeiro and identifies potential metabolic markers associated with the immune status.
Collapse
Affiliation(s)
- Jeane de Souza Nogueira
- Histocompatibility and Cryopreservation Laboratory, IBRAG, Rio de Janeiro State University, CEP 20950-003 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cíntia Barros Santos-Rebouças
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Mina Piergiorge
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Paula Valente
- CENABIO I, Institute of Medical Biochemistry, CNRMN, BioNMR, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos C Gama-Almeida
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana El-Bacha
- LeBioME-Bioactives, Mitochondria and Placental Metabolism Core, Institute of Nutrition Josué de Castro, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | | | - Orlando da Costa Ferreira
- Molecular Virology Laboratory, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luís Cristóvão Porto
- Histocompatibility and Cryopreservation Laboratory, IBRAG, Rio de Janeiro State University, CEP 20950-003 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tatiana Kelly da Silva Fidalgo
- Department of Preventive and Community Dentistry, Dental School, Rio de Janeiro State University, CEP 20551-030 Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilson Costa Dos Santos
- Department of Genetics, IBRAG, Rio de Janeiro State University, CEP 20550-013 Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
17
|
Xu S, Li J, Wang H, Wang F, Yin Z, Wang Z. Real-world effectiveness and factors associated with effectiveness of inactivated SARS-CoV-2 vaccines: a systematic review and meta-regression analysis. BMC Med 2023; 21:160. [PMID: 37106390 PMCID: PMC10134725 DOI: 10.1186/s12916-023-02861-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The two inactivated SARS-CoV-2 vaccines, CoronaVac and BBIBP-CorV, have been widely used to control the COVID-19 pandemic. The influence of multiple factors on inactivated vaccine effectiveness (VE) during long-term use and against variants is not well understood. METHODS We selected published or preprinted articles from PubMed, Embase, Scopus, Web of Science, medRxiv, BioRxiv, and the WHO COVID-19 database by 31 August 2022. We included observational studies that assessed the VE of completed primary series or homologous booster against SARS-CoV-2 infection or severe COVID-19. We used DerSimonian and Laird random-effects models to calculate pooled estimates and conducted multiple meta-regression with an information theoretic approach based on Akaike's Information Criterion to select the model and identify the factors associated with VE. RESULTS Fifty-one eligible studies with 151 estimates were included. For prevention of infection, VE associated with study region, variants, and time since vaccination; VE was significantly decreased against Omicron compared to Alpha (P = 0.021), primary series VE was 52.8% (95% CI, 43.3 to 60.7%) against Delta and 16.4% (95% CI, 9.5 to 22.8%) against Omicron, and booster dose VE was 65.2% (95% CI, 48.3 to 76.6%) against Delta and 20.3% (95% CI, 10.5 to 28.0%) against Omicron; primary VE decreased significantly after 180 days (P = 0.022). For the prevention of severe COVID-19, VE associated with vaccine doses, age, study region, variants, study design, and study population type; booster VE increased significantly (P = 0.001) compared to primary; though VE decreased significantly against Gamma (P = 0.034), Delta (P = 0.001), and Omicron (P = 0.001) compared to Alpha, primary and booster VEs were all above 60% against each variant. CONCLUSIONS Inactivated vaccine protection against SARS-CoV-2 infection was moderate, decreased significantly after 6 months following primary vaccination, and was restored by booster vaccination. VE against severe COVID-19 was greatest after boosting and did not decrease over time, sustained for over 6 months after the primary series, and more evidence is needed to assess the duration of booster VE. VE varied by variants, most notably against Omicron. It is necessary to ensure booster vaccination of everyone eligible for SARS-CoV-2 vaccines and continue monitoring virus evolution and VE. TRIAL REGISTRATION PROSPERO, CRD42022353272.
Collapse
Affiliation(s)
- Shiyao Xu
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Jincheng Li
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China
| | - Hongyuan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Fuzhen Wang
- Chinese Center for Disease Control and Prevention, National Immunization Programme, Beijing, China
| | - Zundong Yin
- Chinese Center for Disease Control and Prevention, National Immunization Programme, Beijing, China.
| | - Zhifeng Wang
- Department of Health Policy and Management, School of Public Health, Peking University, Beijing, China.
| |
Collapse
|
18
|
Méndez C, Peñaloza HF, Schultz BM, Piña-Iturbe A, Ríos M, Moreno-Tapia D, Pereira-Sánchez P, Leighton D, Orellana C, Covarrubias C, Gálvez NMS, Soto JA, Duarte LF, Rivera-Pérez D, Vázquez Y, Cabrera A, Bustos S, Iturriaga C, Urzua M, Navarrete MS, Rojas Á, Fasce RA, Fernández J, Mora J, Ramírez E, Gaete-Argel A, Acevedo M, Valiente-Echeverría F, Soto-Rifo R, Weiskopf D, Grifoni A, Sette A, Zeng G, Meng W, González-Aramundiz JV, González PA, Abarca K, Melo-González F, Bueno SM, Kalergis AM. Humoral and cellular response induced by a second booster of an inactivated SARS-CoV-2 vaccine in adults. EBioMedicine 2023; 91:104563. [PMID: 37099842 PMCID: PMC10129368 DOI: 10.1016/j.ebiom.2023.104563] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND The Omicron variant has challenged the control of the COVID-19 pandemic due to its immuno-evasive properties. The administration of a booster dose of a SARS-CoV-2 vaccine showed positive effects in the immunogenicity against SARS-CoV-2, effect that is even enhanced after the administration of a second booster. METHODS During a phase-3 clinical trial, we evaluated the effect of a second booster of CoronaVac®, an inactivated vaccine administered 6 months after the first booster, in the neutralization of SARS-CoV-2 (n = 87). In parallel, cellular immunity (n = 45) was analyzed in stimulated peripheral mononuclear cells by flow cytometry and ELISPOT. FINDINGS Although a 2.5-fold increase in neutralization of the ancestral SARS-CoV-2 was observed after the second booster when compared with prior its administration (Geometric mean units p < 0.0001; Geometric mean titer p = 0.0002), a poor neutralization against the Omicron variant was detected. Additionally, the activation of specific CD4+ T lymphocytes remained stable after the second booster and, importantly, equivalent activation of CD4+ T lymphocytes against the Omicron variant and the ancestral SARS-CoV-2 were found. INTERPRETATION Although the neutralizing response against the Omicron variant after the second booster of CoronaVac® was slightly increased, these levels are far from those observed against the ancestral SARS-CoV-2 and could most likely fail to neutralize the virus. In contrast, a robust CD4+T cell response may confer protection against the Omicron variant. FUNDING The Ministry of Health, Government of Chile, the Confederation of Production and Commerce, Chile and SINOVAC Biotech.NIHNIAID. The Millennium Institute on Immunology and Immunotherapy.
Collapse
Affiliation(s)
- Constanza Méndez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Hernán F Peñaloza
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara M Schultz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Piña-Iturbe
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Ríos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Moreno-Tapia
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patricia Pereira-Sánchez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diane Leighton
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia Orellana
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Consuelo Covarrubias
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Luisa F Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alex Cabrera
- Flow Cytometry Facility, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio Bustos
- Flow Cytometry Facility, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Iturriaga
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcela Urzua
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María S Navarrete
- Departamento de Enfermedades Infecciosas del Adulto, División de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Álvaro Rojas
- Departamento de Enfermedades Infecciosas del Adulto, División de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo A Fasce
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Chile
| | - Jorge Fernández
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Chile
| | - Judith Mora
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Chile
| | - Eugenio Ramírez
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Chile
| | - Aracelly Gaete-Argel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Mónica Acevedo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Fernando Valiente-Echeverría
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Ricardo Soto-Rifo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago de Chile, Chile
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, 92037, USA
| | | | | | - José V González-Aramundiz
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katia Abarca
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile.
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile; Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile; Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
19
|
Takheaw N, Liwsrisakun C, Laopajon W, Pata S, Chaiwong W, Inchai J, Duangjit P, Pothirat C, Bumroongkit C, Deesomchok A, Theerakittikul T, Limsukon A, Tajarernmuang P, Niyatiwatchanchai N, Trongtrakul K, Kasinrerk W. Levels and durability of neutralizing antibodies against SARS-CoV-2 Omicron and other variants after ChAdOx-1 or BNT162b2 booster in CoronaVac-primed elderly individuals. Heliyon 2023; 9:e15653. [PMID: 37095993 PMCID: PMC10116116 DOI: 10.1016/j.heliyon.2023.e15653] [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: 05/10/2022] [Revised: 04/05/2023] [Accepted: 04/18/2023] [Indexed: 04/26/2023] Open
Abstract
The outbreak of the SARS-CoV-2 Omicron variant raised the need for vaccine boosting. We evaluated the efficiency of the third booster vaccine, ChAdOx-1 or BNT162b2, in causing a neutralizing antibody (NAb) response and its durability against the Omicron and other variants in elderly individuals previously vaccinated with 2-dose CoronaVac inactivated vaccine. After receiving 2-dose CoronaVac, only 2.2% of subjects had NAbs against the Omicron variant above the cut-off value. Four weeks after boosting, the number of subjects who had NAb levels above the cut-off values in the ChAdOx-1 and BNT162b2 vaccine boosting groups increased to 41.7% and 54.5%, respectively. However, after 12 and 24 weeks of boosting with any vaccines, NAb levels against the Omicron variant dramatically waned. Twenty-four weeks after boosting, only 2% had high levels of NAbs against the Omicron variant. Compared to other variants, the Omicron variant was less responsive to boosting vaccines. The waning rate of NAb levels for the Omicron variant was much faster than that observed in the Alpha, Beta and Delta variants. To combat the Omicron variant, the fourth booster dose is, therefore, recommended for elderly individuals.
Collapse
Affiliation(s)
- Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Chalerm Liwsrisakun
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Warawut Chaiwong
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Juthamas Inchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pilaiporn Duangjit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaicharn Pothirat
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiwat Bumroongkit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Athavudh Deesomchok
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Theerakorn Theerakittikul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Atikun Limsukon
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattraporn Tajarernmuang
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nutchanok Niyatiwatchanchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Konlawij Trongtrakul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
20
|
Buckley PR, Lee CH, Antanaviciute A, Simmons A, Koohy H. A systems approach evaluating the impact of SARS-CoV-2 variant of concern mutations on CD8+ T cell responses. IMMUNOTHERAPY ADVANCES 2023; 3:ltad005. [PMID: 37082106 PMCID: PMC10112682 DOI: 10.1093/immadv/ltad005] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/02/2023] [Indexed: 03/17/2023] Open
Abstract
T cell recognition of SARS-CoV-2 antigens after vaccination and/or natural infection has played a central role in resolving SARS-CoV-2 infections and generating adaptive immune memory. However, the clinical impact of SARS-CoV-2-specific T cell responses is variable and the mechanisms underlying T cell interaction with target antigens are not fully understood. This is especially true given the virus' rapid evolution, which leads to new variants with immune escape capacity. In this study, we used the Omicron variant as a model organism and took a systems approach to evaluate the impact of mutations on CD8+ T cell immunogenicity. We computed an immunogenicity potential score for each SARS-CoV-2 peptide antigen from the ancestral strain and Omicron, capturing both antigen presentation and T cell recognition probabilities. By comparing ancestral vs. Omicron immunogenicity scores, we reveal a divergent and heterogeneous landscape of impact for CD8+ T cell recognition of mutated targets in Omicron variants. While T cell recognition of Omicron peptides is broadly preserved, we observed mutated peptides with deteriorated immunogenicity that may assist breakthrough infection in some individuals. We then combined our scoring scheme with an in silico mutagenesis, to characterise the position- and residue-specific theoretical mutational impact on immunogenicity. While we predict many escape trajectories from the theoretical landscape of substitutions, our study suggests that Omicron mutations in T cell epitopes did not develop under cell-mediated pressure. Our study provides a generalisable platform for fostering a deeper understanding of existing and novel variant impact on antigen-specific vaccine- and/or infection-induced T cell immunity.
