1
|
Roa CC, de Los Reyes MRA, Plennevaux E, Smolenov I, Hu B, Gao F, Ilagan H, Ambrosino D, Siber G, Clemens R, Han HH. SCB-2019 protein vaccine as heterologous booster of neutralizing activity against SARS-CoV-2 Omicron variants after immunization with other COVID-19 vaccines. Hum Vaccin Immunother 2024; 20:2301632. [PMID: 38206168 DOI: 10.1080/21645515.2023.2301632] [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: 09/12/2023] [Accepted: 12/31/2023] [Indexed: 01/12/2024] Open
Abstract
We assessed the non-inferiority of homologous boosting compared with heterologous boosting with the recombinant protein vaccine, SCB-2019, in adults previously immunized with different COVID-19 vaccines. Three equal cohorts (N ~ 420) of Philippino adults (18-80 years) previously immunized with Comirnaty, CoronaVac or Vaxzevria COVID-19 vaccines were randomized 1:1 to receive homologous or heterologous (SCB-2019) boosters. Neutralizing antibodies against prototype SARS-CoV-2 (Wuhan-Hu-1) were measured in all participants and against Delta variant and Omicron sub-lineages in subsets (30‒50 per arm) 15 days after boosting. Participants recorded solicited adverse events for 7 days and unsolicited and serious adverse events until Day 60. Prototype SARS-CoV-2 neutralizing responses on Day 15 after SCB-2019 were statistically non-inferior to homologous Vaxzevria boosters, superior to CoronaVac, but lower than homologous Comirnaty. Neutralizing responses against Delta and Omicron BA.1, BA.2, BA.4 and BA.5 variants after heterologous SCB-2019 were higher than homologous CoronaVac or Vaxzevria, but lower than homologous Comirnaty. Responses against Omicron BF.7, BQ.1.1.3, and XBB1.5 following heterologous SCB-2019 were lower than after homologous Comirnaty booster but significantly higher than after Vaxzevria booster. SCB-2019 reactogenicity was similar to CoronaVac or Vaxzevria, but lower than Comirnaty; most frequent events were mild/moderate injection site pain, headache and fatigue. No vaccine-related serious adverse events were reported. Heterologous SCB-2019 boosting was well tolerated and elicited neutralizing responses against all tested SARS-COV-2 viruses including Omicron BA.1, BA.2, BA.4, BA.5, BF.7, BQ.1.1.3, and XBB1.5 sub-lineages that were non-inferior to homologous boosting with CoronaVac or Vaxzevria, but not homologous Comirnaty booster.
Collapse
Affiliation(s)
- Camilo C Roa
- Department of Physiology, Philippine General Hospital, University of the Philippines, Manila, Philippines
| | | | - Eric Plennevaux
- Clinical Development, Clover Biopharmaceuticals, Cambridge, UK
| | - Igor Smolenov
- Clinical Development, Clover Biopharmaceuticals, Boston, MA, USA
| | - Branda Hu
- Clinical Development, Clover Biopharmaceuticals, Boston, MA, USA
| | - Faith Gao
- Clinical Development, Clover Biopharmaceuticals, Boston, MA, USA
| | - Hannalyn Ilagan
- Clinical Development, Clover Biopharmaceuticals, Boston, MA, USA
| | | | | | - Ralf Clemens
- Global Research in Infectious Diseases, Rio de Janeiro, Brazil
| | - Htay Htay Han
- Clinical Development, Clover Biopharmaceuticals, Boston, MA, USA
| |
Collapse
|
2
|
Dolzhikova IV, Tukhvatulin AI, Grousova DM, Zorkov ID, Komyakova ME, Ilyukhina AA, Kovyrshina AV, Shelkov AY, Botikov AG, Samokhvalova EG, Reshetnikov DA, Siniavin AE, Savina DM, Shcheblyakov DV, Izhaeva FM, Dzharullaeva AS, Erokhova AS, Popova O, Ozharovskaya TA, Zrelkin DI, Goldovskaya PP, Semikhin AS, Zubkova OV, Nedorubov AA, Gushchin VA, Naroditsky BS, Logunov DY, Gintsburg AL. Immunogenicity and Protectivity of Sputnik V Vaccine in hACE2-Transgenic Mice against Homologous and Heterologous SARS-CoV-2 Lineages Including Far-Distanced Omicron BA.5. Vaccines (Basel) 2024; 12:1152. [PMID: 39460319 PMCID: PMC11512357 DOI: 10.3390/vaccines12101152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND The SARS-CoV-2 virus continuously acquires mutations, leading to the emergence of new variants. Notably, the effectiveness of global vaccination efforts has significantly declined with the rise and spread of the B.1.1.529 (Omicron) variant. METHODS The study used virological, immunological and histological research methods, as well as methods of working with laboratory animals. In this study, we evaluated the Gam-COVID-Vac (Sputnik V), an adenoviral vaccine developed by the N.F. Gamaleya National Research Center for Epidemiology and Microbiology, and conducted experiments on hemizygous K18-ACE2-transgenic F1 mice. The variants studied included B.1.1.1, B.1.1.7, B.1.351, B.1.1.28/P.1, B.1.617.2, and B.1.1.529 BA.5. RESULTS Our findings demonstrate that the Sputnik V vaccine elicits a robust humoral and cellular immune response, effectively protecting vaccinated animals from challenges posed by various SARS-CoV-2 variants. However, we observed a notable reduction in vaccine efficacy against the B.1.1.529 (Omicron BA.5) variant. CONCLUSIONS Our results indicate that ongoing monitoring of emerging mutations is crucial to assess vaccine efficacy against new SARS-CoV-2 variants to identify those with pandemic potential. If protective efficacy declines, it will be imperative to develop new vaccines tailored to current variants of the virus.
Collapse
Affiliation(s)
- Inna V. Dolzhikova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Amir I. Tukhvatulin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Daria M. Grousova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Ilya D. Zorkov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Marina E. Komyakova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Anna A. Ilyukhina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Anna V. Kovyrshina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Artem Y. Shelkov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Andrey G. Botikov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Ekaterina G. Samokhvalova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Dmitrii A. Reshetnikov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Andrey E. Siniavin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Daria M. Savina
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Dmitrii V. Shcheblyakov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Fatima M. Izhaeva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Alina S. Dzharullaeva
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Alina S. Erokhova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Olga Popova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Tatiana A. Ozharovskaya
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Denis I. Zrelkin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Polina P. Goldovskaya
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Alexander S. Semikhin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Olga V. Zubkova
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Andrey A. Nedorubov
- Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” (Sechenov University), Ministry of Health, Russian Federation, 119991 Moscow, Russia
| | - Vladimir A. Gushchin
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Boris S. Naroditsky
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Denis Y. Logunov
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
| | - Alexander L. Gintsburg
- Federal State Budget Institution “National Research Centre for Epidemiology and Microbiology Named after Honorary Academician N F Gamaleya”, Ministry of Health, Russian Federation, 123098 Moscow, Russia
- Federal State Autonomous Educational Institution of Higher Education “I.M. Sechenov First Moscow State Medical University” (Sechenov University), Ministry of Health, Russian Federation, 119991 Moscow, Russia
| |
Collapse
|
3
|
Tong X, Wang Q, Jung W, Chicz TM, Blanc R, Parker LJ, Barouch DH, McNamara RP. Compartment-specific antibody correlates of protection to SARS-CoV-2 Omicron in macaques. iScience 2024; 27:110174. [PMID: 39224511 PMCID: PMC11367469 DOI: 10.1016/j.isci.2024.110174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/09/2024] [Accepted: 05/31/2024] [Indexed: 09/04/2024] Open
Abstract
Antibodies represent a primary mediator of protection against respiratory viruses. Serum neutralizing antibodies (NAbs) are often considered a primary correlate of protection. However, detailed antibody profiles including characterization of antibody functions in different anatomic compartments are poorly understood. Here we show that antibody correlates of protection against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) challenge are different in systemic versus mucosal compartments in rhesus macaques. In serum, NAbs were the strongest correlate of protection and linked to spike-specific binding antibodies and other extra-NAb functions that create a larger protective network. In bronchiolar lavage (BAL), antibody-dependent cellular phagocytosis (ADCP) proved the strongest correlate of protection rather than NAbs. Within BAL, ADCP was linked to mucosal spike-specific immunoglobulin (Ig)G, IgA/secretory IgA, and Fcγ-receptor binding antibodies. Our results support a model in which antibodies with different functions mediate protection at different anatomic sites.
Collapse
Affiliation(s)
- Xin Tong
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Qixin Wang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Wonyeong Jung
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Taras M. Chicz
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Ross Blanc
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Lily J. Parker
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Dan H. Barouch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ryan P. McNamara
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA 02139, USA
| |
Collapse
|
4
|
Camacho-Moll ME, Mata-Tijerina VL, Gutiérrez-Salazar CC, Silva-Ramírez B, Peñuelas-Urquides K, González-Escalante L, Escobedo-Guajardo BL, Cruz-Luna JE, Corrales-Pérez R, Gómez-García S, Bermúdez-de León M. The impact of comorbidity status in COVID-19 vaccines effectiveness before and after SARS-CoV-2 omicron variant in northeastern Mexico: a retrospective multi-hospital study. Front Public Health 2024; 12:1402527. [PMID: 38932780 PMCID: PMC11199416 DOI: 10.3389/fpubh.2024.1402527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Introduction The end of the coronavirus disease 2019 (COVID-19) pandemic has been declared by the World Health Organization on May 5, 2023. Several vaccines were developed, and new data is being published about their effectiveness. However, the clinical trials for the vaccines were performed before the Omicron variant appeared and there are population groups where vaccine effectiveness still needs to be tested. The overarching goal of the present study was to analyze the effects of COVID-19 vaccination before and after the Omicron variant in patients considering comorbidities in a population from Nuevo Leon, Mexico. Methods Epidemiological COVID-19 data from the Mexican Social Security Institute were collected from 67 hospitals located in northeastern Mexico, from July 2020 to May 2023, and a total of 669,393 cases were compiled, 255,819 reported a SARS-CoV-2 positive reverse transcription quantitative polymerase chain reaction (RT-qPCR) test or a positive COVID-19 antigen rapid test. Results Before Omicron (BO, 2020-2021), after 14 days of two doses of COVID-19 vaccine, BNT162b2 and ChAdOx1 vaccines were effective against infection in non-comorbid and all comorbid subgroups, whereas after Omicron (AO, 2022- 2023) there was no significant effectiveness against infection with none of the vaccines. Regarding hospitalization BO, BNT162b2, ChAdOx1, CoronaVac and mRNA-1273 significantly protected non-comorbid patients whereas BNT162b2, ChAdOx1, and mRNA-1273, protected all comorbid subgroups against hospitalization. AO, BNT162b2, ChAdOx1, CoronaVac and mRNA-1273 were effective against hospitalization in non-comorbid patients whereas for most comorbid subgroups BNT162b2, ChAdOx1 and CoronaVac were effective against hospitalization. Non-comorbid patients were protected against death as an outcome of COVID-19 during the BO period with most vaccines whereas a reduction in effectiveness was observed AO with mRNA-1273 vaccines in patients with hypertension, and diabetes mellitus. Discussion BO, COVID-19 vaccines were effective against infection, hospitalization, and death whereas AO, COVID-19 vaccines failed to protect the population from COVID-19 infection. A varying effectiveness against hospitalization and death is observed AO.