Collapse
Affiliation(s)
- Paul R Buckley
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Chloe H Lee
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Agne Antanaviciute
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Alison Simmons
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Hashem Koohy
- Medical Research Council (MRC) Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine (WIMM), John Radcliffe Hospital, University of Oxford, Oxford, UK
- MRC WIMM Centre for Computational Biology, Medical Research Council (MRC) Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Alan Turing Fellow in Health and Medicine
| |
Collapse
|
21
|
Cao L, Guo J, Li H, Ren H, Xiao K, Zhang Y, Zhu S, Song Y, Zhao W, Wu D, Chen Z, Zhang Y, Xia B, Ji T, Yan D, Wang D, Yang Q, Zhou Y, Li X, Hou Z, Xu W. A Beta Strain-Based Spike Glycoprotein Vaccine Candidate Induces Broad Neutralization and Protection against SARS-CoV-2 Variants of Concern. Microbiol Spectr 2023; 11:e0268722. [PMID: 36847495 PMCID: PMC10100794 DOI: 10.1128/spectrum.02687-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 02/04/2023] [Indexed: 03/01/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is still ongoing. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern (VOCs) are circulating worldwide, making it resistant to existing vaccines and antiviral drugs. Therefore, the evaluation of variant-based expanded spectrum vaccines to optimize the immune response and provide broad protectiveness is very important. In this study, we expressed spike trimer protein (S-TM) based on the Beta variant in a GMP-grade workshop using CHO cells. Mice were immunized twice with S-TM protein combined with aluminum hydroxide (Al) and CpG Oligonucleotides (CpG) adjuvant to evaluate its safety and efficacy. BALB/c immunized with S-TM + Al + CpG induced high neutralizing antibody titers against the Wuhan-Hu-1 strain (wild-type, WT), the Beta and Delta variants, and even the Omicron variant. In addition, compared with the S-TM + Al group, the S-TM + Al + CpG group effectively induced a stronger Th1-biased cell immune response in mice. Furthermore, after the second immunization, H11-K18 hACE2 mice were well protected from challenge with the SARS-CoV-2 Beta strain, with a 100% survival rate. The virus load and pathological lesions in the lungs were significantly reduced, and no virus was detected in mouse brain tissue. Our vaccine candidate is practical and effective for current SARS-CoV-2 VOCs, which will support its further clinical development for potential sequential immune and primary immunization. IMPORTANCE Continuous emergence of adaptive mutations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to challenge the use and development of existing vaccines and drugs. The value of variant-based vaccines that are capable of inducing a higher and broader protection immune response against SARS-CoV-2 variants is currently being evaluated. This article shows that a recombinant prefusion spike protein based on a Beta variant was highly immunogenic and could induced a stronger Th1-biased cell immune response in mice and was effectively protective against challenge with the SARS-CoV-2 Beta variant. Importantly, this Beta-based SARS-CoV-2 vaccine could also offer a robust humoral immune response with effectively broad neutralization ability against the wild type and different variants of concern (VOCs): the Beta, Delta, and Omicron BA.1 variants. To date, the vaccine described here has been produced in a pilot scale (200L), and the development, filling process, and toxicological safety evaluation have also been completed, which provides a timely response to the emerging SARS-CoV-2 variants and vaccine development.
Collapse
Affiliation(s)
- Lei Cao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Jinyuan Guo
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hai Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hu Ren
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Kang Xiao
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Shuangli Zhu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yang Song
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Weijia Zhao
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Dan Wu
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Zhihui Chen
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Yanan Zhang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Baicheng Xia
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tianjiao Ji
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongmei Yan
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dongyan Wang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qian Yang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yangzi Zhou
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaolei Li
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhanjun Hou
- Hwellso Biotechnology (Beijing) Co., Ltd., Beijing, China
| | - Wenbo Xu
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, Hubei, China
| |
Collapse
|
22
|
One-Year Post-Vaccination Longitudinal Follow-Up of Quantitative SARS-CoV-2 Anti-Spike Total Antibodies in Health Care Professionals and Evaluation of Correlation with Surrogate Neutralization Test. Vaccines (Basel) 2023; 11:vaccines11020355. [PMID: 36851233 PMCID: PMC9966239 DOI: 10.3390/vaccines11020355] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/08/2023] Open
Abstract
Numerous vaccines have been generated to decrease the morbidity and mortality of COVID-19. This study aims to evaluate the immunogenicity of the heterologous boosts by BioNTech against homologous boosts by CoronaVac at three-month intervals in two health care worker (HCW) cohorts, with or without prior COVID-19, for one year post-vaccination. This is a prospective cohort study in which the humoral responses of 386 HCWs were followed-up longitudinally in six main groups according to their previous COVID-19 exposure and vaccination status. Anti-SARS-CoV-2 spike-RBD total antibody levels were measured and SARS-CoV-2 neutralization antibody (NAbs) responses against the ancestral Wuhan and the Omicron variant were evaluated comparatively using international standard serum for Wuhan and Omicron, as well as with the aid of a conversion tool. The anti-SARS-CoV-2 spike-RBD total Ab and Nab difference between with and without prior COVID-19, three months after two-dose primary vaccination with CoronaVac, was statistically significant (p = 0.001). In the subsequent follow-ups, this difference was not observed between the groups. Those previously infected (PI) and non-previously infected (NPI) groups receiving BioNTech as the third dose had higher anti-SARS-CoV-2 spike total Ab levels (14.2-fold and 17.4-fold, respectively, p = 0.001) and Nab responses (against Wuhan and Omicron) than those receiving CoronaVac. Ab responses after booster vaccination decreased significantly in all groups at the ninth-month follow-up (p < 0.05); however, Abs were still higher in all booster received groups than that in the primary vaccination. Abs were above the protective level at the twelfth-month measurement in the entire of the second BioNTech received group as the fourth dose of vaccination. In the one-year follow-up period, the increased incidence of COVID-19 in the groups vaccinated with two or three doses of CoronaVac compared with the groups vaccinated with BioNTech as a booster suggested that continuing the heterologous CoronaVac/BioNTech vaccination, revised according to current SARS-CoV-2 variants and with at least a six-month interval booster would be an effective and safe strategy for protection against COVID-19, particularly in health care workers.
Collapse
|
23
|
Tarke A, Zhang Y, Methot N, Narowski TM, Phillips E, Mallal S, Frazier A, Filaci G, Weiskopf D, Dan JM, Premkumar L, Scheuermann RH, Sette A, Grifoni A. Targets and cross-reactivity of human T cell recognition of Common Cold Coronaviruses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.04.522794. [PMID: 36656777 PMCID: PMC9844015 DOI: 10.1101/2023.01.04.522794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The Coronavirus (CoV) family includes a variety of viruses able to infect humans. Endemic CoVs that can cause common cold belong to the alphaCoV and betaCoV genera, with the betaCoV genus also containing subgenera with zoonotic and pandemic concern, including sarbecoCoV (SARS-CoV and SARS-CoV-2) and merbecoCoV (MERS-CoV). It is therefore warranted to explore pan-CoV vaccine concepts, to provide adaptive immune protection against new potential CoV outbreaks, particularly in the context of betaCoV sub lineages. To explore the feasibility of eliciting CD4 + T cell responses widely cross-recognizing different CoVs, we utilized samples collected pre-pandemic to systematically analyze T cell reactivity against representative alpha (NL63) and beta (OC43) common cold CoVs (CCC). Similar to previous findings on SARS-CoV-2, the S, N, M, and nsp3 antigens were immunodominant for both viruses while nsp2 and nsp12 were immunodominant for NL63 and OC43, respectively. We next performed a comprehensive T cell epitope screen, identifying 78 OC43 and 87 NL63-specific epitopes. For a selected subset of 18 epitopes, we experimentally assessed the T cell capability to cross-recognize sequences from representative viruses belonging to alphaCoV, sarbecoCoV, and beta-non-sarbecoCoV groups. We found general conservation within the alpha and beta groups, with cross-reactivity experimentally detected in 89% of the instances associated with sequence conservation of >67%. However, despite sequence conservation, limited cross-reactivity was observed in the case of sarbecoCoV (50% of instances), indicating that previous CoV exposure to viruses phylogenetically closer to this subgenera is a contributing factor in determining cross-reactivity. Overall, these results provided critical insights in the development of future pan-CoV vaccines.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Experimental Medicine and Center of Excellence for Biomedical Research (CEBR), University of Genoa, Genoa, 16132, Italy
| | - Yun Zhang
- J. Craig Venter Institute, La Jolla, CA 92037, USA
| | - Nils Methot
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Tara M Narowski
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Elizabeth Phillips
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia, Australia
| | - Simon Mallal
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, Western Australia, Australia
| | - April Frazier
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Gilberto Filaci
- Center of Excellence for Biomedical Research, Department of Internal Medicine, University of Genoa, Genoa 16132, Italy
- Biotherapy Unit, IRCCS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599-7290, USA
| | - Richard H Scheuermann
- J. Craig Venter Institute, La Jolla, CA 92037, USA
- Department of Pathology, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- These authors contributed equally
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
- These authors contributed equally
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- These authors contributed equally
- Lead Contact
| |
Collapse
|
24
|
Rodríguez-Guilarte L, Ramírez MA, Andrade CA, Kalergis AM. LAG-3 Contribution to T Cell Downmodulation during Acute Respiratory Viral Infections. Viruses 2023; 15:147. [PMID: 36680187 PMCID: PMC9865459 DOI: 10.3390/v15010147] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
LAG-3 is a type I transmembrane protein expressed on immune cells, such as activated T cells, and binds to MHC class II with high affinity. LAG-3 is an inhibitory receptor, and its multiple biological activities on T cell activation and effector functions play a regulatory role in the immune response. Immunotherapies directed at immune checkpoints, including LAG-3, have become a promising strategy for controlling malignant tumors and chronic viral diseases. Several studies have suggested an association between the expression of LAG-3 with an inadequate immune response during respiratory viral infections and the susceptibility to reinfections, which might be a consequence of the inhibition of T cell effector functions. However, important information relative to therapeutic potential during acute viral lower respiratory tract infections and the mechanism of action of the LAG-3 checkpoint remains to be characterized. In this article, we discuss the contribution of LAG-3 to the impairment of T cells during viral respiratory infections. Understanding the host immune response to respiratory infections is crucial for developing effective vaccines and therapies.
Collapse
Affiliation(s)
- Linmar Rodríguez-Guilarte
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Mario A. Ramírez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Catalina A. Andrade
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| |
Collapse
|
25
|
Yu P, Liu Z, Zhu Z, Yang J, Deng M, Chen M, Lai C, Kong W, Xiong S, Wan L, Mai W, Chen L, Lei Y, Khan SA, Ruan J, Kang A, Guo X, Zhou Q, Li W, Chen Z, Liang Y, Li P, Zhang L, Ji T. Omicron variants breakthrough infection elicited higher specific memory immunity than third dose booster in healthy vaccinees. Virol Sin 2023; 38:233-243. [PMID: 36603767 PMCID: PMC10176432 DOI: 10.1016/j.virs.2022.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/30/2022] [Indexed: 01/03/2023] Open
Abstract
Homologous booster, heterologous booster, and Omicron variants breakthrough infection (OBI) could improve the humoral immunity against Omicron variants. Questions concerning about memory B cells (MBCs) and T cells immunity against Omicron variants, features of long-term immunity, after booster and OBI, needs to be explored. Here, comparative analysis demonstrate antibody and T cell immunity against ancestral strain, Delta and Omicron variants in Omicron breakthrough infected patients (OBIPs) are comparable to that in Ad5-nCoV boosted healthy volunteers (HVs), higher than that in inactivated vaccine (InV) boosted HVs. However, memory B cells (MBCs) immunity against Omicron variants was highest in OBIPs, followed by Ad5-nCoV boosted and InV boosted HVs. OBIPs and Ad5-nCoV boosted HVs have higher classical MBCs and activated MBCs, and lower naïve MBCs and atypical MBCs relative to both vaccine boosted HVs. Collectively, these data indicate Omicron breakthrough infection elicit higher MBCs and T cells against SARS-CoV-2 especially Omicron variants relative to homologous InV booster and heterologous Ad5-nCoV booster.
Collapse
Affiliation(s)
- Pei Yu
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Zijian Liu
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China
| | - Zhuoqi Zhu
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Jiaqing Yang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Min Deng
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Mingxiao Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Changchun Lai
- Clinical Laboratory Medicine Department, Maoming People's Hospital, Maoming, 525000, China
| | - Weiya Kong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shilong Xiong
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Li Wan
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Weikang Mai
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Lu Chen
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Yu Lei
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Shahzad Akbar Khan
- Laboratory of Pathology, Department of Pathobiology, University of Poonch Rawalakot Azad Kashmir Pakistan 12350, Pakistan
| | - Jianfeng Ruan
- Hospital Infection-Control Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - An Kang
- Medical Examination Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Xuguang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510140, China
| | - Qiang Zhou
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenrui Li
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China
| | - Zheng Chen
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China.
| | - Yuemei Liang
- Clinical Laboratory Medicine Department, Dongguan Ninth People's Hospital, Dongguan, 523016, China.
| | - Pingchao Li
- State Key Laboratories of Respiratory Diseases, Guangdong-Hong Kong-Macao Joint Laboratory of Infectious Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510535, China.
| | - Lei Zhang
- Kidney Transplant Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Department of Organ Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, 510630, China.
| | - Tianxing Ji
- Clinical Laboratory Medicine Department, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, China; Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
26
|
Wang H, Gan M, Wu B, Zeng R, Wang Z, Xu J, Li J, Zhang Y, Cao J, Chen L, Di D, Peng S, Lei J, Zhao Y, Song X, Yuan T, Zhou T, Liu Q, Yi J, Wang X, Cai H, Lei Y, Wen Y, Li W, Chen Q, Wang Y, Long P, Yuan Y, Wang C, Pan A, Wang Q, Gong R, Fan X, Wu T, Liu L. Humoral and cellular immunity of two-dose inactivated COVID-19 vaccination in Chinese children: A prospective cohort study. J Med Virol 2023; 95:e28380. [PMID: 36478357 PMCID: PMC9877748 DOI: 10.1002/jmv.28380] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/18/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022]
Abstract
Children are the high-risk group for COVID-19, and in need of vaccination. However, humoral and cellular immune responses of COVID-19 vaccine remain unclear in vaccinated children. To establish the rational immunization strategy of inactivated COVID-19 vaccine for children, the immunogenicity of either one dose or two doses of the vaccine in children was evaluated. A prospective cohort study of 322 children receiving inactivated COVID-19 vaccine was established in China. The baseline was conducted after 28 days of the first dose, and the follow-up was conducted after 28 days of the second dose. The median titers of receptor binding domain (RBD)-IgG, and neutralizing antibody (NAb) against prototype strain and Omicron variant after the second dose increased significantly compared to those after the first dose (first dose: 70.0, [interquartile range, 30.0-151.0] vs. second dose: 1261.0 [636.0-2060.0] for RBD-IgG; 2.5 [2.5-18.6] vs. 252.0 [138.6-462.1] for NAb against prototype strain; 2.5 [2.5-2.5] vs. 15.0 [7.8-26.5] for NAb against Omicron variant, all p < 0.05). The flow cytometry results showed that the first dose elicited SARS-CoV-2 specific cellular immunity, while the second dose strengthened SARS-CoV-2 specific IL-2+ or TNF-α+ monofunctional, IFN-γ+ TNF-α+ bifunctional, and IFN-γ- IL-2+ TNF-α+ multifunctional CD4+ T cell responses (p < 0.05). Moreover, SARS-CoV-2 specific memory T cells were generated after the first vaccination, including the central memory T cells and effector memory T cells. The present findings provide scientific evidence for the vaccination strategy of the inactive vaccines among children against COVID-19 pandemic.
Collapse
Affiliation(s)
- Hao Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Mengze Gan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bihao Wu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanChina
| | - Rui Zeng
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Zhen Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Jun Xu
- Qichun Center for Disease Control and PreventionHuanggangChina
| | - Jia Li
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Yandi Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jinge Cao
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Li Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanChina
| | - Dongsheng Di
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Siyuan Peng
- Qichun Center for Disease Control and PreventionHuanggangChina
| | - Jinfeng Lei
- Qichun Center for Disease Control and PreventionHuanggangChina
| | - Yingying Zhao
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Xuemei Song
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Tingting Yuan
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Tingting Zhou
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Qian Liu
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Jing Yi
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Xi Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Hao Cai
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Yanshou Lei
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Yuying Wen
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Wenhui Li
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Qinlin Chen
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Yufei Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Pinpin Long
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Yu Yuan
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Chaolong Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - An Pan
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Qi Wang
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega‐ScienceChinese Academy of SciencesWuhanChina
| | - Xionglin Fan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tangchun Wu
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| | - Li Liu
- Department of Occupational and Environmental Health and Department of Epidemiology and Biostatistics, Ministry of Education and State Key Laboratory of Environmental HealthHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
27
|
Heinen N, Marheinecke CS, Bessen C, Blazquez-Navarro A, Roch T, Stervbo U, Anft M, Plaza-Sirvent C, Busse S, Klöhn M, Schrader J, Vidal Blanco E, Urlaub D, Watzl C, Hoffmann M, Pöhlmann S, Tenbusch M, Steinmann E, Todt D, Hagenbeck C, Zimmer G, Schmidt WE, Quast DR, Babel N, Schmitz I, Pfänder S. In-depth analysis of T cell immunity and antibody responses in heterologous prime-boost-boost vaccine regimens against SARS-CoV-2 and Omicron variant. Front Immunol 2022; 13:1062210. [PMID: 36618413 PMCID: PMC9811676 DOI: 10.3389/fimmu.2022.1062210] [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: 10/05/2022] [Accepted: 11/29/2022] [Indexed: 12/24/2022] Open
Abstract
With the emergence of novel Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) Variants of Concern (VOCs), vaccination studies that elucidate the efficiency and effectiveness of a vaccination campaign are critical to assess the durability and the protective immunity provided by vaccines. SARS-CoV-2 vaccines have been found to induce robust humoral and cell-mediated immunity in individuals vaccinated with homologous vaccination regimens. Recent studies also suggest improved immune response against SARS-CoV-2 when heterologous vaccination strategies are employed. Yet, few data exist on the extent to which heterologous prime-boost-boost vaccinations with two different vaccine platforms have an impact on the T cell-mediated immune responses with a special emphasis on the currently dominantly circulating Omicron strain. In this study, we collected serum and peripheral blood mononuclear cells (PBMCs) from 57 study participants of median 35-year old's working in the health care field, who have received different vaccination regimens. Neutralization assays revealed robust but decreased neutralization of Omicron VOC, including BA.1 and BA.4/5, compared to WT SARS-CoV-2 in all vaccine groups and increased WT SARS-CoV-2 binding and neutralizing antibodies titers in homologous mRNA prime-boost-boost study participants. By investigating cytokine production, we found that homologous and heterologous prime-boost-boost-vaccination induces a robust cytokine response of CD4+ and CD8+ T cells. Collectively, our results indicate robust humoral and T cell mediated immunity against Omicron in homologous and heterologous prime-boost-boost vaccinated study participants, which might serve as a guide for policy decisions.
Collapse
Affiliation(s)
- Natalie Heinen
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| | | | - Clara Bessen
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, Germany
| | - Arturo Blazquez-Navarro
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital, University Hospital of the Ruhr University Bochum, Herne, Germany
- BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Toralf Roch
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital, University Hospital of the Ruhr University Bochum, Herne, Germany
- BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Ulrik Stervbo
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital, University Hospital of the Ruhr University Bochum, Herne, Germany
| | - Moritz Anft
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital, University Hospital of the Ruhr University Bochum, Herne, Germany
| | | | - Sandra Busse
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, Germany
| | - Mara Klöhn
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Jil Schrader
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Elena Vidal Blanco
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Doris Urlaub
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors (IfADo) at TU Dortmund, Dortmund, Germany
| | - Carsten Watzl
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors (IfADo) at TU Dortmund, Dortmund, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
| | - Matthias Tenbusch
- Institut für klinische und molekulare Virologie, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Eike Steinmann
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - Daniel Todt
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
- European Virus Bioinformatics Center, Jena, Germany
| | - Carsten Hagenbeck
- Clinic for Gynecology and Obstetrics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Gert Zimmer
- Clinic for Gynecology and Obstetrics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | | | - Daniel Robert Quast
- Department of Medicine I, St. Josef-Hospital Bochum, Ruhr University Bochum, Bochum, Germany
| | - Nina Babel
- Center for Translational Medicine and Immune Diagnostics Laboratory, Medical Department I, Marien Hospital, University Hospital of the Ruhr University Bochum, Herne, Germany
- BIH Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr University Bochum, Bochum, Germany
| | - Stephanie Pfänder
- Department of Molecular & Medical Virology, Ruhr University Bochum, Bochum, Germany
| |
Collapse
|
28
|
Abstract
Multiple vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been evaluated in clinical trials. However, trials addressing the immune response in the pediatric population are scarce. The inactivated vaccine CoronaVac has been shown to be safe and immunogenic in a phase 1/2 clinical trial in a pediatric cohort in China. Here, we report interim safety and immunogenicity results of a phase 3 clinical trial for CoronaVac in healthy children and adolescents in Chile. Participants 3 to 17 years old received two doses of CoronaVac in a 4-week interval until 31 December 2021. Local and systemic adverse reactions were registered for volunteers who received one or two doses of CoronaVac. Whole-blood samples were collected from a subgroup of 148 participants for humoral and cellular immunity analyses. The main adverse reaction reported after the first and second doses was pain at the injection site. Four weeks after the second dose, an increase in neutralizing antibody titer was observed in subjects relative to their baseline visit. Similar results were found for activation of specific CD4+ T cells. Neutralizing antibodies were identified against the Delta and Omicron variants. However, these titers were lower than those for the D614G strain. Importantly, comparable CD4+ T cell responses were detected against these variants of concern. Therefore, CoronaVac is safe and immunogenic in subjects 3 to 17 years old, inducing neutralizing antibody secretion and activating CD4+ T cells against SARS-CoV-2 and its variants. (This study has been registered at ClinicalTrials.gov under no. NCT04992260.) IMPORTANCE This work evaluated the immune response induced by two doses of CoronaVac separated by 4 weeks in healthy children and adolescents in Chile. To date, few studies have described the effects of CoronaVac in the pediatric population. Therefore, it is essential to generate knowledge regarding the protection of vaccines in this population. Along these lines, we reported the anti-S humoral response and cellular immune response to several SARS-CoV-2 proteins that have been published and recently studied. Here, we show that a vaccination schedule consisting of two doses separated by 4 weeks induces the secretion of neutralizing antibodies against SARS-CoV-2. Furthermore, CoronaVac induces the activation of CD4+ T cells upon stimulation with peptides from the proteome of SARS-CoV-2. These results indicate that, even though the neutralizing antibody response induced by vaccination decreases against the Delta and Omicron variants, the cellular response against these variants is comparable to the response against the ancestral strain D614G, even being significantly higher against Omicron.