Collapse
Affiliation(s)
- Maria Elena Camacho-Moll
- Laboratory of Molecular Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Viviana Leticia Mata-Tijerina
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | | | - Beatriz Silva-Ramírez
- Laboratory of Immunogenetics, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Katia Peñuelas-Urquides
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Laura González-Escalante
- Laboratory of Molecular Microbiology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Brenda Leticia Escobedo-Guajardo
- Laboratory of Molecular Research of Diseases, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Jorge Eleazar Cruz-Luna
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Roberto Corrales-Pérez
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Salvador Gómez-García
- Medical Epidemiological Assistance Coordination of the State of Nuevo Leon, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| | - Mario Bermúdez-de León
- Laboratory of Molecular Biology, Northeast Biomedical Research Center, Mexican Social Security Institute, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
5
|
Hồ NT, Hughes SG, Ta VT, Phan LT, Đỗ Q, Nguyễn TV, Phạm ATV, Thị Ngọc Đặng M, Nguyễn LV, Trịnh QV, Phạm HN, Chử MV, Nguyễn TT, Lương QC, Tường Lê VT, Nguyễn TV, Trần LTL, Thi Van Luu A, Nguyen AN, Nguyen NTH, Vu HS, Edelman JM, Parker S, Sullivan B, Sullivan S, Ruan Q, Clemente B, Luk B, Lindert K, Berdieva D, Murphy K, Sekulovich R, Greener B, Smolenov I, Chivukula P, Nguyễn VT, Nguyen XH. Safety, immunogenicity and efficacy of the self-amplifying mRNA ARCT-154 COVID-19 vaccine: pooled phase 1, 2, 3a and 3b randomized, controlled trials. Nat Commun 2024; 15:4081. [PMID: 38744844 PMCID: PMC11094049 DOI: 10.1038/s41467-024-47905-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Combination of waning immunity and lower effectiveness against new SARS-CoV-2 variants of approved COVID-19 vaccines necessitates new vaccines. We evaluated two doses, 28 days apart, of ARCT-154, a self-amplifying mRNA COVID-19 vaccine, compared with saline placebo in an integrated phase 1/2/3a/3b controlled, observer-blind trial in Vietnamese adults (ClinicalTrial.gov identifier: NCT05012943). Primary safety and reactogenicity outcomes were unsolicited adverse events (AE) 28 days after each dose, solicited local and systemic AE 7 days after each dose, and serious AEs throughout the study. Primary immunogenicity outcome was the immune response as neutralizing antibodies 28 days after the second dose. Efficacy against COVID-19 was assessed as primary and secondary outcomes in phase 3b. ARCT-154 was well tolerated with generally mild-moderate transient AEs. Four weeks after the second dose 94.1% (95% CI: 92.1-95.8) of vaccinees seroconverted for neutralizing antibodies, with a geometric mean-fold rise from baseline of 14.5 (95% CI: 13.6-15.5). Of 640 cases of confirmed COVID-19 eligible for efficacy analysis most were due to the Delta (B.1.617.2) variant. Efficacy of ARCT-154 was 56.6% (95% CI: 48.7- 63.3) against any COVID-19, and 95.3% (80.5-98.9) against severe COVID-19. ARCT-154 vaccination is well tolerated, immunogenic and efficacious, particularly against severe COVID-19 disease.
Collapse
Affiliation(s)
- Nhân Thị Hồ
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | | | | | | | - Quyết Đỗ
- Vietnam Military Medical University, Hanoi, Vietnam
| | | | | | | | | | | | | | - Mến Văn Chử
- Vietnam Military Medical University, Hanoi, Vietnam
| | | | | | | | | | - Lý-Thi-Lê Trần
- Hi-tech Center, Vinmec Healthcare System, Hanoi, Vietnam
- Vietnam Biocare Biotechnology Jointstock Company, Hanoi, Vietnam
| | - Anh Thi Van Luu
- Vietnam Biocare Biotechnology Jointstock Company, Hanoi, Vietnam
| | - Anh Ngoc Nguyen
- Vietnam Biocare Biotechnology Jointstock Company, Hanoi, Vietnam
| | | | - Hai-Son Vu
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam
| | | | | | | | | | - Qian Ruan
- Arcturus Therapeutics, Inc, San Diego, CA, USA
| | | | - Brian Luk
- Arcturus Therapeutics, Inc, San Diego, CA, USA
| | | | | | - Kat Murphy
- Arcturus Therapeutics, Inc, San Diego, CA, USA
| | | | | | | | | | - Vân Thu Nguyễn
- Vietnam Biocare Biotechnology Jointstock Company, Hanoi, Vietnam
| | - Xuan-Hung Nguyen
- Vinmec-VinUni Institute of Immunology, Vinmec Healthcare System, Hanoi, Vietnam.
- Hi-tech Center, Vinmec Healthcare System, Hanoi, Vietnam.
- College of Health Sciences, Vin University, Hanoi, Vietnam.
| |
Collapse
|
6
|
Kumar A, Tripathi P, Kumar P, Shekhar R, Pathak R. From Detection to Protection: Antibodies and Their Crucial Role in Diagnosing and Combatting SARS-CoV-2. Vaccines (Basel) 2024; 12:459. [PMID: 38793710 PMCID: PMC11125746 DOI: 10.3390/vaccines12050459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Understanding the antibody response to SARS-CoV-2, the virus responsible for COVID-19, is crucial to comprehending disease progression and the significance of vaccine and therapeutic development. The emergence of highly contagious variants poses a significant challenge to humoral immunity, underscoring the necessity of grasping the intricacies of specific antibodies. This review emphasizes the pivotal role of antibodies in shaping immune responses and their implications for diagnosing, preventing, and treating SARS-CoV-2 infection. It delves into the kinetics and characteristics of the antibody response to SARS-CoV-2 and explores current antibody-based diagnostics, discussing their strengths, clinical utility, and limitations. Furthermore, we underscore the therapeutic potential of SARS-CoV-2-specific antibodies, discussing various antibody-based therapies such as monoclonal antibodies, polyclonal antibodies, anti-cytokines, convalescent plasma, and hyperimmunoglobulin-based therapies. Moreover, we offer insights into antibody responses to SARS-CoV-2 vaccines, emphasizing the significance of neutralizing antibodies in order to confer immunity to SARS-CoV-2, along with emerging variants of concern (VOCs) and circulating Omicron subvariants. We also highlight challenges in the field, such as the risks of antibody-dependent enhancement (ADE) for SARS-CoV-2 antibodies, and shed light on the challenges associated with the original antigenic sin (OAS) effect and long COVID. Overall, this review intends to provide valuable insights, which are crucial to advancing sensitive diagnostic tools, identifying efficient antibody-based therapeutics, and developing effective vaccines to combat the evolving threat of SARS-CoV-2 variants on a global scale.
Collapse
Affiliation(s)
- Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India
| | - Prajna Tripathi
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, USA;
| | - Prashant Kumar
- R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Ritu Shekhar
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
7
|
Corrao G, Porcu G, Tratsevich A, Cereda D, Pavesi G, Bertolaso G, Franchi M. Estimating All-Cause Deaths Averted in the First Two Years of the COVID-19 Vaccination Campaign in Italy. Vaccines (Basel) 2024; 12:413. [PMID: 38675795 PMCID: PMC11055119 DOI: 10.3390/vaccines12040413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/10/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Comparing deaths averted by vaccination campaigns is a crucial public health endeavour. Excess all-cause deaths better reflect the impact of the pandemic than COVID-19 deaths. We used a seasonal autoregressive integrated moving average with exogenous factors model to regress daily all-cause deaths on annual trend, seasonality, and environmental temperature in three Italian regions (Lombardy, Marche and Sicily) from 2015 to 2019. The model was used to forecast excess deaths during the vaccinal period (December 2020-October 2022). We used the prevented fraction to estimate excess deaths observed during the vaccinal campaigns, those which would have occurred without vaccination, and those averted by the campaigns. At the end of the vaccinal period, the Lombardy region proceeded with a more intensive COVID-19 vaccination campaign than other regions (on average, 1.82 doses per resident, versus 1.67 and 1.56 in Marche and Sicily, respectively). A higher prevented fraction of all-cause deaths was consistently found in Lombardy (65% avoided deaths, as opposed to 60% and 58% in Marche and Sicily). Nevertheless, because of a lower excess mortality rate found in Lombardy compared to Marche and Sicily (12, 24 and 23 per 10,000 person-years, respectively), a lower rate of averted deaths was observed (22 avoided deaths per 10,000 person-years, versus 36 and 32 in Marche and Sicily). In Lombardy, early and full implementation of adult COVID-19 vaccination was associated with the largest reduction in all-cause deaths compared to Marche and Sicily.
Collapse
Affiliation(s)
- Giovanni Corrao
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (A.T.); (M.F.)
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
| | - Gloria Porcu
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (A.T.); (M.F.)
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
- Specialization School of Health Statistics and Biometrics, University of Padua, 35131 Padua, Italy
| | - Alina Tratsevich
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (A.T.); (M.F.)
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
| | - Danilo Cereda
- Preventive Unit of Welfare Department, Lombardy Region, 20124 Milan, Italy;
| | - Giovanni Pavesi
- General Directorate of Welfare Department, Lombardy Region, 20124 Milan, Italy;
| | | | - Matteo Franchi
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, 20126 Milan, Italy; (G.C.); (A.T.); (M.F.)
- Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, 20126 Milan, Italy
| |
Collapse
|
8
|
Abraham R, Biju A, Ittyachen AM. Diabetes Mellitus and Other Comorbidities: Outcome among Covid-19 Patients in Kerala: A Retrospective Observational Study. J Family Med Prim Care 2024; 13:1544-1549. [PMID: 38827707 PMCID: PMC11141971 DOI: 10.4103/jfmpc.jfmpc_1529_23] [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: 09/13/2023] [Revised: 12/06/2023] [Accepted: 12/07/2023] [Indexed: 06/04/2024] Open
Abstract
Background Covid-19 was declared by the WHO as a pandemic in 2020; India was also severely affected. Diabetes, a major lifestyle disorder, has seen its prevalence rate rising in developing countries. India is home to the world's second-largest population of diabetes. Several studies have reported greater severity and mortality of Covid-19 in diabetic patients. Methodology This was a hospital-based retrospective study done in a rural-based medical college in Kerala State. Data was collected using a semi-structured proforma and analysis was performed using Statistical Package for Social Sciences software version 25. The study was part of the STS research program of the Indian Council of Medical Research (ICMR). Results There were 567 patients in the study. Those with pre-existing diabetes had a worse outcome compared to those with newly detected diabetes. The presence of CKD was associated with a poor outcome. Patients admitted to the ICU, and those on assisted ventilation also had a lower survival rate; within the subgroup, those on non-invasive ventilation had a better outcome. Conclusion Mortality in Covid-19 is multifactorial. Those with diabetes have a poor outcome. Comorbidities have been reported to confer a high mortality rate in Covid-19 but this was not so in our study (except for CKD). Variability in outcome with respect to comorbidities and better outcomes in those who were non-invasively ventilated calls for more research to establish the relationship between pre-existing conditions and severity of disease. The use of non-invasive ventilation could also provide succor to resource-limited communities.
Collapse
Affiliation(s)
- Rahael Abraham
- Department of Medicine, M.O.S.C Medical College and Hospital, Kolenchery, Ernakulam District, Kerala, India
| | - Asha Biju
- Department of Medicine, M.O.S.C Medical College and Hospital, Kolenchery, Ernakulam District, Kerala, India
| | - Abraham M. Ittyachen
- Department of Medicine, M.O.S.C Medical College and Hospital, Kolenchery, Ernakulam District, Kerala, India
| |
Collapse
|
9
|
Tong X, Wang Q, Jung W, Chicz TM, Blanc R, Parker LJ, Barouch DH, McNamara RP. Compartment-Specific Antibody Correlates of Protection to SARS-CoV-2 Omicron in Macaques. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.582951. [PMID: 38464001 PMCID: PMC10925337 DOI: 10.1101/2024.03.01.582951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Antibodies represent a primary mediator of protection against respiratory viruses such as SARS-CoV-2. Serum neutralizing antibodies (NAbs) are often considered a primary correlate of protection. However, detailed antibody profiles including characterization of antibody functions in different anatomic compartments are not well understood. Here we show that antibody correlates of protection against SARS-CoV-2 challenge are different in systemic versus mucosal compartments in rhesus macaques. In serum, neutralizing antibodies were the strongest correlate of protection and were linked to Spike-specific binding antibodies and other extra-neutralizing antibody functions that create a larger protective network. In contrast, in bronchiolar lavage (BAL), antibody-dependent cellular phagocytosis (ADCP) proved the strongest correlate of protection rather than NAbs. Within BAL, ADCP was linked to mucosal Spike-specific IgG, IgA/secretory IgA, and Fcγ-receptor binding antibodies. Our results support a model in which antibodies with different functions mediate protection at different anatomic sites. The correlation of ADCP and other Fc functional antibody responses with protection in BAL suggests that these antibody responses may be critical for protection against SARS-CoV-2 Omicron challenge in mucosa.