Collapse
|
29
|
Villegas C, Ortiz A, Arriagada V, Ortega S, Walker J, Arriagada E, Kalergis AM, Huepe C. Influence of online opinions and interactions on the Covid-19 vaccination in Chile. Sci Rep 2022; 12:21288. [PMID: 36494384 PMCID: PMC9734170 DOI: 10.1038/s41598-022-23738-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/04/2022] [Indexed: 12/13/2022] Open
Abstract
We analyze 6 months of Twitter conversations related to the Chilean Covid-19 vaccination process, in order to understand the online forces that argue for or against it and suggest effective digital communication strategies. Using AI, we classify accounts into four categories that emerge from the data as a result of the type of language used. This classification naturally distinguishes pro- and anti-vaccine activists from moderates that promote or inhibit vaccination in discussions, which also play a key role that should be addressed by public policies. We find that all categories display relatively constant opinions, but that the number of tweeting accounts grows in each category during controversial periods. We also find that accounts disfavoring vaccination tend to appear in the periphery of the interaction network, which is consistent with Chile's high immunization levels. However, these are more active in addressing those favoring vaccination than vice-versa, revealing a potential communication problem even in a society where the antivaccine movement has no central role. Our results highlight the importance of social network analysis to understand public discussions and suggest online interventions that can help achieve successful immunization campaigns.
Collapse
Affiliation(s)
- Claudio Villegas
- grid.7870.80000 0001 2157 0406Social Listening Lab SoL-UC, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.441788.60000 0001 2154 0610School of Anthropology, Universidad Academia de Humanismo Cristiano, Santiago, Chile
| | - Abril Ortiz
- grid.7870.80000 0001 2157 0406Social Listening Lab SoL-UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Víctor Arriagada
- grid.7870.80000 0001 2157 0406Social Listening Lab SoL-UC, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.441788.60000 0001 2154 0610School of Anthropology, Universidad Academia de Humanismo Cristiano, Santiago, Chile
| | - Sofía Ortega
- grid.7870.80000 0001 2157 0406Social Listening Lab SoL-UC, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.441788.60000 0001 2154 0610School of Anthropology, Universidad Academia de Humanismo Cristiano, Santiago, Chile
| | - Juan Walker
- grid.7870.80000 0001 2157 0406School of Communications, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo Arriagada
- grid.7870.80000 0001 2157 0406Social Listening Lab SoL-UC, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.7870.80000 0001 2157 0406School of Communications, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.7870.80000 0001 2157 0406Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- grid.7870.80000 0001 2157 0406Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile ,grid.7870.80000 0001 2157 0406Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristián Huepe
- grid.7870.80000 0001 2157 0406School of Communications, Pontificia Universidad Católica de Chile, Santiago, Chile ,CHuepe Labs, Chicago, IL 60622 USA ,grid.16753.360000 0001 2299 3507Northwestern Institute on Complex Systems and ESAM, Northwestern University, Evanston, IL 60208 USA
| |
Collapse
|
30
|
Tanriover MD, Aydin OA, Guner R, Yildiz O, Celik I, Doganay HL, Kose S, Akhan S, Akalin EH, Sezer Z, Ozdarendeli A, Unal S. Efficacy, Immunogenicity, and Safety of the Two-Dose Schedules of TURKOVAC versus CoronaVac in Healthy Subjects: A Randomized, Observer-Blinded, Non-Inferiority Phase III Trial. Vaccines (Basel) 2022; 10:1865. [PMID: 36366373 PMCID: PMC9698857 DOI: 10.3390/vaccines10111865] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 09/29/2023] Open
Abstract
We present the interim results of the efficacy, immunogenicity, and safety of the two-dose schedules of TURKOVAC versus CoronaVac. This was a randomized, observer-blinded, non-inferiority trial (NCT04942405). Volunteers were 18-55 years old and randomized at a 1:1 ratio to receive either TURKOVAC or CoronaVac at Day 0 and Day 28, both of which are 3 μg/0.5 mL of inactivated severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) adsorbed to aluminum hydroxide. The primary efficacy outcome was the prevention of polymerase chain reaction (PCR)-confirmed symptomatic coronavirus disease 2019 (COVID-19) at least 14 days after the second dose in the modified per-protocol (mPP) group. Safety analyses were performed in the modified intention-to-treat (mITT) group. Between 22 June 2021 and 7 January 2022, 1290 participants were randomized. The mITT group consisted of 915 participants, and the mPP group consisted of 732 participants. During a median follow-up of 90 (IQR 86-90) days, the relative risk reduction with TURKOVAC compared to CoronaVac was 41.03% (95% CI 12.95-60.06) for preventing PCR-confirmed symptomatic COVID-19. The incidences of adverse events (AEs) overall were 58.8% in TURKOVAC and 49.7% in CoronaVac arms (p = 0.006), with no fatalities or grade four AEs. TURKOVAC was non-inferior to CoronaVac in terms of efficacy and demonstrated a good safety and tolerability profile.
Collapse
Affiliation(s)
- Mine Durusu Tanriover
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, 06230 Ankara, Türkiye
- Vaccine Institute, Hacettepe University, 06230 Ankara, Türkiye
| | - Ozlem Altuntas Aydin
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Başaksehir Cam and Sakura City Hospital, 34480 Istanbul, Türkiye
| | - Rahmet Guner
- Infectious Diseases and Clinical Microbiology Clinic, Ankara Yildirim Beyazit University, Ankara City Hospital, 06800 Ankara, Türkiye
| | - Orhan Yildiz
- Department of Infectious Diseases and Clinical Microbiology, Erciyes University Faculty of Medicine, 38030 Kayseri, Türkiye
| | - Ilhami Celik
- Department of Infectious Diseases and Clinical Microbiology, Kayseri City Training and Research Hospital, 38080 Kayseri, Türkiye
| | - Hamdi Levent Doganay
- Department of Gastroenterology, Medical Park Pendik Hospital, 34899 Istanbul, Türkiye
- Department of Internal Medicine, Bahcesehir University School of Medicine, 34734 Istanbul, Türkiye
| | - Sukran Kose
- Infectious Diseases Clinic, University of Health Sciences, Izmir Tepecik Training and Research Hospital, 35020 Izmir, Türkiye
| | - Sila Akhan
- Department of Infectious Diseases and Clinical Microbiology, Kocaeli University Faculty of Medicine, 41001 Kocaeli, Türkiye
| | - Emin Halis Akalin
- Department of Infectious Diseases and Clinical Microbiology, Bursa Uludag University Faculty of Medicine, 16059 Bursa, Türkiye
| | - Zafer Sezer
- Department of Medical Pharmacology, Erciyes University Faculty of Medicine, 38030 Kayseri, Türkiye
| | - Aykut Ozdarendeli
- Department of Microbiology, Erciyes University Faculty of Medicine, 38030 Kayseri, Türkiye
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, 38280 Kayseri, Türkiye
| | - Serhat Unal
- Vaccine Institute, Hacettepe University, 06230 Ankara, Türkiye
- Department of Infectious Diseases and Clinical Microbiology, Hacettepe University Faculty of Medicine, 06230 Ankara, Türkiye
| | | |
Collapse
|
31
|
Covarrubias CE, Rivera TA, Soto CA, Deeks T, Kalergis AM. Current GMP standards for the production of vaccines and antibodies: An overview. Front Public Health 2022; 10:1021905. [PMID: 36743162 PMCID: PMC9891391 DOI: 10.3389/fpubh.2022.1021905] [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: 08/17/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
The manufacture of pharmaceutical products made under good manufacturing practices (GMP) must comply with the guidelines of national regulatory bodies based on international or regional compendia. The existence of this type of regulation allows pharmaceutical laboratories to count on the standardization of high-quality production processes, obtaining a safe product for human use, with a positive impact on public health. In addition, the COVID-19 pandemic highlights the importance of having more and better-distributed manufacturing plants, emphasizing regions such as Latin America. This review shows the most important GMP standards in the world and, in particular, their relevance in the production of vaccines and antibodies.
Collapse
Affiliation(s)
- Consuelo E. Covarrubias
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Thomas A. Rivera
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Soto
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Trevor Deeks
- Deeks Pharmaceutical Consulting Services, Rockville, MD, United States
| | - Alexis M. Kalergis
- Millenium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Gálvez NMS, Pacheco GA, Schultz BM, Melo-González F, Soto JA, Duarte LF, González LA, Rivera-Pérez D, Ríos M, Berrios RV, Vázquez Y, Moreno-Tapia D, Vallejos OP, Andrade CA, Hoppe-Elsholz G, Iturriaga C, Urzua M, Navarrete MS, Rojas Á, Fasce R, Fernández J, Mora J, Ramírez E, Gaete-Argel A, Acevedo ML, Valiente-Echeverría F, Soto-Rifo R, Weiskopf D, Grifoni A, Sette A, Zeng G, Meng W, González-Aramundiz JV, Johnson M, Goldblatt D, González PA, Abarca K, Bueno SM, Kalergis AM. Differences in the immune response elicited by two immunization schedules with an inactivated SARS-CoV-2 vaccine in a randomized phase 3 clinical trial. eLife 2022; 11:e81477. [PMID: 36226829 PMCID: PMC9596164 DOI: 10.7554/elife.81477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/09/2022] [Indexed: 11/29/2022] Open
Abstract
Background The development of vaccines to control the coronavirus disease 2019 (COVID-19) pandemic progression is a worldwide priority. CoronaVac is an inactivated severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine approved for emergency use with robust efficacy and immunogenicity data reported in trials in China, Brazil, Indonesia, Turkey, and Chile. Methods This study is a randomized, multicenter, and controlled phase 3 trial in healthy Chilean adults aged ≥18 years. Volunteers received two doses of CoronaVac separated by 2 (0-14 schedule) or 4 weeks (0-28 schedule); 2302 volunteers were enrolled, 440 were part of the immunogenicity arm, and blood samples were obtained at different times. Samples from a single center are reported. Humoral immune responses were evaluated by measuring the neutralizing capacities of circulating antibodies. Cellular immune responses were assessed by ELISPOT and flow cytometry. Correlation matrixes were performed to evaluate correlations in the data measured. Results Both schedules exhibited robust neutralizing capacities with the response induced by the 0-28 schedule being better. No differences were found in the concentration of antibodies against the virus and different variants of concern (VOCs) between schedules. Stimulation of peripheral blood mononuclear cells (PBMCs) with Mega pools of Peptides (MPs) induced the secretion of interferon (IFN)-γ and the expression of activation induced markers in CD4+ T cells for both schedules. Correlation matrixes showed strong correlations between neutralizing antibodies and IFN-γ secretion. Conclusions Immunization with CoronaVac in Chilean adults promotes robust cellular and humoral immune responses. The 0-28 schedule induced a stronger humoral immune response than the 0-14 schedule. Funding Ministry of Health, Government of Chile, Confederation of Production and Commerce & Millennium Institute on Immunology and Immunotherapy, Chile. Clinical trial number NCT04651790.
Collapse
Affiliation(s)
- Nicolás MS Gálvez
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Gaspar A Pacheco
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Bárbara M Schultz
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Felipe Melo-González
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés BelloSantiagoChile
| | - Jorge A Soto
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés BelloSantiagoChile
| | - Luisa F Duarte
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Liliana A González
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Daniela Rivera-Pérez
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Mariana Ríos
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Roslye V Berrios
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Daniela Moreno-Tapia
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Omar P Vallejos
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Catalina A Andrade
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Guillermo Hoppe-Elsholz
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Carolina Iturriaga
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| | - Marcela Urzua
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| | - María S Navarrete
- Centro de Investigación Clínica UC, Pontificia Universidad Católica de ChileSantiagoChile
| | - Álvaro Rojas
- Departamento de Enfermedades Infecciosas del Adulto, División de Medicina, Escuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| | - Rodrigo Fasce
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de ChileSantiagoChile
| | - Jorge Fernández
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de ChileSantiagoChile
| | - Judith Mora
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de ChileSantiagoChile
| | - Eugenio Ramírez
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de ChileSantiagoChile
| | - Aracelly Gaete-Argel
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de ChileSantiagoChile
| | - Mónica L Acevedo
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de ChileSantiagoChile
| | - Fernando Valiente-Echeverría
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de ChileSantiagoChile
| | - Ricardo Soto-Rifo
- Laboratory of Molecular and Cellular Virology, Virology Program, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de ChileSantiagoChile
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for ImmunologyLa JollaUnited States
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for ImmunologyLa JollaUnited States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for ImmunologyLa JollaUnited States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of CaliforniaSan DiegoUnited States
| | | | | | | | - José V González-Aramundiz
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de ChileSantiagoChile
| | - Marina Johnson
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| | - David Goldblatt
- Department of Infection, Inflammation and Immunity, Great Ormond Street Institute of Child Health, University College LondonLondonUnited Kingdom
| | - Pablo A González
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Katia Abarca
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| | - Susan M Bueno
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and ImmunotherapySantiagoChile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de ChileSantiagoChile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
33
|
Rabaan AA, Mutair AA, Hajissa K, Alfaraj AH, Al-Jishi JM, Alhajri M, Alwarthan S, Alsuliman SA, Al-Najjar AH, Al Zaydani IA, Al-Absi GH, Alshaikh SA, Alkathlan MS, Almuthree SA, Alawfi A, Alshengeti A, Almubarak FZ, Qashgari MS, Abdalla ANK, Alhumaid S. A Comprehensive Review on the Current Vaccines and Their Efficacies to Combat SARS-CoV-2 Variants. Vaccines (Basel) 2022; 10:1655. [PMID: 36298520 PMCID: PMC9611209 DOI: 10.3390/vaccines10101655] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
Since the first case of Coronavirus disease (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019, SARS-CoV-2 infection has affected many individuals worldwide. Eventually, some highly infectious mutants-caused by frequent genetic recombination-have been reported for SARS-CoV-2 that can potentially escape from the immune responses and induce long-term immunity, linked with a high mortality rate. In addition, several reports stated that vaccines designed for the SARS-CoV-2 wild-type variant have mixed responses against the variants of concern (VOCs) and variants of interest (VOIs) in the human population. These results advocate the designing and development of a panvaccine with the potential to neutralize all the possible emerging variants of SARS-CoV-2. In this context, recent discoveries suggest the design of SARS-CoV-2 panvaccines using nanotechnology, siRNA, antibodies or CRISPR-Cas platforms. Thereof, the present comprehensive review summarizes the current vaccine design approaches against SARS-CoV-2 infection, the role of genetic mutations in the emergence of new viral variants, the efficacy of existing vaccines in limiting the infection of emerging SARS-CoV-2 variants, and efforts or challenges in designing SARS panvaccines.
Collapse
Affiliation(s)
- Ali A. Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran 31311, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur 22610, Pakistan
| | - Abbas Al Mutair
- Research Center, Almoosa Specialist Hospital, Al-Ahsa 36342, Saudi Arabia
- College of Nursing, Princess Norah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia
- School of Nursing, Wollongong University, Wollongong, NSW 2522, Australia
- Nursing Department, Prince Sultan Military College of Health Sciences, Dhahran 33048, Saudi Arabia
| | - Khalid Hajissa
- Department of Medical Microbiology & Parasitology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
| | - Amal H. Alfaraj
- Pediatric Department, Abqaiq General Hospital, First Eastern Health Cluster, Abqaiq 33261, Saudi Arabia
| | - Jumana M. Al-Jishi
- Internal Medicine Department, Qatif Central Hospital, Qatif 635342, Saudi Arabia
| | - Mashael Alhajri
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Sara Alwarthan
- Department of Internal Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Shahab A. Alsuliman
- Infectious Disease Division, Department of Internal Medicine, Dammam Medical Complex, Dammam 32245, Saudi Arabia
| | - Amal H. Al-Najjar
- Drug & Poison Information Center, Pharmacy Department, Security Forces Hospital Program, Riyadh 3643, Saudi Arabia
| | - Ibrahim A. Al Zaydani
- Department of Pediatric Infectious Diseases, Abha Maternity and Children Hospital, Abha 62526, Saudi Arabia
| | - Ghadeer Hassan Al-Absi
- Department of Pharmacy Practice, College of Pharmacy, Alfaisal University, Riyadh 325476, Saudi Arabia
| | - Sana A. Alshaikh
- Diagnostic Virology Laboratory, Maternity and Children Hospital, Eastern Health Cluster, Dammam 32253, Saudi Arabia
| | - Mohammed S. Alkathlan
- Infectious Diseases Department, King Fahad Specialist Hospital, Buraydah 52382, Saudi Arabia
| | - Souad A. Almuthree
- Department of Infectious Disease, King Abdullah Medical City, Makkah 43442, Saudi Arabia
| | - Abdulsalam Alawfi
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah 41491, Saudi Arabia
| | - Amer Alshengeti
- Department of Pediatrics, College of Medicine, Taibah University, Al-Madinah 41491, Saudi Arabia
- Department of Infection Prevention and Control, Prince Mohammad Bin Abdulaziz Hospital, National Guard Health Affairs, Al-Madinah 41491, Saudi Arabia
| | - Fatimah Z. Almubarak
- Department of Family Medicine, Family Medicine Academy, Dammam 36365, Saudi Arabia
| | - Mohammed S. Qashgari
- Communicable Diseases Prevention Department, Saudi Public Health Authority, Riyadh 13354, Saudi Arabia
| | - Areeg N. K. Abdalla
- Department of Intensive Care Unit, Saudi German Hospital, Dammam 32313, Saudi Arabia
| | - Saad Alhumaid
- Administration of Pharmaceutical Care, Al-Ahsa Health Cluster, Ministry of Health, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
34
|
Alhinai Z, Park S, Choe YJ, Michelow IC. A global epidemiological analysis of COVID-19 vaccine types and clinical outcomes. Int J Infect Dis 2022; 124:206-211. [PMID: 36155824 PMCID: PMC9499984 DOI: 10.1016/j.ijid.2022.09.014] [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: 06/26/2022] [Revised: 08/30/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Objectives To compare messenger RNA (mRNA)–based and adenovirus-vectored vaccines (ADVVs) with inactivated virus vaccines (IVVs) using real-world aggregate data. Methods We performed longitudinal analyses of publicly accessible epidemiological, clinical, virological, vaccine-related, and other public health data from 41 eligible countries during the first half of 2021. The relationships between vaccination coverage and clinical outcomes were analyzed using repeated measures correlation analyses and mixed-effects modeling to adjust for potential mediating and confounding factors. Results Countries that used mRNA and/or ADVV (n = 31) vs IVV, among other vaccine types (n = 10), had different distributions of age (42.4 vs 33.9 years, respectively; P-value = 0.0006), gross domestic product per capita ($ 38,606 vs $ 20,422, respectively; P <0.0001), and population sizes (8,655,541 vs 5,139,162, respectively; P-value = 0.36). After adjustment for country differences, the stringency of nonpharmaceutical interventions, and dominant SARS-CoV-2 variant types, populations that received mRNA and/or ADVV had significantly lower rates of cases and deaths over time (P <0.001 for each analysis). Populations vaccinated with IVV, among others, had significantly higher rates of cases and deaths over time (P <0.05 for each analysis). Conclusion The real-world effectiveness of IVV may be inferior to mRNA and/or ADVV, and prospective comparative studies are needed to critically evaluate the role of IVV in the context of contemporary SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Zaid Alhinai
- Department of Child Health, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, 123, Oman.
| | - Sangshin Park
- Graduate School of Urban Public Health & Department of Urban Big Data Convergence, University of Seoul, Seoul, 02504, Republic of Korea
| | - Young-June Choe
- Department of Pediatrics, Korea University Anam Hospital, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Ian C Michelow
- Department of Pediatrics, Division of Infectious Diseases, The Warren Alpert Medical School of Brown University, Providence, 02903, Rhode Island, United States
| |
Collapse
|
35
|
SARS-CoV-2 Variants, Current Vaccines and Therapeutic Implications for COVID-19. Vaccines (Basel) 2022; 10:vaccines10091538. [PMID: 36146616 PMCID: PMC9504858 DOI: 10.3390/vaccines10091538] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Over the past two years, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused hundreds of millions of infections, resulting in an unprecedented pandemic of coronavirus disease 2019 (COVID-19). As the virus spreads through the population, ongoing mutations and adaptations are being discovered. There is now substantial clinical evidence that demonstrates the SARS-CoV-2 variants have stronger transmissibility and higher virulence compared to the wild-type strain of SARS-CoV-2. Hence, development of vaccines against SARS-CoV-2 variants to boost individual immunity has become essential. However, current treatment options are limited for COVID-19 caused by the SARS-CoV-2 variants. In this review, we describe current distribution, variation, biology, and clinical features of COVID-19 caused by SARS-CoV-2 variants (including Alpha (B.1.1.7 Lineage) variant, Beta (B.1.351 Lineage) variant, Gamma (P.1 Lineage) variant, Delta (B.1.617.2 Lineage) variant, and Omicron (B.1.1.529 Lineage) variant and others. In addition, we review currently employed vaccines in clinical or preclinical phases as well as potential targeted therapies in an attempt to provide better preventive and treatment strategies for COVID-19 caused by different SARS-CoV-2 variants.
Collapse
|
36
|
Young A. T cells in SARS-CoV-2 infection and vaccination. Ther Adv Vaccines Immunother 2022; 10:25151355221115011. [PMID: 36051003 PMCID: PMC9425900 DOI: 10.1177/25151355221115011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 07/04/2022] [Indexed: 12/24/2022] Open
Abstract
While antibodies garner the lion’s share of attention in SARS-CoV-2 immunity, cellular immunity (T cells) may be equally, if not more important, in controlling infection. Both CD8+ and CD4+ T cells are elicited earlier and are associated with milder disease, than antibodies, and T-cell activation appears to be necessary for control of infection. Variants of concern (VOCs) such as Omicron have escaped the neutralizing antibody responses after two mRNA vaccine doses, but T-cell immunity is largely intact. The breadth and patient-specific nature of the latter offers a formidable line of defense that can limit the severity of illness, and are likely to be responsible for most of the protection from natural infection or vaccination against VOCs which have evaded the antibody response. Comprehensive searches for T-cell epitopes, T-cell activation from infection and vaccination of specific patient groups, and elicitation of cellular immunity by various alternative vaccine modalities are here reviewed. Development of vaccines that specifically target T cells is called for, to meet the needs of patient groups for whom cellular immunity is weaker, such as the elderly and the immunosuppressed. While VOCs have not yet fully escaped T-cell immunity elicited by natural infection and vaccines, some early reports of partial escape suggest that future VOCs may achieve the dreaded result, dislodging a substantial proportion of cellular immunity, enough to cause a grave public health burden. A proactive, rather than reactive, solution which identifies and targets immutable sequences in SARS-CoV-2, not just those which are conserved, may be the only recourse humankind has to disarm these future VOCs before they disarm us.