Collapse
Affiliation(s)
- Xin Tong
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Qixin Wang
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Wonyeong Jung
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Taras M. Chicz
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Ross Blanc
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Lily J. Parker
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| | - Dan H. Barouch
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Ryan P. McNamara
- Ragon Institute of Mass General, MIT, and Harvard, Cambridge, MA, United States
| |
Collapse
|
10
|
Park JS, Jeon J, Um J, Choi YY, Kim MK, Lee KS, Sung HK, Jang HC, Chin B, Kim CK, Oh MD, Lee CS. Magnitude and Duration of Serum Neutralizing Antibody Titers Induced by a Third mRNA COVID-19 Vaccination against Omicron BA.1 in Older Individuals. Infect Chemother 2024; 56:25-36. [PMID: 38014726 PMCID: PMC10990888 DOI: 10.3947/ic.2023.0057] [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: 05/26/2023] [Accepted: 08/21/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron variant (B.1.1.529) is dominating coronavirus disease 2019 (COVID-19) worldwide. The waning protective effect of available vaccines against the Omicron variant is a critical public health issue. This study aimed to assess the impact of the third COVID-19 vaccination on immunity against the SARS-CoV-2 Omicron BA.1 strain in older individuals. MATERIALS AND METHODS Adults aged ≥60 years who had completed two doses of the homologous COVID-19 vaccine with either BNT162b2 (Pfizer/BioNTech, New York, NY, USA, BNT) or ChAdOx1 nCoV (SK bioscience, Andong-si, Gyeongsangbuk-do, Korea, ChAd) were registered to receive the third vaccination. Participants chose either BNT or mRNA-1273 (Moderna, Norwood, MA, USA, m1273) mRNA vaccine for the third dose and were categorized into four groups: ChAd/ChAd/BNT, ChAd/ChAd/m1273, BNT/BNT/BNT, and BNT/BNT/m1273. Four serum specimens were obtained from each participant at 0, 4, 12, and 24 weeks after the third dose (V1, V2, V3, and V4, respectively). Serum-neutralizing antibody (NAb) activity against BetaCoV/Korea/KCDC03/2020 (NCCP43326, ancestral strain) and B.1.1.529 (NCCP43411, Omicron BA.1 variant) was measured using plaque reduction neutralization tests. A 50% neutralizing dilution (ND50) >10 was considered indicative of protective NAb titers. RESULTS In total, 186 participants were enrolled between November 24, 2021, and June 30, 2022. The respective groups received the third dose at a median (interquartile range [IQR]) of 132 (125 - 191), 123 (122 - 126), 186 (166 - 193), and 182 (175 - 198) days after the second dose. Overall, ND50 was lower at V1 against Omicron BA.1 than against the ancestral strain. NAb titers against the ancestral strain and Omicron BA.1 variant at V2 were increased at least 30-fold (median [IQR], 1235.35 [1021.45 - 2374.65)] and 129.8 [65.3 - 250.7], respectively). ND50 titers against the ancestral strain and Omicron variant did not differ significantly among the four groups (P = 0.57). NAb titers were significantly lower against the Omicron variant than against the ancestral strain at V3 (median [IQR], 36.4 (17.55 - 75.09) vs. 325.9 [276.07 - 686.97]; P = 0.012). NAb titers against Omicron at V4 were 16 times lower than that at V3. Most sera exhibited a protective level (ND50 >10) at V4 (75.0% [24/32], 73.0% [27/37], 73.3% [22/30], and 70.6% [12/17] in the ChAd/ChAd/BNT, ChAd/ChAd/m1273, BNT/BNT/BNT, and BNT/BNT/m1273 groups, respectively), with no significant differences among groups (P = 0.99). CONCLUSION A third COVID-19 mRNA vaccine dose restored waning NAb titers against Omicron BA.1. Our findings support a third-dose vaccination program to prevent the waning of humoral immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Jun-Sun Park
- Research Institute for Public Healthcare, National Medical Center, Seoul, Korea
| | - Jaehyun Jeon
- Department of Infectious Diseases, National Medical Center, Seoul, Korea
| | - Jihye Um
- Research Institute for Public Healthcare, National Medical Center, Seoul, Korea
| | - Youn Young Choi
- Department of Pediatrics, National Medical Center, Seoul, Korea
| | - Min-Kyung Kim
- Department of Infectious Diseases, National Medical Center, Seoul, Korea
| | - Kyung-Shin Lee
- Research Institute for Public Healthcare, National Medical Center, Seoul, Korea
| | - Ho Kyung Sung
- Research Institute for Public Healthcare, National Medical Center, Seoul, Korea
| | - Hee-Chang Jang
- National Institute of Infectious Diseases, National Institute of Health, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - BumSik Chin
- Department of Infectious Diseases, National Medical Center, Seoul, Korea
| | - Choon Kwan Kim
- Division of Infectious Diseases, VHS Medical Center, Seoul, Korea
| | - Myung-don Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Korea
| | - Chang-Seop Lee
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, Korea
| |
Collapse
|
11
|
Alleva DG, Feitsma EA, Janssen YF, Boersma HH, Lancaster TM, Sathiyaseelan T, Murikipudi S, Delpero AR, Scully MM, Ragupathy R, Kotha S, Haworth JR, Shah NJ, Rao V, Nagre S, Ronca SE, Green FM, Shaw SA, Aminetzah A, Kruijff S, Brom M, van Dam GM, Zion TC. Immunogenicity phase II study evaluating booster capacity of nonadjuvanted AKS-452 SARS-Cov-2 RBD Fc vaccine. NPJ Vaccines 2024; 9:40. [PMID: 38383578 PMCID: PMC10881471 DOI: 10.1038/s41541-024-00830-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/08/2024] [Indexed: 02/23/2024] Open
Abstract
AKS-452, a subunit vaccine comprising an Fc fusion of the ancestral wild-type (WT) SARS-CoV-2 virus spike protein receptor binding domain (SP/RBD), was evaluated without adjuvant in a single cohort, non-randomized, open-labelled phase II study (NCT05124483) at a single site in The Netherlands for safety and immunogenicity. A single 90 µg subcutaneous booster dose of AKS-452 was administered to 71 adults previously primed with a registered mRNA- or adenovirus-based vaccine and evaluated for 273 days. All AEs were mild and no SAEs were attributable to AKS-452. While all subjects showed pre-existing SP/RBD binding and ACE2-inhibitory IgG titers, 60-68% responded to AKS-452 via ≥2-fold increase from days 28 to 90 and progressively decreased back to baseline by day 180 (days 28 and 90 mean fold-increases, 14.7 ± 6.3 and 8.0 ± 2.2). Similar response kinetics against RBD mutant proteins (including omicrons) were observed but with slightly reduced titers relative to WT. There was an expected strong inverse correlation between day-0 titers and the fold-increase in titers at day 28. AKS-452 enhanced neutralization potency against live virus, consistent with IgG titers. Nucleocapsid protein (Np) titers suggested infection occurred in 66% (46 of 70) of subjects, in which only 20 reported mild symptomatic COVID-19. These favorable safety and immunogenicity profiles support booster evaluation in a planned phase III universal booster study of this room-temperature stable vaccine that can be rapidly and inexpensively manufactured to serve vaccination at a global scale without the need of a complex distribution or cold chain.
Collapse
Affiliation(s)
- David G Alleva
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Eline A Feitsma
- Department of Surgery, University Medical Center Groningen (UMCG), Hanzeplein 1, 9700 RB, Groningen, The Netherlands
| | - Yester F Janssen
- Department of Nuclear Medicine and Molecular Imaging, UMCG, Groningen, The Netherlands
| | - Hendrikus H Boersma
- Department of Nuclear Medicine and Molecular Imaging, UMCG, Groningen, The Netherlands
- Department of Clinical Pharmacy and Pharmacology, UMCG, Groningen, The Netherlands
| | - Thomas M Lancaster
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | | | - Sylaja Murikipudi
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Andrea R Delpero
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Melanie M Scully
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Ramya Ragupathy
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Sravya Kotha
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Jeffrey R Haworth
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Nishit J Shah
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Vidhya Rao
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Shashikant Nagre
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA
| | - Shannon E Ronca
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX, 77030, USA
| | - Freedom M Green
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX, 77030, USA
| | - Stephen A Shaw
- Department of Pediatrics, Division of Tropical Medicine, Baylor College of Medicine and Texas Children's Hospital, Baylor, College of Medicine, 1102 Bates Ave, 300.15, Houston, TX, 77030, USA
| | - Ari Aminetzah
- TRACER BV, Aarhusweg 2-1/2-2, 9723 JJ, Groningen, The Netherlands
| | - Schelto Kruijff
- Department of Surgery, University Medical Center Groningen (UMCG), Hanzeplein 1, 9700 RB, Groningen, The Netherlands
- Department of Nuclear Medicine and Molecular Imaging, UMCG, Groningen, The Netherlands
| | - Maarten Brom
- TRACER BV, Aarhusweg 2-1/2-2, 9723 JJ, Groningen, The Netherlands
| | - Gooitzen M van Dam
- Department of Nuclear Medicine and Molecular Imaging, UMCG, Groningen, The Netherlands
- TRACER BV, Aarhusweg 2-1/2-2, 9723 JJ, Groningen, The Netherlands
| | - Todd C Zion
- Akston Biosciences Corporation, 100 Cummings Center, Suite 454C, Beverly, MA, 01915, USA.
| |
Collapse
|
12
|
Cheng MQ, Weng ZY, Li R, Song G. Efficacy of adjuvant-associated COVID-19 vaccines against SARS-CoV-2 variants of concern in randomized controlled trials: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e35201. [PMID: 38363919 PMCID: PMC10869057 DOI: 10.1097/md.0000000000035201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 08/23/2023] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Adjuvants may enhance the efficacy of vaccines. however, the efficacy of adjuvant-associated COVID-19 vaccines (ACVs) remains unclear since the emergence of the COVID-19 pandemic. This study aimed to address this gap by conducting a systematic review and meta-analysis of the efficacy of ACVs against Severe Acute Respiratory Syndrome Coronavirus 2 CoV (SARS-CoV-2) variants of concern (VOC). METHODS A systematic search was conducted of randomized controlled trials (RCTs) evaluating the vaccine efficacy (VE) of ACVs against VOC (alpha, beta, gamma, delta, or Omicron), up to May 27, 2023. The DerSimonian-Laird random-effects model was used to assess VE with 95% confidence intervals (CI) through meta-analysis. Cochrane Risk of Bias tools were used to assess the risk of bias in RCTs. RESULTS Eight RCTs with 113,202 participants were included in the analysis, which incorporated 4 ACVs [Matrix-M (NVX-CoV2373), Alum (BBV152), CpG-1018/Alum (SCB-2019), and AS03 (CoVLP]). The pooled efficacy of full vaccination with ACVs against VOC was 88.0% (95% CI: 83.0-91.5). Full vaccination was effective against Alpha, Beta, Delta, and Gamma variants, with VE values of 93.66% (95% CI: 86.5-100.74), 64.70% (95% CI: 41.87-87.54), 75.95% (95% CI: 67.9-83.99), and 91.26% (95% CI: 84.35-98.17), respectively. Currently, there is a lack of RCT evidence regarding the efficacy of ACVs against the Omicron variant. CONCLUSION In this meta-analysis, it should be that full vaccination with ACVs has high efficacy against Alpha or Gamma variants and moderate efficacy against Beta and Delta variants. Notably, with the exception of the aluminum-adjuvanted vaccine, the other ACVs had moderate to high efficacy against the SARS-CoV-2 variant. This raises concerns about the effectiveness of ACVs booster vaccinations against Omicron.
Collapse
Affiliation(s)
- Meng-qun Cheng
- Department of Reproductive Medicine, The Pu’er People’s Hospital, Pu’er, China
| | - Zhi-Ying Weng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Rong Li
- Department of Pharmacy, The Pu’er People’s Hospital, Pu’er, China
| | - Gao Song
- Department of Pharmacy, The Pu’er People’s Hospital, Pu’er, China
| |
Collapse
|
13
|
Tamada Y, Takeuchi K, Kusama T, Maeda M, Murata F, Osaka K, Fukuda H. Bivalent mRNA vaccine effectiveness against COVID-19 among older adults in Japan: a test-negative study from the VENUS study. BMC Infect Dis 2024; 24:135. [PMID: 38287337 PMCID: PMC10823731 DOI: 10.1186/s12879-024-09035-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Bivalent COVID-19 vaccines have been implemented worldwide since the booster vaccination campaigns of autumn of 2022, but little is known about their effectiveness. Thus, this study holistically evaluated the effectiveness of bivalent vaccines against infection in older adults in Japan. METHODS We adopted the test-negative design using COVID-19 test data of individuals, aged ≥ 65 years, residing in three municipalities in Japan, who underwent tests in medical institutions between October 1 and December 30, 2022. Logistic regression analyses were conducted to estimate the odds of testing positive according to vaccination status. Vaccine effectiveness was defined as (1 - odds ratio) × 100%. RESULTS A total of 3,908 positive and 16,090 negative results were included in the analyses. Receiving a bivalent dose in addition to ≥ 2 monovalent doses was 33.6% (95% confidence interval [CI]: 20.8, 44.3%) more effective than receiving no vaccination, and 18.2% (95% CI: 9.4, 26.0%) more effective than receiving ≥ 2 monovalent doses but not receiving a bivalent vaccination. In addition, the effectiveness peaked at 14-20 days after administration and then gradually declined over time. Furthermore, a bivalent booster dose provided 18.6% (95% CI: 9.9, 26.5%) additional protection among those vaccinated with ≥ 2 monovalent doses, in the absence of a previous infection history. However, we did not find sufficient evidence of effectiveness of bivalent vaccines among previously infected older adults. CONCLUSIONS Bivalent vaccines are effective against COVID-19 infections among older adults without a history of infection.