Collapse
Affiliation(s)
- Arthur Young
- InvVax, 2265 E. Foohill Blvd., Pasadena, CA 91107, USA
| |
Collapse
|
37
|
Schultz BM, Melo-González F, Duarte LF, Gálvez NMS, Pacheco GA, Soto JA, Berríos-Rojas RV, González LA, Moreno-Tapia D, Rivera-Pérez D, Ríos M, Vázquez Y, Hoppe-Elsholz G, Andrade-Parra CA, Vallejos OP, Piña-Iturbe A, Iturriaga C, Urzua M, Navarrete MS, Rojas Á, Fasce R, Fernández J, Mora J, Ramírez E, Gaete-Argel A, Acevedo ML, Valiente-Echeverría F, Soto-Rifo R, Weiskopf D, Grifoni A, Sette A, Zeng G, Meng W, González-Aramundiz JV, González PA, Abarca K, Kalergis AM, Bueno SM. A Booster Dose of CoronaVac Increases Neutralizing Antibodies and T Cells that Recognize Delta and Omicron Variants of Concern. mBio 2022; 13:e0142322. [PMID: 35946814 PMCID: PMC9426482 DOI: 10.1128/mbio.01423-22] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/11/2022] [Indexed: 01/10/2023] Open
Abstract
CoronaVac is an inactivated SARS-CoV-2 vaccine approved by the World Health Organization (WHO). Previous studies reported increased levels of neutralizing antibodies and specific T cells 2 and 4 weeks after two doses of CoronaVac; these levels were significantly reduced at 6 to 8 months after the two doses. Here, we report the effect of a booster dose of CoronaVac on the anti-SARS-CoV-2 immune response generated against the variants of concern (VOCs), Delta and Omicron, in adults participating in a phase III clinical trial in Chile. Volunteers immunized with two doses of CoronaVac in a 4-week interval received a booster dose of the same vaccine between 24 and 30 weeks after the second dose. Neutralization capacities and T cell activation against VOCs Delta and Omicron were assessed 4 weeks after the booster dose. We observed a significant increase in neutralizing antibodies 4 weeks after the booster dose. We also observed a rise in anti-SARS-CoV-2-specific CD4+ T cells over time, and these cells reached a peak 4 weeks after the booster dose. Furthermore, neutralizing antibodies and SARS-CoV-2-specific T cells induced by the booster showed activity against VOCs Delta and Omicron. Our results show that a booster dose of CoronaVac increases adults' humoral and cellular anti-SARS-CoV-2 immune responses. In addition, immunity induced by a booster dose of CoronaVac is active against VOCs, suggesting adequate protection. IMPORTANCE CoronaVac is an inactivated vaccine against SARS-CoV-2 that has been approved by WHO for emergency use. Phase III clinical trials are in progress in several countries, including China, Brazil, Turkey, and Chile, and have shown safety and immunogenicity after two doses of the vaccine. This report characterizes immune responses induced by two doses of CoronaVac followed by a booster dose 5 months after the second dose in healthy Chilean adults. The data reported here show that a booster dose increased the immune responses against SARS-CoV-2, enhancing levels of neutralizing antibodies against the ancestral strain and VOCs. Similarly, anti-SARS-CoV-2 CD4+ T cell responses were increased following the booster dose. In contrast, levels of gamma interferon secretion and T cell activation against the VOCs Delta and Omicron were not significantly different from those for the ancestral strain. Therefore, a third dose of CoronaVac in a homologous vaccination schedule improves its immunogenicity in healthy volunteers.
Collapse
Affiliation(s)
- Bárbara M. Schultz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Luisa F. Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gaspar A. Pacheco
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Jorge A. Soto
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Roslye V. Berríos-Rojas
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Liliana A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Moreno-Tapia
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniela Rivera-Pérez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Ríos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yaneisi Vázquez
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Hoppe-Elsholz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade-Parra
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro Piña-Iturbe
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina Iturriaga
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Marcela Urzua
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María S. Navarrete
- Centro de Investigación Clínica UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Álvaro Rojas
- Centro de Investigación Clínica UC, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rodrigo Fasce
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Jorge Fernández
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Judith Mora
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Eugenio Ramírez
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Santiago, Chile
| | - Aracelly Gaete-Argel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mónica L. Acevedo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Fernando Valiente-Echeverría
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ricardo Soto-Rifo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, California, USA
| | | | | | - CoronaVac03CL Study Group
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Investigación Clínica UC, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Laboratorio Biomédico, Instituto de Salud Pública de Chile, Santiago, Chile
- Laboratorio de Virología Molecular y Celular, Programa de Virología, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, California, USA
- Sinovac Biotech, Beijing, China
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José V. González-Aramundiz
- Departamento de Farmacia, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Katia Abarca
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Enfermedades Infecciosas e Inmunología Pediátrica, División de Pediatría, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
38
|
Fujio K, Sung J, Nakatani S, Yamamoto K, Iwagami M, Fujimoto K, Shokirova H, Okumura Y, Akasaki Y, Nagino K, Midorikawa-Inomata A, Hirosawa K, Miura M, Huang T, Morooka Y, Kuwahara M, Murakami A, Inomata T. Characteristics and Clinical Ocular Manifestations in Patients with Acute Corneal Graft Rejection after Receiving the COVID-19 Vaccine: A Systematic Review. J Clin Med 2022; 11:jcm11154500. [PMID: 35956115 PMCID: PMC9369681 DOI: 10.3390/jcm11154500] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 02/04/2023] Open
Abstract
This study aimed to determine the characteristics and clinical ocular manifestations of acute corneal graft rejection after coronavirus disease 2019 (COVID-19) vaccination. We conducted an online search of the PubMed and EMBASE databases. Data on recipients’ characteristics, corneal transplantation types, interval between vaccination and allograft rejection, clinical manifestations, and graft rejection medication were extracted. Thirteen articles on 21 patients (23 eyes) with acute corneal graft rejection after COVID-19 vaccination, published between April and December 2021, were included. The median (interquartile range) age at the onset of rejection was 68 (27–83) years. Types of transplantation included penetrating keratoplasty (12 eyes), Descemet membrane endothelial keratoplasty (six eyes), Descemet stripping automated endothelial keratoplasty (four eyes), and living-related conjunctival-limbal allograft (one eye). The interval between vaccination and rejection ranged from 1 day to 6 weeks. Corneal edema was the leading clinical manifestation (20 eyes), followed by keratic precipitates (14 eyes) and conjunctival or ciliary injection (14 eyes). Medications included frequently applied topical corticosteroids (12 eyes), followed by a combination of topical and oral corticosteroids (four eyes). In addition, the clinical characteristics of corneal allograft rejection after COVID-19 vaccination were identified. Corneal transplant recipients may require further vaccination, necessitating appropriate management and treatment.
Collapse
Affiliation(s)
- Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Jaemyoung Sung
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| | - Satoru Nakatani
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
| | - Kazuko Yamamoto
- Department of Respiratory Medicine, Nagasaki University Hospital, Nagasaki 852-8102, Japan;
| | - Masao Iwagami
- Department of Health Services Research, Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan;
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Ken Nagino
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Akie Midorikawa-Inomata
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Tianxiang Huang
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Yuki Morooka
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan; (K.F.); (J.S.); (S.N.); (K.F.); (H.S.); (Y.O.); (Y.A.); (K.H.); (M.M.); (T.H.); (Y.M.); (M.K.); (A.M.)
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan;
- AI Incubation Farm, Juntendo University Graduate School of Medicine, Tokyo 113-0033, Japan
- Correspondence: ; Tel.: +81-3-3813-3111; Fax: +81-3-3817-0260
| |
Collapse
|
39
|
Petrone L, Picchianti-Diamanti A, Sebastiani GD, Aiello A, Laganà B, Cuzzi G, Vanini V, Gualano G, Grifoni A, Ferraioli M, Castilletti C, Meschi S, Vaia F, Nicastri E, Sette A, Goletti D. Humoral and cellular responses to spike of δ SARS-CoV-2 variant in vaccinated patients with immune-mediated inflammatory diseases. Int J Infect Dis 2022; 121:24-30. [PMID: 35462039 PMCID: PMC9023365 DOI: 10.1016/j.ijid.2022.04.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 04/12/2022] [Accepted: 04/16/2022] [Indexed: 01/28/2023] Open
Abstract
OBJECTIVES We assessed vaccination-induced antibody and cellular responses against spike from the ancestral strain and from the delta (δ) SARS-CoV-2 variant in patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressive therapy in comparison with immunocompetent subjects. METHODS We enrolled patients with IMID and immunocompetent subjects who completed the vaccination schedule within 4-6 months from the first dose. The interferon (IFN)-γ-response to spike peptides that were derived from the ancestral and the δ SARS-CoV-2 were measured by ELISA. Anti-Receptor Binding Domain IgG antibodies were also evaluated. RESULTS We enrolled 43 patients with IMID and nine immunocompetent subjects. No significant differences were found after comparing the specific immune response (IFN-γ) between patients with IMID and immunocompetent subjects to the ancestral (p = 0.36) or δ peptide pool (p = 0.51). Nevertheless, IFN-γ-specific responses to the ancestral or to the δ pools were reduced in subjects taking CTLA4-IgG or TNF-α inhibitors compared with subjects treated with IL-6 inhibitors or Disease Modifying Anti-Rheumatic Drugs. Regarding the antibody response, no significant differences were observed between patients with IMID and immunocompetent individuals. CONCLUSIONS Cellular responses to δ SARS-CoV-2 variant remain largely intact in patients with IMID. However, the magnitude of these responses is dependent on the specific IMID immunosuppressive regimen. Serological response was also similar between the IMID and control groups.
Collapse
Affiliation(s)
- Linda Petrone
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Andrea Picchianti-Diamanti
- Department of Clinical and Molecular Medicine, "Sapienza" University, S. Andrea University Hospital, 00189 Rome, Italy
| | | | - Alessandra Aiello
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Bruno Laganà
- Department of Clinical and Molecular Medicine, "Sapienza" University, S. Andrea University Hospital, 00189 Rome, Italy
| | - Gilda Cuzzi
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Valentina Vanini
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy; Unità Operativa Semplice (UOS) Professioni Sanitarie Tecniche, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Gina Gualano
- Respiratory Infectious Diseases Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Mario Ferraioli
- Rheumatology, Allergology and Clinical Immunology, Dipartimento di medicina dei sistemi, University of Rome Tor Vergata, Rome, Italy
| | - Concetta Castilletti
- Virology Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Silvia Meschi
- Virology Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Francesco Vaia
- UOC Direzione Sanitaria, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Emanuele Nicastri
- UOC Malattie Infettive ad Alta Intensità di Cura, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, 92037, USA
| | - Delia Goletti
- Translational Research Unit, National Institute for Infectious Diseases Lazzaro Spallanzani-IRCCS, 00149 Rome, Italy.
| |
Collapse
|
40
|
Cao L, Lou J, Chan SY, Zheng H, Liu C, Zhao S, Li Q, Mok CKP, Chan RWY, Chong MKC, Wu WKK, Chen Z, Wong ELY, Chan PKS, Zee BCY, Yeoh EK, Wang MH. Rapid evaluation of COVID-19 vaccine effectiveness against symptomatic infection with SARS-CoV-2 variants by analysis of genetic distance. Nat Med 2022; 28:1715-1722. [PMID: 35710987 PMCID: PMC9388371 DOI: 10.1038/s41591-022-01877-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 05/20/2022] [Indexed: 11/20/2022]
Abstract
Timely evaluation of the protective effects of Coronavirus Disease 2019 (COVID-19) vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants of concern is urgently needed to inform pandemic control planning. Based on 78 vaccine efficacy or effectiveness (VE) data from 49 studies and 1,984,241 SARS-CoV-2 sequences collected from 31 regions, we analyzed the relationship between genetic distance (GD) of circulating viruses against the vaccine strain and VE against symptomatic infection. We found that the GD of the receptor-binding domain of the SARS-CoV-2 spike protein is highly predictive of vaccine protection and accounted for 86.3% (P = 0.038) of the VE change in a vaccine platform-based mixed-effects model and 87.9% (P = 0.006) in a manufacturer-based model. We applied the VE-GD model to predict protection mediated by existing vaccines against new genetic variants and validated the results by published real-world and clinical trial data, finding high concordance of predicted VE with observed VE. We estimated the VE against the Delta variant to be 82.8% (95% prediction interval: 68.7-96.0) using the mRNA vaccine platform, closely matching the reported VE of 83.0% from an observational study. Among the four sublineages of Omicron, the predicted VE varied between 11.9% and 33.3%, with the highest VE predicted against BA.1 and the lowest against BA.2, using the mRNA vaccine platform. The VE-GD framework enables predictions of vaccine protection in real time and offers a rapid evaluation method against novel variants that may inform vaccine deployment and public health responses.