Collapse
Affiliation(s)
- Yudai Tamada
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan
| | - Kenji Takeuchi
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan.
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Aichi, Japan.
- Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan.
| | - Taro Kusama
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
- Division of Statistics and Data Science, Liaison Center for Innovative Dentistry, Tohoku University Graduate School of Dentistry, Miyagi, Japan
| | - Megumi Maeda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumiko Murata
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Ken Osaka
- Department of International and Community Oral Health, Tohoku University Graduate School of Dentistry, 4-1, Seiryo-Machi, Aoba-Ku, Sendai, Miyagi, 980-8575, Japan
| | - Haruhisa Fukuda
- Department of Health Care Administration and Management, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
14
|
Diego JGB, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian hamsters. PLoS Pathog 2024; 20:e1011805. [PMID: 38198521 PMCID: PMC10805310 DOI: 10.1371/journal.ppat.1011805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 01/23/2024] [Accepted: 11/06/2023] [Indexed: 01/12/2024] Open
Abstract
Hybrid immunity (vaccination + natural infection) to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. The mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed for T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera, reflected by smaller antigenic cartography distances. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Salamanca, Spain
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Kim Handrejk
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lauren A. Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sara S. El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, La Jolla, California, United States of America
| | - Max W. Chang
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Brett T. Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, Wyoming, United States of America
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| |
Collapse
|
15
|
Penetra SLS, Santos HFP, Resende PC, Bastos LS, da Silva MFB, Pina-Costa A, Lopes RS, Saboia-Vahia L, de Oliveira ACA, Pereira EC, Filho FM, Wakimoto MD, Calvet GA, Fuller TL, Whitworth J, Smith C, Nielsen-Saines K, Carvalho MS, Espíndola OM, Guaraldo L, Siqueira MM, Brasil P. SARS-CoV-2 Reinfection Cases in a Household-Based Prospective Cohort in Rio de Janeiro. J Infect Dis 2023; 228:1680-1689. [PMID: 37571849 PMCID: PMC11032242 DOI: 10.1093/infdis/jiad336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/02/2023] [Accepted: 08/09/2023] [Indexed: 08/13/2023] Open
Abstract
This was a household-based prospective cohort study conducted in Rio de Janeiro, in which people with laboratory-confirmed coronavirus disease 2019 (COVID-19) and their household contacts were followed from April 2020 through June 2022. Ninety-eight reinfections were identified, with 71 (72.5%) confirmed by genomic analyses and lineage definition in both infections. During the pre-Omicron period, 1 dose of any COVID-19 vaccine was associated with a reduced risk of reinfection, but during the Omicron period not even booster vaccines had this effect. Most reinfections were asymptomatic or milder in comparison with primary infections, a justification for continuing active surveillance to detect infections in vaccinated individuals. Our findings demonstrated that vaccination may not prevent infection or reinfection with severe acute respiratory syndrome coronavirus 2 (SARS CoV-2). Therefore we highlight the need to continuously update the antigenic target of SARS CoV-2 vaccines and administer booster doses to the population regularly, a strategy well established in the development of vaccines for influenza immunization programs.
Collapse
Affiliation(s)
- Stephanie L S Penetra
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heloisa F P Santos
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola Cristina Resende
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Soares Bastos
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michele F B da Silva
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Anielle Pina-Costa
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Serrano Lopes
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leonardo Saboia-Vahia
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Any Caroline Alves de Oliveira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Elisa Cavalcante Pereira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Medeiros Filho
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mayumi D Wakimoto
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Guilherme A Calvet
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Trevon L Fuller
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
- University of California, Los Angeles, Los Angeles, California, USA
| | - Jimmy Whitworth
- International Public Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christopher Smith
- International Public Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | | | - Marilia Sá Carvalho
- Scientific Computing Program, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otávio M Espíndola
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lusiele Guaraldo
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marilda M Siqueira
- Laboratory of Respiratory Viruses and Measles National Influenza Centre, Americas Regional Reference Lab for Measles and Rubella, Reference Laboratory for COVID-19 World Health Organization, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patricia Brasil
- Evandro Chagas National Institute of Infectious Diseases, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Southworth T, Jackson N, Singh D. Airway and Systemic Immune Responses Following the Third COVID-19 Vaccination in COPD Patients. Int J Chron Obstruct Pulmon Dis 2023; 18:3027-3036. [PMID: 38143919 PMCID: PMC10749100 DOI: 10.2147/copd.s433269] [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: 07/31/2023] [Accepted: 11/30/2023] [Indexed: 12/26/2023] Open
Abstract
Introduction Booster vaccinations are required to maintain protection against COVID-19. COPD patients are at higher risk of developing severe illness following SARS-CoV-2 infection. Previous cross-sectional analysis after the second COVID-19 booster showed similar immune responses in COPD patients and controls, but pre-vaccination samples were not available. This longitudinal study evaluated systemic and airway immune responses in COPD patients using samples obtained pre- and post-third COVID-19 vaccination. Methods Twelve COPD patients were recruited, with plasma, nasal and sputum (n = 10) samples collected pre-vaccination and 4- and 14-weeks post vaccination. Samples were analyzed for anti-spike IgA and IgG and cellular immunity. The ability of plasma and nasal samples to block ACE2-spike protein interaction was assessed for Wild type, Delta, and Omicron spike variants. Results Vaccinations increased anti-spike IgG in plasma (p < 0.001), nasal (IgG p < 0.001) and sputum (p = 0.002) samples, IgA in plasma (p < 0.001) and blood cellular immunity (p = 0.001). Plasma and nasal anti-spike IgA levels correlated (rho: 0.6, p = 0.02), with similar results for IgG (rho: 0.79, p = 0.003). Post-vaccination nasal (p = 0.002) and plasma (p < 0.001) samples were less effective at blocking Omicron spike binding to ACE2 compared to the Wild type spike variant. Discussion Airway and systemic immune responses against SARS-CoV-2 increased in COPD patients following a third COVID-19 vaccination. Nasal and systemic responses in COPD patients were less effective against Omicron variant compared to previous variants.
Collapse
Affiliation(s)
- Thomas Southworth
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
- Medicines Evaluation Unit, Manchester, UK
| | | | - Dave Singh
- Division of Infection, Immunity and Respiratory Medicine, University of Manchester, Manchester, UK
- Medicines Evaluation Unit, Manchester, UK
- Manchester University NHS Foundation Trust, Manchester, UK
| |
Collapse
|
17
|
Rosolen V, Turoldo F, Zamaro G, Del Bianco F, Pezzotti P, Castriotta L, Barbone F. COVID-19 vaccination effectiveness in the population of Friuli Venezia Giulia, North-East Italy. Control of bias associated with divergent compliance to policies in a test-negative case-control study. BMC Public Health 2023; 23:2476. [PMID: 38082276 PMCID: PMC10714502 DOI: 10.1186/s12889-023-17244-9] [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: 08/24/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Vaccine effectiveness (VE) studies consolidate knowledge of real-world effectiveness in different contexts. However, methodological issues may undermine their conclusions: to assess the VE against COVID-19 within the Italian population, a specific threat to validity is related to the consequences of divergent compliance to the Green Pass policy. METHODS To address this challenge we conducted a test negative case-control (TNCC) study and multiple sensitivity analysis among residents aged ≥ 12 in Friuli Venezia Giulia Region (FVG), North-east Italy, from February 1, 2021 to March 31, 2022. Information regarding 211,437 cases of COVID-19 infection and 845,748 matched controls was obtained from the regional computerized health database. The investigation considered: COVID-19 infection, hospitalization, and death. Multiple conditional logistic regressions adjusted for covariates were performed and VE was estimated as (1-OR COVID-19vaccinated vs. unvaccinated)x100. Mediation analyses were carried out to offset potential collider variables, particularly, the number of swabs performed after the introduction of pandemic restrictions. RESULTS Full-cycle VE against infection decreased from 96% (95% CI: 96, 97) in the Alpha period to 43% (95% CI: 42, 45) in the Omicron period. Booster dose raised the protection in Omicron period to 67% (95% CI: 66, 67). Against the evasive Omicron variant, the protection of the booster dose was 87% (95% CI: 83, 90) for hospitalization and 90% (95% CI: 82, 95) for death. The number of swabs performed was included as a covariate in the adjustments, and the mediation analysis confirmed that it was a strong mediator between vaccination and COVID-19-related outcomes. CONCLUSIONS The study suggests that, under similar TNCC settings, mediation analysis and adjustment for number of diagnostic tests should be included, as an effective approach to the challenge of differential testing behavior that may determine substantial selection bias. This correction allowed us to align with results from other studies that show how full-cycle VE against infection was initially high but decreased over time by variant circulation, counterbalanced by booster dose that raised protection across variants and outcome severity.
Collapse
Affiliation(s)
- Valentina Rosolen
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Via Cassa Di Risparmio 10, Trieste, 34121, Italy
| | - Federico Turoldo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, Trieste, 34149, Italy
| | - Gianna Zamaro
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Via Cassa Di Risparmio 10, Trieste, 34121, Italy
| | - Flavio Del Bianco
- Prevention Technical Platform, "AS FO" Western Friuli Health Authority, Via della Vecchia Ceramica 1, Pordenone, 33170, Italy
| | - Patrizio Pezzotti
- Department of Infectious Diseases, National Institute of Health (ISS), Viale Regina Elena 299, Rome, 00161, Italy
| | - Luigi Castriotta
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Via Cassa Di Risparmio 10, Trieste, 34121, Italy
- Institute of Hygiene and Evaluative Epidemiology, Friuli Centrale University Health Authority, Via Colugna 50, Udine, 33100, Italy
| | - Fabio Barbone
- Central Directorate for Health, Social Policies and Disability, Friuli Venezia Giulia Region, Via Cassa Di Risparmio 10, Trieste, 34121, Italy.
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, Trieste, 34149, Italy.
| |
Collapse
|
18
|
Sritipsukho P, Khawcharoenporn T, Siribumrungwong B, Damronglerd P, Suwantarat N, Satdhabudha A, Chaiyakulsil C, Sinlapamongkolkul P, Tangsathapornpong A, Bunjoungmanee P, Nanthapisal S, Tanprasertkul C, Sritipsukho N, Mingmalairak C, Apisarnthanarak A, Tantiyavarong P. Real-life effectiveness of COVID-19 vaccine during the Omicron variant-dominant pandemic: how many booster doses do we need? Emerg Microbes Infect 2023; 12:2174779. [PMID: 36715323 PMCID: PMC9936995 DOI: 10.1080/22221751.2023.2174779] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The surge in coronavirus disease 2019 (COVID-19) caused by the Omicron variants of the severe acute respiratory syndrome coronavirus 2 necessitates researches to inform vaccine effectiveness (VE) and other preventive measures to halt the pandemic. A test-negative case-control study was conducted among adults (age ≥18 years) who were at-risk for COVID-19 and presented for nasopharyngeal real-time polymerase chain reaction testing during the Omicron variant-dominant period in Thailand (1 January 2022-15 June 2022). All participants were prospectively followed up for COVID-19 development for 14 days after the enrolment. Vaccine effectiveness was estimated and adjusted for characteristics associated with COVID-19. Of the 7971 included individuals, there were 3104 cases and 4867 controls. The adjusted VE among persons receiving 2-dose, 3-dose, and 4-dose vaccine regimens for preventing infection and preventing moderate-to-critical diseases were 33%, 48%, 62% and 60%, 74%, 76%, respectively. The VE were generally higher among those receiving the last dose of vaccine within 90 days compared to those receiving the last dose more than 90 days prior to the enrolment. The highest VE were observed in individuals receiving the 4-dose regimen, CoronaVac-CoronaVac-ChAdOx1 nCoV-19-BNT162b2 for both preventing infection (65%) and preventing moderate-to-critical diseases (82%). Our study demonstrated increased VE along with an increase in number of vaccine doses received. Current vaccination programmes should focus on reducing COVID-19 severity and mandate at least one booster dose. The heterologous boosters with viral vector and mRNA vaccines were highly effective and can be used in individuals who previously received the primary series of inactivated vaccine.
Collapse
Affiliation(s)
- Paskorn Sritipsukho
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Thana Khawcharoenporn
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand, Thana Khawcharoenporn Division of Infectious Diseases, Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathumthani, 12120, Thailand
| | - Boonying Siribumrungwong
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Surgery, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Pansachee Damronglerd
- Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Nuntra Suwantarat
- Department of Internal Medicine, Chulabhorn International College of Medicine, Thammasat University, Pathumthani, Thailand
| | - Araya Satdhabudha
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Chanapai Chaiyakulsil
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | | | | | - Pornumpa Bunjoungmanee
- Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Sira Nanthapisal
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Pediatrics, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Chamnan Tanprasertkul
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Obstetrics & Gynecology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Naiyana Sritipsukho
- Thammasat Postdoctoral Fellowship, Thammasat University, Pathumthani, Thailand
| | - Chatchai Mingmalairak
- Department of Surgery, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Anucha Apisarnthanarak
- Center of Excellence in Applied Epidemiology, Thammasat University, Pathumthani, Thailand,Department of Internal Medicine, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| | - Pichaya Tantiyavarong
- Department of Clinical Epidemiology, Faculty of Medicine, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
19
|
Tan-Lhernould L, Tamandjou C, Deschamps G, Platon J, Sommen C, Chereau F, Parent du Châtelet I, Cauchemez S, Vaux S, Paireau J. Impact of vaccination against severe COVID-19 in the French population aged 50 years and above: a retrospective population-based study. BMC Med 2023; 21:426. [PMID: 37940955 PMCID: PMC10633992 DOI: 10.1186/s12916-023-03119-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 10/19/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Given the widespread implementation of COVID-19 vaccination to mitigate the pandemic from the end of 2020, it is important to retrospectively evaluate its impact, in particular by quantifying the number of severe outcomes prevented through vaccination. METHODS We estimated the number of hospitalizations, intensive care unit (ICU) admissions and deaths directly averted by vaccination in France, in people aged ≥ 50 years, from December 2020 to March 2022, based on (1) the number of observed events, (2) vaccination coverage, and (3) vaccine effectiveness. We accounted for the effect of primary vaccination and the first booster dose, the circulating variants, the age groups, and the waning of vaccine-induced protection over time. RESULTS An estimated 480,150 (95% CI: 260,072-582,516) hospitalizations, 132,156 (50,409-157,767) ICU admissions and 125,376 (53,792-152,037) deaths were directly averted by vaccination in people aged ≥ 50 years, which corresponds to a reduction of 63.2% (48.2-67.6), 68.7% (45.6-72.4) and 62.7% (41.9-67.1) respectively, compared to what would have been expected without vaccination over the study period. An estimated 5852 (2285-6853) deaths were directly averted among the 50-59 years old, 16,837 (6568-19,473) among the 60-69 years old, 32,136 (13,651-36,758) among the 70-79 years old and 70,551 (31,288-88,953) among the ≥ 80 years old. CONCLUSIONS The vaccination campaign in France considerably reduced COVID-19 morbidity and mortality, as well as stress on the healthcare system.
Collapse
Affiliation(s)
- Laetitia Tan-Lhernould
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Cynthia Tamandjou
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Guilhem Deschamps
- Direction Appui, Traitements et Analyses de données, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Johnny Platon
- Direction Appui, Traitements et Analyses de données, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Cécile Sommen
- Direction Appui, Traitements et Analyses de données, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Fanny Chereau
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Isabelle Parent du Châtelet
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Simon Cauchemez
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Paris, F-75015, France
| | - Sophie Vaux
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France
| | - Juliette Paireau
- Direction des Maladies Infectieuses, Santé publique France, 12 Rue du Val d'Osne, Saint-Maurice, 94415, France.
- Mathematical Modelling of Infectious Diseases Unit, Institut Pasteur, Université Paris Cité, CNRS UMR 2000, Paris, F-75015, France.
| |
Collapse
|
20
|
Roa CC, de Los Reyes MRA, Plennevaux E, Smolenov I, Hu B, Gao F, Ilagan H, Ambrosino D, Siber G, Clemens R. Superior Boosting of Neutralizing Titers Against Omicron SARS-CoV-2 Variants by Heterologous SCB-2019 Vaccine vs a Homologous Booster in CoronaVac-Primed Adults. J Infect Dis 2023; 228:1253-1262. [PMID: 37439701 PMCID: PMC10629704 DOI: 10.1093/infdis/jiad262] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/30/2023] [Accepted: 07/07/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND We compared homologous and heterologous boosting in adults in the Philippines primed with 2 or 3 doses of CoronaVac, with recombinant protein vaccine, SCB-2019. METHODS CoronaVac-immunized adults (18-72 years) received a homologous or heterologous full or half dose SCB-2019 booster. We assessed all neutralizing antibody (NAb) responses against prototype SARS-CoV-2 after 15 days and NAb against SARS-CoV-2 Delta and Omicron variants in subsets (30‒50 per arm). Participants recorded adverse events. RESULTS In 2-dose CoronaVac-primed adults prototype NAb geometric mean titers (GMT) were 203 IU/mL (95% confidence interval [CI], 182-227) and 939 IU/mL (95% CI, 841-1049) after CoronaVac and SCB-2019 boosters; the GMT ratio (4.63; 95% CI, 3.95-5.41) met predefined noninferiority and post-hoc superiority criteria. After 3-dose CoronaVac-priming prototype NAb GMTs were 279 IU/mL (95% CI, 240-325), 1044 IU/mL (95% CI, 898-1213), and 668 IU/mL (95% CI, 520-829) following CoronaVac, full and half-dose SCB-2019 boosters, respectively. NAb GMT ratios against Delta and Omicron comparing SCB-2019 with CoronaVac were all greater than 2. Mild to moderate reactogenicity was evenly balanced between groups. No vaccine-related serious adverse events were reported. CONCLUSIONS Full or half dose SCB-2019 boosters were well tolerated with superior immunogenicity than homologous CoronaVac, particularly against newly emerged variants. Clinical Trials Registration. NCT05188677.
Collapse
Affiliation(s)
- Camilo C Roa
- Department of Physiology, Philippine General Hospital, University of the Philippines, Manila, Philippines
| | | | | | - Igor Smolenov
- Clover Biopharmaceuticals, Boston, Massachusetts, USA
| | - Branda Hu
- Clover Biopharmaceuticals, Boston, Massachusetts, USA
| | - Faith Gao
- Clover Biopharmaceuticals, Boston, Massachusetts, USA
| | | | | | | | - Ralf Clemens
- International Vaccine Institute, Seoul, Republic of Korea
| |
Collapse
|
21
|
Liu H, Chen J, Shao W, Yan S, Ding S. Efficacy and Safety of Novel Oral Antivirals in Hospitalized COVID-19 Patients: A Network Meta-Analysis of Randomized Clinical Trials. Clin Epidemiol 2023; 15:1041-1053. [PMID: 37933389 PMCID: PMC10625770 DOI: 10.2147/clep.s422386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/26/2023] [Indexed: 11/08/2023] Open
Abstract
Objective Numerous pharmacological interventions are now under investigation for the treatment of the 2019 coronavirus pandemic (COVID-19), and the evidence is rapidly evolving. Our aim is to evaluate the comparative efficacy and safety of these drugs. Methods We searched for randomized clinical trials (RCTs) on the efficacy and safety of novel oral antivirals for the treatment of hospitalized COVID-19 patients until November 30, 2022, including baricitinib, ivermectin (IVM), favipiravir (FVP), chloroquine (CQ), lopinavir and ritonavir (LPV/RTV), hydroxychloroquine (HCQ), and hydroxychloroquine plus azithromycin (HCQ+AZT). The main outcomes of this network meta-analysis (NMA) were in-hospital mortality, adverse event (AE), recovery time, and improvement in peripheral capillary oxygen saturation (SpO2). For dichotomous results, the odds ratio (OR) was used, and the 95% confidence interval (CI) was determined. We also used meta-regression to explore whether different treatments affected efficacy and safety. STATA 15.0 was used to conduct the NMA. The research protocol was registered with PROSPERO (#CRD 42023415743). Results Thirty-six RCTs, with 33,555 hospitalized COVID-19 patients, were included in this analysis. First, we compared the efficacy of different novel oral antivirals. Baricitinib (OR 0.56, 95% CI: 0.35 to 0.90) showed the highest probability of being the optimal probiotic species in reducing in-hospital mortality and suggested that none of the interventions reduced AE better than placebo. In terms of safety outcomes, IVM ranked first in improving the recovery time of hospitalized COVID-19 patients (mean difference (MD) -1.36, 95% CI: -2.32 to -0.39). In addition, patients were most likely to increase SpO2 (OR 1.77, 95% CI: 0.09 to 3.45). The meta-regression revealed no significant differences between participants using different novel oral antivirals in all outcomes in hospitalized COVID-19 patients. Conclusion Currently, baricitinib has reduced in-hospital mortality in hospitalized COVID-19 patients, with moderate certainty of evidence. IVM appeared to be a safer option than placebo in improving recovery time, while FVP was associated with increased SpO2 safety outcomes. These preliminary evidence-based observations should guide clinical practice until more data are made public.
Collapse
Affiliation(s)
- Haoshuang Liu
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Jingfeng Chen
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Weihao Shao
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, People's Republic of China
| | - Su Yan
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| | - Suying Ding
- Health Management Center, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, People’s Republic of China
| |
Collapse
|
22
|
Nii-Trebi NI, Mughogho TS, Abdulai A, Tetteh F, Ofosu PM, Osei MM, Yalley AK. Dynamics of viral disease outbreaks: A hundred years (1918/19-2019/20) in retrospect - Loses, lessons and emerging issues. Rev Med Virol 2023; 33:e2475. [PMID: 37602770 DOI: 10.1002/rmv.2475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/24/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
Infectious diseases continue to be the leading cause of morbidity and mortality, and a formidable obstacle to the development and well-being of people worldwide. Viruses account for more than half of infectious disease outbreaks that have plagued the world. The past century (1918/19-2019/20) has witnessed some of the worst viral disease outbreaks the world has recorded, with overwhelming impact especially in low- and middle-income countries (LMIC). The frequency of viral disease outbreak appears to be increasing. Generally, although infectious diseases have afflicted the world for centuries and humankind has had opportunities to examine the nature of their emergence and mode of spread, almost every new outbreak poses a formidable challenge to humankind, beating the existing pandemic preparedness systems, if any, and causing significant losses. These underscore inadequacy in our understanding of the dynamics and preparedness against viral disease outbreaks that lead to epidemics and pandemics. Despite these challenges, the past 100 years of increasing frequencies of viral disease outbreaks have engendered significant improvements in response to epidemics and pandemics, and offered lessons to inform preparedness. Hence, the increasing frequency of emergence of viral outbreaks and the challenges these outbreaks pose to humankind, call for the continued search for effective ways to tackle viral disease outbreaks in real time. Through a PRISMA-based approach, this systematic review examines the outbreak of viral diseases in retrospect to decipher the outbreak patterns, losses inflicted on humanity and highlights lessons these offer for meaningful preparation against future viral disease outbreaks and pandemics.
Collapse
Affiliation(s)
- Nicholas I Nii-Trebi
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | | | - Anisa Abdulai
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Francis Tetteh
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Priscilla M Ofosu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | - Mary-Magdalene Osei
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Akua K Yalley
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
23
|
Liu X, Wang Q, Ren L, Fang X, He Z, Ding J, Wang K, Xu H, Zhang H, Song Y, Lu Q, Sun M, Han X, Cao L, Lin W, Li X, Zhang Q, Ding Y, Wang F, Wang T, Wang J, Liu X, Wu Y, Chen Y, Feng Z, Wang S, Wang X, Guan Y, Xie X, Huang H, Zhang M, Wang X, Hong Z, Jiang W, Han Y, Deng Y, Zhao J, Liao J, Wang Y, Lian Y. COVID-19 vaccination for patients with epilepsy: A Chinese expert consensus. Epilepsy Behav 2023; 147:109387. [PMID: 37625346 DOI: 10.1016/j.yebeh.2023.109387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/29/2023] [Accepted: 07/30/2023] [Indexed: 08/27/2023]
Abstract
Coronavirus disease-2019 (COVID-19) first emerged in late 2019 and has since spread worldwide. More than 600 million people have been diagnosed with COVID-19, and over 6 million have died. Vaccination against COVID-19 is one of the best ways to protect humans. Epilepsy is a common disease, and there are approximately 10 million patients with epilepsy (PWE) in China. However, China has listed "uncontrolled epilepsy" as a contraindication for COVID-19 vaccination, which makes many PWE reluctant to get COVID-19 vaccination, greatly affecting the health of these patients in the COVID-19 epidemic. However, recent clinical practice has shown that although a small percentage of PWE may experience an increased frequency of seizures after COVID-19 vaccination, the benefits of COVID-19 vaccination for PWE far outweigh the risks, suggesting that COVID-19 vaccination is safe and recommended for PWE. Nonetheless, vaccination strategies vary for different PWE, and this consensus provides specific recommendations for PWE to be vaccinated against COVID-19.
Collapse
Affiliation(s)
- Xuewu Liu
- Department of Neurology, Shandong Provincial Hospital, Affiliated to Shandong First Medical University, Jinan, China.