Collapse
Affiliation(s)
- Lirong Cao
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Jingzhi Lou
- Beth Bioinformatics Co. Ltd., Hong Kong SAR, China
| | - See Yeung Chan
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- Beth Bioinformatics Co. Ltd., Hong Kong SAR, China
| | - Hong Zheng
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Caiqi Liu
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shi Zhao
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Qi Li
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Chris Ka Pun Mok
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Renee Wan Yi Chan
- Department of Paediatrics, The Chinese University of Hong Kong, Hong Kong SAR, China
- Hong Kong Hub of Paediatric Excellence, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Marc Ka Chun Chong
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - William Ka Kei Wu
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zigui Chen
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Eliza Lai Yi Wong
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Health Systems and Policy Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul Kay Sheung Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
- Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Benny Chung Ying Zee
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Eng Kiong Yeoh
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China
- Centre for Health Systems and Policy Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Maggie Haitian Wang
- JC School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong SAR, China.
- CUHK Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
41
|
Reyes H, Diethelm-Varela B, Méndez C, Rebolledo-Zelada D, Lillo-Dapremont B, Muñoz SR, Bueno SM, González PA, Kalergis AM. Contribution of Two-Dose Vaccination Toward the Reduction of COVID-19 Cases, ICU Hospitalizations and Deaths in Chile Assessed Through Explanatory Generalized Additive Models for Location, Scale, and Shape. Front Public Health 2022; 10:815036. [PMID: 35968462 PMCID: PMC9364872 DOI: 10.3389/fpubh.2022.815036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 05/24/2022] [Indexed: 12/14/2022] Open
Abstract
Objectives To assess the impact of the initial two-dose-schedule mass vaccination campaign in Chile toward reducing adverse epidemiological outcomes due to SARS-CoV-2 infection. Methods Publicly available epidemiological data ranging from 3 February 2021 to 30 September 2021 were used to construct GAMLSS models that explain the beneficial effect of up to two doses of vaccination on the following COVID-19-related outcomes: new cases per day, daily active cases, daily occupied ICU beds and daily deaths. Results Administered first and second vaccine doses, and the statistical interaction between the two, are strong, statistically significant predictors for COVID-19-related new cases per day (R2 = 0.847), daily active cases (R2 = 0.903), ICU hospitalizations (R2 = 0.767), and deaths (R2 = 0.827). Conclusion Our models stress the importance of completing vaccination schedules to reduce the adverse outcomes during the pandemic. Future work will continue to assess the influence of vaccines, including booster doses, as the pandemic progresses, and new variants emerge. Policy Implications This work highlights the importance of attaining full (two-dose) vaccination status and reinforces the notion that a second dose provides increased non-additive protection. The trends we observed may also support the inclusion of booster doses in vaccination plans. These insights could contribute to guiding other countries in their vaccination campaigns.
Collapse
Affiliation(s)
- Humberto Reyes
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Benjamin Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Constanza Méndez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Diego Rebolledo-Zelada
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bastián Lillo-Dapremont
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sergio R. Muñoz
- Centro de Excelencia en Capacitación, Investigación y Gestión para la Salud Basada en Evidencia (CIGES), Facultad de Medicina, Universidad de La Frontera, Temuco, Chile
- Centro de Investigación en Epidemiología Cardiovascular y Nutricional (EPICYN), Facultad de Medicina, Universidad de La Frontera, Temuco, Chile
- Departamento de Salud Pública, Facultad de Medicina, Universidad de La Frontera, Temuco, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Alexis M. Kalergis
| |
Collapse
|
42
|
Zhao Z, Zhang X, Wang R, Wang Y, Gong L, Li C. Vaccine‐induced erythrodermic psoriasis in a child successfully treated with secukinumab: A case report and brief literature review. Dermatol Ther 2022; 35:e15684. [PMID: 35789520 DOI: 10.1111/dth.15684] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/24/2022] [Accepted: 07/03/2022] [Indexed: 11/27/2022]
Affiliation(s)
- Zhenkai Zhao
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| | - Xiaoning Zhang
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| | - Rui Wang
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| | - Youlin Wang
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| | - Lingling Gong
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| | - Chengxin Li
- Department of Dermatology First Medical Center of Chinese PLA General Hospital Beijing China
| |
Collapse
|
43
|
Xu K, Fan C, Han Y, Dai L, Gao GF. Immunogenicity, efficacy and safety of COVID-19 vaccines: an update of data published by 31 December 2021. Int Immunol 2022; 34:595-607. [PMID: 35778913 PMCID: PMC9278184 DOI: 10.1093/intimm/dxac031] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 06/30/2022] [Indexed: 02/01/2023] Open
Abstract
The unprecedented coronavirus disease 2019 (COVID-19) pandemic has caused a disaster for public health in the last 2 years, without any sign of an ending. Various vaccines were developed rapidly as soon as the outbreak occurred. Clinical trials demonstrated the reactogenicity, immunogenicity and protection efficacy in humans, and some of the vaccines have been approved for clinical use. However, waves of infections such as the recently circulating Omicron variant still occur. Newly emerging variants, especially the variants of concern, and waning humoral responses pose serious challenges to the control of the COVID-19 pandemic. Previously, we summarized the humoral and cellular immunity, safety profiles and protection efficacy of COVID-19 vaccines with clinical data published by 21 May 2021. In this review, we summarize and update the published clinical data of COVID-19 vaccines and candidates up to 31 December 2021.
Collapse
Affiliation(s)
- Kun Xu
- Research Network of Immunity and Health (RNIH), Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China,Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan, China
| | - Chunxiang Fan
- National Immunization Programme, Chinese Center for Diseases Control and Prevention, Beijing, China
| | - Yuxuan Han
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China
| | - Lianpan Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Tropical Medicine and Laboratory Medicine, The First Affiliated Hospital, Hainan Medical University, Hainan, China,Savaid Medical School, University of Chinese Academy of Sciences, Beijing, China,CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
44
|
Moga E, Lynton-Pons E, Domingo P. The Robustness of Cellular Immunity Determines the Fate of SARS-CoV-2 Infection. Front Immunol 2022; 13:904686. [PMID: 35833134 PMCID: PMC9271749 DOI: 10.3389/fimmu.2022.904686] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/27/2022] [Indexed: 12/11/2022] Open
Abstract
Two years after the appearance of the SARS-CoV-2 virus, the causal agent of the current global pandemic, it is time to analyze the evolution of the immune protection that infection and vaccination provide. Cellular immunity plays an important role in limiting disease severity and the resolution of infection. The early appearance, breadth and magnitude of SARS-CoV-2 specific T cell response has been correlated with disease severity and it has been thought that T cell responses may be sufficient to clear infection with minimal disease in COVID-19 patients with X-linked or autosomal recessive agammaglobulinemia. However, our knowledge of the phenotypic and functional diversity of CD8+ cytotoxic lymphocytes, CD4+ T helper cells, mucosal-associated invariant T (MAIT) cells and CD4+ T follicular helper (Tfh), which play a critical role in infection control as well as long-term protection, is still evolving. It has been described how CD8+ cytotoxic lymphocytes interrupt viral replication by secreting antiviral cytokines (IFN-γ and TNF-α) and directly killing infected cells, negatively correlating with stages of disease progression. In addition, CD4+ T helper cells have been reported to be key pieces, leading, coordinating and ultimately regulating antiviral immunity. For instance, in some more severe COVID-19 cases a dysregulated CD4+ T cell signature may contribute to the greater production of pro-inflammatory cytokines responsible for pathogenic inflammation. Here we discuss how cellular immunity is the axis around which the rest of the immune system components revolve, since it orchestrates and leads antiviral response by regulating the inflammatory cascade and, as a consequence, the innate immune system, as well as promoting a correct humoral response through CD4+ Tfh cells. This review also analyses the critical role of cellular immunity in modulating the development of high-affinity neutralizing antibodies and germinal center B cell differentiation in memory and long-lived antibody secreting cells. Finally, since there is currently a high percentage of vaccinated population and, in some cases, vaccine booster doses are even being administered in certain countries, we have also summarized newer approaches to long-lasting protective immunity and the cross-protection of cellular immune response against SARS-CoV-2.
Collapse
Affiliation(s)
- Esther Moga
- Department of Immunology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain,*Correspondence: Esther Moga,
| | - Elionor Lynton-Pons
- Department of Immunology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Pere Domingo
- Unidad de enfermedades infecciosas, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| |
Collapse
|
45
|
Immunological Study of Combined Administration of SARS-CoV-2 DNA Vaccine and Inactivated Vaccine. Vaccines (Basel) 2022; 10:vaccines10060929. [PMID: 35746536 PMCID: PMC9228235 DOI: 10.3390/vaccines10060929] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Objective: We constructed two DNA vaccines containing the receptor-binding domain (RBD) genes of multiple SARS-CoV-2 variants and used them in combination with inactivated vaccines in a variety of different protocols to explore potential novel immunization strategies against SARS-CoV-2 variants. Methods: Two DNA vaccine candidates with different signal peptides (namely, secreted and membrane signal peptides) and RBD protein genes of different SARS-CoV-2 strains (Wuhan-Hu-1, B.1.351, B.1.617.2, C.37) were used. Four different combinations of DNA and inactivated vaccines were tested, namely, Group A: three doses of DNA vaccine; B: three doses of DNA vaccine and one dose of inactivated vaccine; C: two doses of inactivated vaccine and one dose of DNA vaccine; and D: coadministration of DNA and inactivated vaccines in two doses. Subgroups were grouped according to the signal peptide used (subgroup 1 contained secreted signal peptides, and subgroup 2 contained membrane signal peptides). The in vitro expression of the DNA vaccines, the humoral and cellular immunity responses of the immunized mice, the immune cell population changes in local lymph nodes, and proinflammatory cytokine levels in serum samples were evaluated. Results: The antibody responses and cellular immunity in Group A were weak for all SARS-CoV-2 strains; for Group B, there was a great enhancement of neutralizing antibody (Nab) titers against the B.1.617.2 variant strain. Group C showed a significant increase in antibody responses (NAb titers against the Wuhan-Hu-1 strain were 768 and 1154 for Group C1 and Group C2, respectively, versus 576) and cellular immune responses, especially for variant B.1.617.2 (3240 (p < 0.001) and 2430 (p < 0.05) for Group C1 and Group C2, versus 450); Group D showed an improvement in immunogenicity. Group C induced higher levels of multiple cytokines. Conclusion: The DNA vaccine candidates we constructed, administered as boosters, could enhance the humoral and cellular immune responses of inactivated vaccines against COVID-19, especially for B.1.617.2.