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing, China.
| | - Liankun Ren
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China; Clinical Research Center for Epilepsy, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Disorders, Beijing, China
| | - Xiqin Fang
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, China; Institute of Epilepsy, Shandong University, Jinan, China
| | - Zhiyi He
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jing Ding
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kang Wang
- Department of Neurology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Huiqin Xu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hua Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an JiaoTong University, Xi'an, China
| | - Yijun Song
- Hematology Hospital, Chinese Academy of Medical Sciences (Institute of Hematology, Chinese Academy of Medical Sciences), Tianjing, China
| | - Qiang Lu
- Department of Neurosurgery, Peking Union Medical College Hospital, Beijing, China
| | - Meizhen Sun
- Departmen of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiong Han
- Department of Neurology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Lili Cao
- Department of Neurology, Qilu Hospital, Shandong University, Jinan, China
| | - Weihong Lin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xiaoyi Li
- Department of Neurology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qing Zhang
- Department of Neurology, General Hospital of Ningxia Medical University, Yinchuang, China
| | - Yao Ding
- Department of Neurology, Epilepsy Center, Zhejiang University School of Medicine Second Affiliated Hospital, Hangzhou, China
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Tiancheng Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiwen Wang
- Department of Neurology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaorong Liu
- Department of Neurology, Second Affiliated Hospital of Guangzhou Medical University, China
| | - Yuan Wu
- Department of Neurology, The First Affiliated Hospital, Guangxi Medical University, Guangzhou, China
| | - Yangmei Chen
- Department of Neurology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhanhui Feng
- Department of Neurology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Shoulei Wang
- Department of Pediatrics, Qinghai Province Women and Children's Hospital, Xining, China
| | - Xiangqing Wang
- Department of Neurology, The First Medical Center of PLA General Hospital, Beijnig, China
| | - Yuguang Guan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijnig, China
| | - Xufang Xie
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huapin Huang
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ming Zhang
- Department of Neurology, Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoshan Wang
- Department of Neurology, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| | - Zhen Hong
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| | - Wen Jiang
- Department of Neurology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanbing Han
- Department of Neurology, First Affiliated Hospital Kunming Medical University, Kunming, China
| | - Yulei Deng
- Department of Neurology & Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiangming Zhao
- Department of Neurology, The First Affiliated Hospital of USTC (Anhui Provincial Hospital), Hefei, China
| | - Jianxiang Liao
- Department of Pediatrics, Shenzhen Children's Hospital, Shenzhen, China
| | - Yu Wang
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yajun Lian
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
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
|
25
|
Tong X, McNamara RP, Avendaño MJ, Serrano EF, García-Salum T, Pardo-Roa C, Bertera HL, Chicz TM, Levican J, Poblete E, Salinas E, Muñoz A, Riquelme A, Alter G, Medina RA. Waning and boosting of antibody Fc-effector functions upon SARS-CoV-2 vaccination. Nat Commun 2023; 14:4174. [PMID: 37443074 PMCID: PMC10345146 DOI: 10.1038/s41467-023-39189-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 06/01/2023] [Indexed: 07/15/2023] Open
Abstract
Since the emergence of SARS-CoV-2, vaccines targeting COVID-19 have been developed with unprecedented speed and efficiency. CoronaVac, utilising an inactivated form of the COVID-19 virus and the mRNA26 based Pfizer/BNT162b2 vaccines are widely distributed. Beyond the ability of vaccines to induce production of neutralizing antibodies, they might lead to the generation of antibodies attenuating the disease by recruiting cytotoxic and opsonophagocytic functions. However, the Fc-effector functions of vaccine induced antibodies are much less studied than virus neutralization. Here, using systems serology, we follow the longitudinal Fc-effector profiles induced by CoronaVac and BNT162b2 up until five months following the two-dose vaccine regimen. Compared to BNT162b2, CoronaVac responses wane more slowly, albeit the levels remain lower than that of BNT162b2 recipients throughout the entire observation period. However, mRNA vaccine boosting of CoronaVac responses, including response to the Omicron variant, induce significantly higher peak of antibody functional responses with increased humoral breadth. In summary, we show that vaccine platform-induced humoral responses are not limited to virus neutralization but rather utilise antibody dependent effector functions. We demonstrate that this functionality wanes with different kinetics and can be rescued and expanded via boosting with subsequent homologous and heterologous vaccination.
Collapse
Affiliation(s)
- X Tong
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - R P McNamara
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - M J Avendaño
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E F Serrano
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - T García-Salum
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - C Pardo-Roa
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - H L Bertera
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - T M Chicz
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA
| | - J Levican
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E Poblete
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - E Salinas
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - A Muñoz
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
| | - A Riquelme
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Catolica de Chile, Santiago, 8331150, Chile
| | - G Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, 02139, USA.
| | - R A Medina
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile.
- Advanced Interdisciplinary Rehabilitation Register (AIRR) - COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, 8331150, Chile.
- Department of Pathology and Laboratory Medicine, School of Medicine, Emory University, Atlanta, GA, 30322, USA.
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
26
|
She Z, Yao Y, Wang C, Li Y, Xiong X, Liu Y. M pro-targeted anti-SARS-CoV-2 inhibitor-based drugs. JOURNAL OF CHEMICAL RESEARCH 2023; 47:17475198231184799. [PMID: 37455837 PMCID: PMC10333551 DOI: 10.1177/17475198231184799] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
The COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 is a global health emergency. The main protease is an important drug target in coronaviruses. It plays an important role in the processing of viral RNA-translated polyproteins and is highly conserved in the amino acid sequence and three-dimensional structure, making it a good drug target for which several small molecule inhibitors are available. This paper describes the various anti-severe acute respiratory syndrome coronavirus 2 inhibitor drugs targeting Mpro discovered since the severe acute respiratory syndrome coronavirus 2 outbreak at the end of 2019, with all these compounds inhibiting severe acute respiratory syndrome coronavirus 2 Mpro activity in vitro. This provides a reference for the development of severe acute respiratory syndrome coronavirus 2 Mpro-targeted inhibitors and the design of therapeutic approaches to address newly emerged severe acute respiratory syndrome coronavirus 2 mutant strains with immune evasion capabilities.
Collapse
Affiliation(s)
- Zhuxin She
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, P.R. China
| | - Yinuo Yao
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, P.R. China
| | - Conglong Wang
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, P.R. China
| | - Yi Li
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, P.R. China
| | - Xiaohui Xiong
- College of Food Science and Light Industry, Nanjing Tech University, Nanjing, P.R. China
| | - Yuanyuan Liu
- School of Pharmaceutical and Chemical Engineering, ChengXian College, Southeast University, Nanjing, P.R. China
| |
Collapse
|
27
|
Goshina A, Matyushenko V, Mezhenskaya D, Rak A, Katelnikova A, Gusev D, Rudenko L, Isakova-Sivak I. RDE Treatment Prevents Non-Specific Detection of SARS-CoV-2- and Influenza-Specific IgG Antibodies in Heat-Inactivated Serum Samples. Antibodies (Basel) 2023; 12:39. [PMID: 37366655 PMCID: PMC10295076 DOI: 10.3390/antib12020039] [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: 05/01/2023] [Revised: 05/28/2023] [Accepted: 06/12/2023] [Indexed: 06/28/2023] Open
Abstract
Assessing the levels of serum IgG antibodies is widely used to measure immunity to influenza and the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) after natural infection or vaccination with specific vaccines, as well as to study immune responses to these viruses in animal models. For safety reasons, sometimes serum specimens collected from infected individuals are subjected to heat inactivation at 56 °C to reduce the risk of infecting personnel during serological studies. However, this procedure may affect the level of virus-specific antibodies, making the results of antibody immunoassays uninterpretable. Here, we evaluated the effect of the heat inactivation of human, ferret and hamster serum samples on the binding of IgG antibodies to the influenza and SARS-CoV-2 antigens. For this, serum samples of naive and immune hosts were analyzed in three variants: (i) untreated sera, (ii) heated at 56 °C for 1 h, and (iii) treated with receptor-destroying enzyme (RDE). The samples were studied through an in-house enzyme-linked immunosorbent assay (ELISA) using whole influenza virus or recombinant proteins corresponding to nucleocapsid (N) protein and the receptor-binding domain of SARS-CoV-2 Spike (RBD) as antigens. We demonstrated that the heat inactivation of the naive serum samples of various hosts can lead to false-positive results, while RDE treatment abolished the effect of the non-specific binding of IgG antibodies to the viral antigens. Furthermore, RDE also significantly decreased the level of virus-specific IgG antibodies in SARS-CoV-2 and influenza-immune sera of humans and animals, although it is unknown whether it actually removes true virus-specific IgG antibodies or only non-specifically binding artifacts. Nevertheless, we suggest that the RDE treatment of human and animal sera may be useful in preventing false-positive results in various immunoassays, while also neutralizing infectious virus, since the standard protocol for the use of RDE also includes heating the sample at 56 °C.
Collapse
Affiliation(s)
- Arina Goshina
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Alexandra Rak
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Anastasia Katelnikova
- Department of Toxicology and Microbiology, Institute of Preclinical Research Ltd., 188663 Saint Petersburg, Russia;
| | - Denis Gusev
- Botkin Infectious Diseases Hospital, Piskarovskiy Ave 49, 195067 Saint Petersburg, Russia
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, 197376 Saint Petersburg, Russia; (A.G.); (V.M.); (D.M.); (A.R.)
| |
Collapse
|
28
|
Cheng Q, Hao X, Wu D, Wang Q, Spear RC, Wei S. Feasible intervention combinations for achieving a safe exit of the Zero-COVID policy in China and its determinants: an individual-based model study. BMC Infect Dis 2023; 23:390. [PMID: 37308872 PMCID: PMC10258473 DOI: 10.1186/s12879-023-08382-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 06/07/2023] [Indexed: 06/14/2023] Open
Abstract
BACKGROUND Although several pathways have been proposed as the prerequisite for a safe phase-out in China, it is not clear which of them are the most important for keeping the mortality rate low, what thresholds should be achieved for these most important interventions, and how the thresholds change with the assumed key epidemiological parameters and population characteristics. METHODS We developed an individual-based model (IBM) to simulate the transmission of the Omicron variant in the synthetic population, accounting for the age-dependent probabilities of severe clinical outcomes, waning vaccine-induced immunity, increased mortality rates when hospitals are overburdened, and reduced transmission when self-isolated at home after testing positive. We applied machine learning algorithms on the simulation outputs to examine the importance of each intervention parameter and the feasible intervention parameter combinations for safe exits, which is defined as having mortality rates lower than that of influenza in China (14.3 per 100, 000 persons). RESULTS We identified vaccine coverage in those above 70 years old, number of ICU beds per capita, and the availability of antiviral treatment as the most important interventions for safe exits across all studied locations, although the thresholds required for safe exits vary remarkably with the assumed vaccine effectiveness, as well as the age structure, age-specific vaccine coverage, community healthcare capacity of the studied locations. CONCLUSIONS The analytical framework developed here can provide the basis for further policy decisions that incorporate considerations about economic costs and societal impacts. Achieving safe exits from the Zero-COVID policy is possible, but challenging for China's cities. When planning for safe exits, local realities such as the age structure and current age-specific vaccine coverage must be taken into consideration.
Collapse
Affiliation(s)
- Qu Cheng
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Degang Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Departments of Epidemiology and Biostatistics, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Qi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Robert C Spear
- Division of Environmental Health, of Public Health, University of California, Berkeley, CA, USA
| | - Sheng Wei
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
29
|
Xu J, Lan X, Zhang L, Zhang X, Zhang J, Song M, Liu J. The effectiveness of the first dose COVID-19 booster vs. full vaccination to prevent SARS-CoV-2 infection and severe COVID-19 clinical event: a meta-analysis and systematic review of longitudinal studies. Front Public Health 2023; 11:1165611. [PMID: 37325336 PMCID: PMC10267329 DOI: 10.3389/fpubh.2023.1165611] [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: 02/14/2023] [Accepted: 05/09/2023] [Indexed: 06/17/2023] Open
Abstract
Background The effectiveness of full Coronavirus Disease 2019 (COVID-19) vaccination against COVID-19 wanes over time. This study aimed to synthesize the clinical effectiveness of the first dose of COVID-19 booster by comparing it to the full vaccination. Methods Studies in PubMed, Web of Science, Embase, and clinical trials databases were searched from 1 January 2021 to 10 September 2022. Studies were eligible if they comprised general adult participants who were not ever or currently infected with SARS-CoV-2, did not have impaired immunity or immunosuppression, and did not have severe diseases. The seroconversion rate of antibodies to S and S subunits and antibody titers of SARS-CoV-2, frequency, phenotype of specific T and B cells, and clinical events involving confirmed infection, admission to the intensive care unit (ICU), and death were compared between the first booster dose of COVID-19 vaccination group and full vaccination group. The DerSimonian and Laird random effects models were used to estimate the pooled risk ratios (RRs) and corresponding 95% confidence intervals (CIs) for the outcomes of clinical interest. While a qualitative description was mainly used to compare the immunogenicity between the first booster dose of COVID-19 vaccination group and full vaccination group. Sensitivity analysis was used to deal with heterogenicity. Results Of the 10,173 records identified, 10 studies were included for analysis. The first dose COVID-19 booster vaccine could induce higher seroconversion rates of antibodies against various SAS-CoV-2 fragments, higher neutralization antibody titers against various SARS-CoV-2 variants, and robust cellular immune response compared to the full vaccination. The risk of SARS-CoV-2 infection, the risk of admission to the ICU, and the risk of death were all higher in the non-booster group than those in the booster group, with RRs of 9.45 (95% CI 3.22-27.79; total evaluated population 12,422,454 vs. 8,441,368; I2 = 100%), 14.75 (95% CI 4.07-53.46; total evaluated population 12,048,224 vs. 7,291,644; I2 = 91%), and 13.63 (95% CI 4.72-39.36; total evaluated population 12,385,960 vs. 8,297,037; I2 = 85%), respectively. Conclusion A homogenous or heterogeneous booster COVID-19 vaccination could elicit strong humoral and cellular immune responses to SARS-CoV-2. Furthermore, it could significantly reduce the risk of SARS-CoV-2 infection and severe COVID-19 clinical events on top of two doses. Future studies are needed to investigate the long-term clinical effectiveness of the first booster dose of the COVID-19 vaccine and compare the effectiveness between homogenous and heterogeneous booster COVID-19 vaccination. Systematic review registration https://inplasy.com/inplasy-2022-11-0114/, identifier: INPLASY2022110114.
Collapse
Affiliation(s)
- Junjie Xu
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Xinquan Lan
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Liangyuan Zhang
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Xiangjun Zhang
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Jiaqi Zhang
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Moxin Song
- Clinical Research Academy, Peking University Shenzhen Hospital, Peking University, Shenzhen, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Jiaye Liu
- School of Public Health, Shenzhen University Medical School, Shenzhen, China
| |
Collapse
|
30
|
Pandit R, Matthews QL. A SARS-CoV-2: Companion Animal Transmission and Variants Classification. Pathogens 2023; 12:775. [PMID: 37375465 DOI: 10.3390/pathogens12060775] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The continuous emergence of novel viruses and their diseases are a threat to global public health as there have been three outbreaks of coronaviruses that are highly pathogenic to humans in the span of the last two decades, severe acute respiratory syndrome (SARS)-CoV in 2002, Middle East respiratory syndrome (MERS)-CoV in 2012, and novel SARS-CoV-2 which emerged in 2019. The unprecedented spread of SARS-CoV-2 worldwide has given rise to multiple SARS-CoV-2 variants that have either altered transmissibility, infectivity, or immune escaping ability, causing diseases in a broad range of animals including human and non-human hosts such as companion, farm, zoo, or wild animals. In this review, we have discussed the recent SARS-CoV-2 outbreak, potential animal reservoirs, and natural infections in companion and farm animals, with a particular focus on SARS-CoV-2 variants. The expeditious development of COVID-19 vaccines and the advancements in antiviral therapeutics have contained the COVID-19 pandemic to some extent; however, extensive research and surveillance concerning viral epidemiology, animal transmission, variants, or seroprevalence in diverse hosts are essential for the future eradication of COVID-19.
Collapse
Affiliation(s)
- Rachana Pandit
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| | - Qiana L Matthews
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
31
|
García-Bernalt Diego J, Singh G, Jangra S, Handrejk K, Laporte M, Chang LA, El Zahed SS, Pache L, Chang MW, Warang P, Aslam S, Mena I, Webb BT, Benner C, García-Sastre A, Schotsaert M. Breakthrough infections by SARS-CoV-2 variants boost cross-reactive hybrid immune responses in mRNA-vaccinated Golden Syrian Hamsters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541294. [PMID: 37425792 PMCID: PMC10327228 DOI: 10.1101/2023.05.22.541294] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Hybrid immunity to SARS-CoV-2 provides superior protection to re-infection. We performed immune profiling studies during breakthrough infections in mRNA-vaccinated hamsters to evaluate hybrid immunity induction. mRNA vaccine, BNT162b2, was dosed to induce binding antibody titers against ancestral spike, but inefficient serum virus neutralization of ancestral SARS-CoV-2 or variants of concern (VoCs). Vaccination reduced morbidity and controlled lung virus titers for ancestral virus and Alpha but allowed breakthrough infections in Beta, Delta and Mu-challenged hamsters. Vaccination primed T cell responses that were boosted by infection. Infection back-boosted neutralizing antibody responses against ancestral virus and VoCs. Hybrid immunity resulted in more cross-reactive sera. Transcriptomics post-infection reflects both vaccination status and disease course and suggests a role for interstitial macrophages in vaccine-mediated protection. Therefore, protection by vaccination, even in the absence of high titers of neutralizing antibodies in the serum, correlates with recall of broadly reactive B- and T-cell responses.
Collapse
Affiliation(s)
- Juan García-Bernalt Diego
- Infectious and Tropical Diseases Research Group (e-INTRO), Biomedical Research Institute of Salamanca-Research Centre for Tropical Diseases at the University of Salamanca (IBSAL-CIETUS), Faculty of Pharmacy, University of Salamanca, Spain
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Gagandeep Singh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Sonia Jangra
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Kim Handrejk
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Manon Laporte
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sara S El Zahed
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Lars Pache
- NCI Designated Cancer Center, Sanford-Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Max W Chang
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Sadaf Aslam
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Ignacio Mena
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Brett T Webb
- Department of Veterinary Sciences, University of Wyoming, Laramie, WY, USA
| | - Christopher Benner
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, NY, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai New York, NY, USA
| |
Collapse
|
32
|
Schulze-Wundling K, Frank Ottensmeyer P, Maria Meyer-Schlinkmann K, Deckena M, Krüger S, Schlinkert S, Budde A, Münstermann D, Töpfner N, Petersmann A, Nauck M, Karch A, Lange B, Blaschke S, Tiemann C, Streeck H. Immunity Against SARS-CoV-2 in the German Population. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:337-344. [PMID: 37155224 PMCID: PMC10408282 DOI: 10.3238/arztebl.m2023.0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 04/08/2023]
Abstract
BACKGROUND Early during the SARS-CoV-2 pandemic, national population-based seroprevalence surveys were conducted in some countries; however, this was not done in Germany. In particular, no seroprevalence surveys were planned for the summer of 2022. In the context of the IMMUNEBRIDGE project, the GUIDE study was carried out to estimate seroprevalence on the national and regional levels. METHODS To obtain an overview of the population-wide immunity against SARS-CoV-2 among adults in Germany that would be as statistically robust as possible, serological tests were carried out using self-sampling dried blood spot cards in conjunction with surveys, one by telephone and one online. Blood samples were analyzed for the presence of antibodies to the S and N antigens of SARS-CoV-2. RESULTS Among the 15 932 participants, antibodies to the S antigen were detected in 95.7%, and to the N antigen in 44.4%. In the higher-risk age groups of persons aged 65 and above and persons aged 80 and above, anti-S antibodies were found in 97,4% and 98.8%, respectively. Distinct regional differences in the distribution of anti-S and anti-N antibodies emerged. Immunity gaps were found both regionally and in particular subgroups of the population. High anti-N antibody levels were especially common in eastern German states, and high anti-S antibody levels in western German states. CONCLUSION These findings indicate that a large percentage of the adult German population has formed antibodies against the SARS-CoV-2 virus. This will markedly lower the probability of an overburdening of the health care system by hospitalization and high occupancy of intensive care units due to future SARS-CoV-2 waves, depending on the viral characteristics of then prevailing variants.
Collapse
Affiliation(s)
- Kai Schulze-Wundling
- Institute of Virology, University Hospital, Faculty of Medicine, Rhenish Friedrich Wilhelm University Bonn
- German Center for Infection Research (DZIF), Locations: Bonn – Cologne, Hannover – Brunswick
| | - Patrick Frank Ottensmeyer
- Institute of Virology, University Hospital, Faculty of Medicine, Rhenish Friedrich Wilhelm University Bonn
- German Center for Infection Research (DZIF), Locations: Bonn – Cologne, Hannover – Brunswick
| | | | - Marek Deckena
- Medical Care Center Laboratory Krone GbR, Bad Salzuflen
| | - Stefan Krüger
- dimap – Institute for Market and Political Research, Bonn: Stefan Krüger, Simon Schlinkert
| | - Simon Schlinkert
- dimap – Institute for Market and Political Research, Bonn: Stefan Krüger, Simon Schlinkert
| | - Axel Budde
- Institute of Virology, University Hospital, Faculty of Medicine, Rhenish Friedrich Wilhelm University Bonn
- German Center for Infection Research (DZIF), Locations: Bonn – Cologne, Hannover – Brunswick
| | | | - Nicole Töpfner
- Center for Pediatric and Adolescent Medicine, University Hospital, Faculty of Medicine Carl Gustav Carus, TU Dresden
| | - Astrid Petersmann
- Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Oldenburg
| | - Matthias Nauck
- Institute for Clinical Chemistry and Laboratory Medicine, Greifswald Medical School, and German Center for Cardiovascular Research (DZHK), Location Greifswald, Greifswald Medical School
| | - André Karch
- Institute of Epidemiology and Social Medicine, Faculty of Medicine, Westphalian Wilhelms University of Münster
| | - Berit Lange
- Institute of Virology, University Hospital, Faculty of Medicine, Rhenish Friedrich Wilhelm University Bonn
| | | | | | - Hendrik Streeck
- Institute of Virology, University Hospital, Faculty of Medicine, Rhenish Friedrich Wilhelm University Bonn
- German Center for Infection Research (DZIF), Locations: Bonn – Cologne, Hannover – Brunswick
| |
Collapse
|
33
|
Huang C, Wei Y, Yan VKC, Ye X, Kang W, Yiu HHE, Shami JJP, Cowling BJ, Tse ML, Castle DJ, Chui CSL, Lai FTT, Li X, Wan EYF, Wong CKH, Hayes JF, Chang WC, Chung AKK, Lau CS, Wong ICK, Chan EW. Vaccine effectiveness of BNT162b2 and CoronaVac against SARS-CoV-2 omicron infection and related hospital admission among people with substance use disorder in Hong Kong: a matched case-control study. Lancet Psychiatry 2023; 10:403-413. [PMID: 37141907 DOI: 10.1016/s2215-0366(23)00111-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/15/2023] [Accepted: 03/20/2023] [Indexed: 05/06/2023]
Abstract
BACKGROUND People with substance use disorder have a high risk of SARS-CoV-2 infection and subsequent poor outcomes. Few studies have evaluated COVID-19 vaccine effectiveness among people with substance use disorder. We aimed to estimate the vaccine effectiveness of BNT162b2 (Fosun-BioNTech) and CoronaVac (Sinovac) against SARS-CoV-2 omicron (B.1.1.529) infection and related hospital admission in this population. METHODS We did a matched case-control study using electronic health databases in Hong Kong. Individuals diagnosed with substance use disorder between Jan 1, 2016, and Jan 1, 2022, were identified. People aged 18 years and older with SARS-CoV-2 infection from Jan 1 to May 31, 2022, and people with COVID-19-related hospital admission from Feb 16 to May 31, 2022, were included as cases and were matched by age, sex, and previous clinical history with controls from all individuals diagnosed with substance use disorder who attended the Hospital Authority health services: up to three controls for SARS-CoV-2 infection and up to ten controls for hospital admission. Conditional logistical regression was used to evaluate the association between vaccination status (ie, one, two, or three doses of BNT162b2 or CoronaVac) and the risk of SARS-CoV-2 infection and COVID-19-related hospital admission, adjusted for baseline comorbidities and medication use. FINDINGS Among 57 674 individuals with substance use disorder, 9523 people with SARS-CoV-2 infections (mean age 61·00 years, SD 14·90; 8075 [84·8%] males and 1448 [15·2%] females) were identified and matched to 28 217 controls (mean age 60·99 years, 14·67; 24 006 [85·1%] males and 4211 [14·9%] females), and 843 people with COVID-19-related hospital admissions (mean age 70·48 years, SD 14·68; 754 [89·4%] males and 89 [10·6%] females) were identified and matched to 7459 controls (mean age 70·24 years, 13·87; 6837 [91·7%] males and 622 [8·3%] females). Data on ethnicity were not available. We observed significant vaccine effectiveness against SARS-CoV-2 infection for two-dose BNT162b2 vaccination (20·7%, 95% CI 14·0-27·0, p<0·0001) and three-dose vaccination (all BNT162b2 41·5%, 34·4-47·8, p<0·0001; all CoronaVac 13·6%, 5·4-21·0, p=0·0015; BNT162b2 booster after two-dose CoronaVac 31·3%, 19·8-41·1, p<0·0001), but not for one dose of either vaccine or two doses of CoronaVac. Significant vaccine effectiveness against COVID-19-related hospital admission was detected after one dose of BNT162b2 vaccination (35·7%, 3·8-57·1, p=0·032), two-dose vaccination (both BNT162b2 73·3%, 64·3 to 80·0, p<0·0001; both CoronaVac 59·9%, 50·2-67·7, p<0·0001), and three-dose vaccination (all BNT162b2 86·3%, 75·6-92·3, p<0·0001; all CoronaVac 73·5% 61·0-81·9, p<0·0001; BNT162b2 booster after two-dose CoronaVac 83·7%, 64·6-92·5, p<0·0001), but not after one dose of CoronaVac. INTERPRETATION For both BNT162b2 and CoronaVac, two-dose or three-dose vaccination was protective against COVID-19-related hospital admission and the booster dose provided protection against SARS-CoV-2 infection among people with substance use disorder. Our findings confirm the importance of booster doses in this population during the period dominated by the omicron variant. FUNDING Health Bureau, the Government of the Hong Kong Special Administrative Region.