Collapse
|
46
|
Tarke A, Grifoni A, Sette A. Bioinformatic and Experimental Analysis of T Cell Immune Reactivity to SARS-CoV-2 and its Variants. FRONTIERS IN BIOINFORMATICS 2022; 2:876380. [PMID: 36304267 PMCID: PMC9580847 DOI: 10.3389/fbinf.2022.876380] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/12/2022] [Indexed: 02/05/2023] Open
Abstract
Definition of the T cells responses to SARS-CoV-2 and associated variants is critical to understanding the complexity of adaptive immunity against SARS-CoV-2 infection. Several groups have investigated the T cells responses by both experimental and bioinformatical approaches. Here we summarize recent findings on CD4 and CD8 T cell responses to SARS-CoV-2 with particular emphasis on emerging variants of concern, consolidating the results on the impact of SARS-CoV-2 variants on T cell responses by performing an additional metanalysis emphasizing the lower impact of variant mutations in dominant T cell epitopes. The consensus is that the majority of T cell responses are conserved across all current SARS-CoV-2 variants, including Delta and Omicron. Thus, even in concomitance with reduced antibody and B cell responses, T cells can still provide a second line of antiviral immunity.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
- Department of Internal Medicine and Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, San Diego, CA, United States
- *Correspondence: Alessandro Sette,
| |
Collapse
|
47
|
Liwsrisakun C, Pata S, Laopajon W, Takheaw N, Chaiwong W, Inchai J, Pothirat C, Bumroongkit C, Deesomchok A, Theerakittikul T, Limsukon A, Tajarernmuang P, Niyatiwatchanchai N, Trongtrakul K, Chuensirikulchai K, Kasinrerk W. Neutralizing antibody and T cell responses against SARS-CoV-2 variants of concern following ChAdOx-1 or BNT162b2 boosting in the elderly previously immunized with CoronaVac vaccine. Immun Ageing 2022; 19:24. [PMID: 35610643 PMCID: PMC9126751 DOI: 10.1186/s12979-022-00279-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/07/2022] [Indexed: 12/16/2022]
Abstract
Background The existence of SARS-CoV-2 variants of concern (VOCs) in association with evidence of breakthrough infections despite vaccination resulted in the need for vaccine boosting. In elderly individuals, information on the immunogenicity of booster vaccinations is limited. In countries where the CoronaVac inactivated vaccine is the primary vaccine, the appropriate boosting regimen is not clear. Immunologic studies of the effects of booster vaccination against VOCs, particularly Delta and Omicron, following CoronaVac in elderly individuals are helpful for policy makers. In this study, we determined the immune responses against VOCs following ChAdOx-1 or BNT162b2 boosting in elderly individuals previously immunized with CoronaVac. Results Before boosting, the median % inhibition of neutralizing antibodies (NAbs) against the wild-type (WT), Alpha, Beta, Delta and Omicron variants in the ChAdOx-1 and BNT162b2 groups was 52.8% vs. 53.4, 36.6% vs. 39.9, 5.2% vs. 13.7, 34.3% vs. 44.9, and 20.8% vs. 18.8%, respectively. After boosting with ChAdOx-1 or BNT162b2, the % inhibition of NAbs were increased to 97.3% vs. 97.4, 94.3% vs. 97.3%, 79.9 vs. 93.7, 95.5% vs. 97.5, and 26.9% vs. 31.9% for WT, Alpha, Beta, Delta and Omicron variants, respectively. Boosting with BNT162b2 induced significantly higher NAb levels than boosting with ChAdOx-1 against the Alpha, Beta and Delta variants but not the WT and Omicron variants. NAb levels against Omicron variant were not significantly different before and after boosting with ChAdOx-1 or BNT162b2. To evaluate T-cell responses, S peptides of the WT, Alpha, Beta and Delta variants were used to stimulate T cells. Upon stimulation, the expression of IL-17A in CD8 T cells was higher in the BNT162b2 group than in the ChAdOx-1 boosting group. However, IFN-γ production in CD4 and CD8 T cells did not significantly differ under all vaccination regimens. The expression of FasL in CD4 T cells, but not CD8 T cells, was higher in the BNT162b2-boosted group. Conclusion Boosting with either ChAdOx-1 or BNT162b2 in CoronaVac-primed healthy elderly individuals induced high NAb production against all examined VOCs except Omicron. BNT162b2 stimulated higher NAb and some T-cell responses than ChAdOx-1. Vaccine boosting is, therefore, recommended for elderly individuals previously immunized with CoronaVac. Supplementary Information The online version contains supplementary material available at 10.1186/s12979-022-00279-8.
Collapse
Affiliation(s)
- Chalerm Liwsrisakun
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Supansa Pata
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Witida Laopajon
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Nuchjira Takheaw
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Warawut Chaiwong
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Juthamas Inchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaicharn Pothirat
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chaiwat Bumroongkit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Athavudh Deesomchok
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Theerakorn Theerakittikul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Atikun Limsukon
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Pattraporn Tajarernmuang
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nutchanok Niyatiwatchanchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Konlawij Trongtrakul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kantinan Chuensirikulchai
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Watchara Kasinrerk
- Division of Clinical Immunology, Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand. .,Biomedical Technology Research Center, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency at the Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
48
|
Isaeva OI, Ketelaars SLC, Kvistborg P. In Silico Analysis Predicts a Limited Impact of SARS-CoV-2 Variants on CD8 T Cell Recognition. Front Immunol 2022; 13:891524. [PMID: 35572563 PMCID: PMC9094405 DOI: 10.3389/fimmu.2022.891524] [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: 03/07/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Since the start of the COVID-19 pandemic, mutations have led to the emergence of new SARS-CoV-2 variants, and some of these have become prominent or dominant variants of concern. This natural course of development can have an impact on how protective the previously naturally or vaccine induced immunity is. Therefore, it is crucial to understand whether and how variant specific mutations influence host immunity. To address this, we have investigated how mutations in the recent SARS-CoV-2 variants of interest and concern influence epitope sequence similarity, predicted binding affinity to HLA, and immunogenicity of previously reported SARS-CoV-2 CD8 T cell epitopes. Our data suggests that the vast majority of SARS-CoV-2 CD8 T cell recognized epitopes are not altered by variant specific mutations. Interestingly, for the CD8 T cell epitopes that are altered due to variant specific mutations, our analyses show there is a high degree of sequence similarity between mutated and reference SARS-CoV-2 CD8 T cell epitopes. However, mutated epitopes, primarily derived from the spike protein, in SARS-CoV-2 variants Delta, AY.4.2 and Mu display reduced predicted binding affinity to their restriction element. These findings indicate that the recent SARS-CoV-2 variants of interest and concern have limited ability to escape memory CD8 T cell responses raised by vaccination or prior infection with SARS-CoV-2 early in the pandemic. The overall low impact of the mutations on CD8 T cell cross-recognition is in accordance with the notion that mutations in SARS-CoV-2 are primarily the result of receptor binding affinity and antibody selection pressures exerted on the spike protein, unrelated to T cell immunity.
Collapse
Affiliation(s)
- Olga I Isaeva
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands.,Department of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Steven L C Ketelaars
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Pia Kvistborg
- Department of Molecular Oncology and Immunology, Netherlands Cancer Institute, Amsterdam, Netherlands
| |
Collapse
|
49
|
Sette A, Saphire EO. Inducing broad-based immunity against viruses with pandemic potential. Immunity 2022; 55:738-748. [PMID: 35545026 PMCID: PMC10286218 DOI: 10.1016/j.immuni.2022.04.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 02/08/2023]
Abstract
The brutal toll of another viral pandemic can be blunted by investing now in research that uncovers mechanisms of broad-based immunity so we may have vaccines and therapeutics at the ready. We do not know exactly what pathogen may trigger the next wave or next pandemic. We do know, however, that the human immune system must respond and must be bolstered with effective vaccines and other therapeutics to preserve lives and livelihoods. These countermeasures must focus on features conserved among families of pathogens in order to be responsive against something yet to emerge. Here, we focus on immunological approaches to mitigate the impact of the next emerging virus pandemic by developing vaccines that elicit both broadly protective antibodies and T cells. Identifying human immune mechanisms of broad protection against virus families with pandemic potential will be our best defense for humanity in the future.
Collapse
Affiliation(s)
- Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
| | - Erica Ollmann Saphire
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA.
| |
Collapse
|
50
|
Lu L, Chen LL, Zhang RRQ, Tsang OTY, Chan JMC, Tam AR, Leung WS, Chik TSH, Lau DPL, Choi CYC, Fong CHY, Cai JP, Tsoi HW, Choi CYK, Zhang X, Abdullah SMU, Chan BPC, Chan KH, Yuen KY, Hung IFN, To KKW. Boosting of serum neutralizing activity against the Omicron variant among recovered COVID-19 patients by BNT162b2 and CoronaVac vaccines. EBioMedicine 2022; 79:103986. [PMID: 35398786 PMCID: PMC8989491 DOI: 10.1016/j.ebiom.2022.103986] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/17/2022] [Accepted: 03/22/2022] [Indexed: 12/05/2022] Open
Abstract
Background SARS-CoV-2 Omicron variant evades immunity from past infection or vaccination and is associated with a greater risk of reinfection among recovered COVID-19 patients. We assessed the serum neutralizing antibody (NAb) activity against Omicron variant (Omicron NAb) among recovered COVID-19 patients with or without vaccination. Methods In this prospective cohort study with 135 recovered COVID-19 patients, we determined the serum NAb titers against ancestral virus or variants using a live virus NAb assay. We used the receiver operating characteristic analysis to determine the optimal cutoff for a commercially-available surrogate NAb assay. Findings Among recovered COVID-19 patients, the serum live virus geometric mean Omicron NAb titer was statistically significantly higher among BNT162b2 recipients compared to non-vaccinated individuals (85.4 vs 5.6,P < 0.0001). The Omicron seropositive rates in live virus NAb test (NAb titer ≥10) were statistically significantly higher among BNT162b2 (90.6% [29/32];P < 0.0001) or CoronaVac (36.7% [11/30]; P = 0.0115) recipients when compared with non-vaccinated individuals (12.3% [9/73]). Subgroup analysis of CoronaVac recipients showed that the Omicron seropositive rates were higher among individuals with two doses than those with one dose (85.7% vs 21.7%; P = 0.0045). For the surrogate NAb assay, a cutoff of 109.1 AU/ml, which is 7.3-fold higher than the manufacturer's recommended cutoff, could achieve a sensitivity and specificity of 89.5% and 89.8%, respectively, in detecting Omicron NAb. Interpretation Among individuals with prior COVID-19, one dose of BNT162b2 or two doses of CoronaVac could induce detectable serum Omicron NAb. Our result would be particularly important for guiding vaccine policies in countries with COVID-19 vaccine shortage. Funding Health and Medical Research Fund, Richard and Carol Yu, Michael Tong (see acknowledgments for full list).
Collapse
Affiliation(s)
- Lu Lu
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Lin-Lei Chen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Ricky Rui-Qi Zhang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Owen Tak-Yin Tsang
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Jacky Man-Chun Chan
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Anthony Raymond Tam
- Department of Medicine, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Wai-Shing Leung
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Thomas Shiu-Hong Chik
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Daphne Pui-Ling Lau
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Chris Yau-Chung Choi
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong Special Administrative Region, China
| | - Carol Ho-Yan Fong
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Jian-Piao Cai
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Hoi-Wah Tsoi
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Charlotte Yee-Ki Choi
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Xiaojuan Zhang
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Syed Muhammad Umer Abdullah
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Brian Pui-Chun Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kwok-Hung Chan
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Kwok-Yung Yuen
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ivan Fan-Ngai Hung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Department of Medicine, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China
| | - Kelvin Kai-Wang To
- State Key Laboratory for Emerging Infectious Diseases, Carol Yu Centre for Infection, Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong Special Administrative Region, China; Department of Microbiology, Queen Mary Hospital, Pokfulam, Hong Kong Special Administrative Region, China; Centre for Virology, Vaccinology and Therapeutics, Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China.; Department of Clinical Microbiology and Infection Control, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|