Collapse
Affiliation(s)
- Caige Huang
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yue Wei
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Vincent K C Yan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Xuxiao Ye
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wei Kang
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Hei Hang Edmund Yiu
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jessica J P Shami
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Benjamin J Cowling
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, University of Hong Kong, Hong Kong Special Administrative Region, China; School of Public Health, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Man Li Tse
- Hong Kong Poison Information Centre, United Christian Hospital, Hong Kong Special Administrative Region, China
| | - David J Castle
- Centre for Addiction and Mental Health and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Celine S L Chui
- School of Public Health, University of Hong Kong, Hong Kong Special Administrative Region, China; School of Nursing, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Francisco T T Lai
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Xue Li
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Eric Y F Wan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Family Medicine and Primary Care, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Carlos K H Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Family Medicine and Primary Care, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China
| | - Joseph F Hayes
- Division of Psychiatry, University College London, London, UK
| | - Wing Chung Chang
- School of Clinical Medicine, Li Ka Shing Faculty of Medicine, Department of Psychiatry, Queen Mary Hospital, University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Brain and Cognitive Sciences, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Albert K K Chung
- School of Clinical Medicine, Li Ka Shing Faculty of Medicine, Department of Psychiatry, Queen Mary Hospital, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Chak Sing Lau
- Department of Medicine, University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Ian C K Wong
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China; Aston School of Pharmacy, Aston University, Birmingham, UK; Expert Committee on Clinical Events Assessment Following COVID-19 Immunization, Department of Health, The Government of the Hong Kong SAR, Hong Kong Special Administrative Region, China
| | - Esther W Chan
- Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong Special Administrative Region, China; Laboratory of Data Discovery for Health (D(2)4H), Hong Kong Science and Technology Park, Hong Kong Special Administrative Region, China; Department of Pharmacy, Shenzhen Hospital and Shenzhen Institute of Research and Innovation, University of Hong Kong, Shenzhen, China.
| |
Collapse
|
34
|
Amemiya Y, Li T, Nishiura H. Age-dependent final size equation to anticipate mortality impact of COVID-19 in China. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:11353-11366. [PMID: 37322985 DOI: 10.3934/mbe.2023503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Before reopening society in December 2022, China had not achieved sufficiently high vaccination coverage among people aged 80 years and older, who are vulnerable to severe infection and death owing to COVID-19. Suddenly ending the zero-COVID policy was anticipated to lead to substantial mortality. To investigate the mortality impact of COVID-19, we devised an age-dependent transmission model to derive a final size equation, permitting calculation of the expected cumulative incidence. Using an age-specific contact matrix and published estimates of vaccine effectiveness, final size was computed as a function of the basic reproduction number, R0. We also examined hypothetical scenarios in which third-dose vaccination coverage was increased in advance of the epidemic, and also in which mRNA vaccine was used instead of inactivated vaccines. Without additional vaccination, the final size model indicated that a total of 1.4 million deaths (half of which were among people aged 80 years and older) were anticipated with an assumed R0 of 3.4. A 10% increase in third-dose coverage would prevent 30,948, 24,106, and 16,367 deaths, with an assumed second-dose effectiveness of 0%, 10%, and 20%, respectively. With mRNA vaccine, the mortality impact would have been reduced to 1.1 million deaths. The experience of reopening in China indicates the critical importance of balancing pharmaceutical and non-pharmaceutical interventions. Ensuring sufficiently high vaccination coverage is vital in advance of policy changes.
Collapse
Affiliation(s)
- Yuri Amemiya
- Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tianwen Li
- Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| | - Hiroshi Nishiura
- Kyoto University School of Public Health, Yoshida-Konoe-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
35
|
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
|
36
|
Nichols JH, Williams EP, Parvathareddy J, Cao X, Kong Y, Fitzpatrick E, Webby RJ, Jonsson CB. Upper Respiratory Infection Drives Clinical Signs and Inflammatory Responses Following Heterologous Challenge of SARS-CoV-2 Variants of Concern in K18 Mice. Viruses 2023; 15:v15040946. [PMID: 37112926 PMCID: PMC10144791 DOI: 10.3390/v15040946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
The evolution of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in the emergence of several variants of concern (VOC) with increased immune evasion and transmissibility. This has motivated studies to assess protection conferred by earlier strains following infection or vaccination to each new VOC. We hypothesized that while NAbs play a major role in protection against infection and disease, a heterologous reinfection or challenge may gain a foothold in the upper respiratory tract (URT) and result in a self-limited viral infection accompanied by an inflammatory response. To test this hypothesis, we infected K18-hACE2 mice with SARS-CoV-2 USA-WA1/2020 (WA1) and, after 24 days, challenged with WA1, Alpha, or Delta. While NAb titers against each virus were similar across all cohorts prior to challenge, the mice challenged with Alpha and Delta showed weight loss and upregulation of proinflammatory cytokines in the URT and lower RT (LRT). Mice challenged with WA1 showed complete protection. We noted increased levels of viral RNA transcripts only in the URT of mice challenged with Alpha and Delta. In conclusion, our results suggested self-limiting breakthrough infections of Alpha or Delta in the URT, which correlated with clinical signs and a significant inflammatory response in mice.
Collapse
Affiliation(s)
- Jacob H Nichols
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Evan P Williams
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jyothi Parvathareddy
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Xueyuan Cao
- Department of Health Promotion and Disease Prevention, College of Nursing, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ying Kong
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Elizabeth Fitzpatrick
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Richard J Webby
- St. Jude Children's Research Hospital, Memphis, TN 38163, USA
| | - Colleen B Jonsson
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN 38163, USA
- Institute for the Study of Host-Pathogen Systems, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
37
|
Kalyoncu S, Yilmaz S, Kuyucu AZ, Sayili D, Mert O, Soyturk H, Gullu S, Akinturk H, Citak E, Arslan M, Taskinarda MG, Tarman IO, Altun GY, Ozer C, Orkut R, Demirtas A, Tilmensagir I, Keles U, Ulker C, Aralan G, Mercan Y, Ozkan M, Caglar HO, Arik G, Ucar MC, Yildirim M, Yildirim TC, Karadag D, Bal E, Erdogan A, Senturk S, Uzar S, Enul H, Adiay C, Sarac F, Ekiz AT, Abaci I, Aksoy O, Polat HU, Tekin S, Dimitrov S, Ozkul A, Wingender G, Gursel I, Ozturk M, Inan M. Process development for an effective COVID-19 vaccine candidate harboring recombinant SARS-CoV-2 delta plus receptor binding domain produced by Pichia pastoris. Sci Rep 2023; 13:5224. [PMID: 36997624 PMCID: PMC10062263 DOI: 10.1038/s41598-023-32021-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 03/21/2023] [Indexed: 04/01/2023] Open
Abstract
Recombinant protein-based SARS-CoV-2 vaccines are needed to fill the vaccine equity gap. Because protein-subunit based vaccines are easier and cheaper to produce and do not require special storage/transportation conditions, they are suitable for low-/middle-income countries. Here, we report our vaccine development studies with the receptor binding domain of the SARS-CoV-2 Delta Plus strain (RBD-DP) which caused increased hospitalizations compared to other variants. First, we expressed RBD-DP in the Pichia pastoris yeast system and upscaled it to a 5-L fermenter for production. After three-step purification, we obtained RBD-DP with > 95% purity from a protein yield of > 1 g/L of supernatant. Several biophysical and biochemical characterizations were performed to confirm its identity, stability, and functionality. Then, it was formulated in different contents with Alum and CpG for mice immunization. After three doses of immunization, IgG titers from sera reached to > 106 and most importantly it showed high T-cell responses which are required for an effective vaccine to prevent severe COVID-19 disease. A live neutralization test was performed with both the Wuhan strain (B.1.1.7) and Delta strain (B.1.617.2) and it showed high neutralization antibody content for both strains. A challenge study with SARS-CoV-2 infected K18-hACE2 transgenic mice showed good immunoprotective activity with no viruses in the lungs and no lung inflammation for all immunized mice.
Collapse
Affiliation(s)
| | - Semiramis Yilmaz
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | | | - Dogu Sayili
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Olcay Mert
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | - Seyda Gullu
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | - Erhan Citak
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- VIB-UGent Center for Medical Biotechnology, Gent, Belgium
| | - Merve Arslan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | | | | | - Ceren Ozer
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Ridvan Orkut
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | | | | | - Umur Keles
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Lund University, Lund, Sweden
| | - Ceren Ulker
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Gizem Aralan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Yavuz Mercan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Muge Ozkan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Hasan Onur Caglar
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Erzurum Technical University, Erzurum, Turkey
| | - Gizem Arik
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Ankara Medipol University, Ankara, Turkey
| | - Mehmet Can Ucar
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Imperial College London, London, UK
| | | | | | | | - Erhan Bal
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir Tinaztepe University, Izmir, Turkey
| | - Aybike Erdogan
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Serif Senturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Serdar Uzar
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Hakan Enul
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Cumhur Adiay
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | - Fahriye Sarac
- Pendik Veterinary Research and Control Institute, Istanbul, Turkey
| | | | - Irem Abaci
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | - Ozge Aksoy
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
| | | | - Saban Tekin
- Marmara Research Center, TUBITAK, Kocaeli, Turkey
- University of Health Sciences, Istanbul, Turkey
| | | | | | | | - Ihsan Gursel
- Izmir Biomedicine and Genome Center, Izmir, Turkey
| | - Mehmet Ozturk
- Izmir Biomedicine and Genome Center, Izmir, Turkey
- Izmir Tinaztepe University, Izmir, Turkey
| | - Mehmet Inan
- Izmir Biomedicine and Genome Center, Izmir, Turkey.
- Akdeniz University, Antalya, Turkey.
| |
Collapse
|
38
|
Vaccine effectiveness against severe COVID-19 during the Omicron wave in Germany: results from the COViK study. Infection 2023:10.1007/s15010-023-02012-z. [PMID: 36913112 PMCID: PMC10009838 DOI: 10.1007/s15010-023-02012-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/22/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE COViK, a prospective hospital-based multicenter case-control study in Germany, aims to assess the effectiveness of COVID-19 vaccines against severe disease. Here, we report vaccine effectiveness (VE) against COVID-19-caused hospitalization and intensive care treatment during the Omicron wave. METHODS We analyzed data from 276 cases with COVID-19 and 494 control patients recruited in 13 hospitals from 1 December 2021 to 5 September 2022. We calculated crude and confounder-adjusted VE estimates. RESULTS 21% of cases (57/276) were not vaccinated, compared to 5% of controls (26/494; p < 0.001). Confounder-adjusted VE against COVID-19-caused hospitalization was 55.4% (95% CI: 12-78%), 81.5% (95% CI: 68-90%) and 95.6% (95%CI: 88-99%) after two, three and four vaccine doses, respectively. VE against hospitalization due to COVID-19 remained stable up to one year after three vaccine doses. CONCLUSION Three vaccine doses remained highly effective in preventing severe disease and this protection was sustained; a fourth dose further increased protection.
Collapse
|
39
|
Kohli MA, Maschio M, Joshi K, Lee A, Fust K, Beck E, Van de Velde N, Weinstein MC. The potential clinical impact and cost-effectiveness of the updated COVID-19 mRNA fall 2023 vaccines in the United States. J Med Econ 2023; 26:1532-1545. [PMID: 37961887 DOI: 10.1080/13696998.2023.2281083] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023]
Abstract
AIMS To assess the potential clinical impact and cost-effectiveness of COVID-19 mRNA vaccines updated for fall 2023 in adults aged ≥18 years over a 1-year analytic time horizon (September 2023-August 2024). MATERIALS AND METHODS A compartmental Susceptible-Exposed-Infected-Recovered model was updated to reflect COVID-19 cases in summer 2023. The numbers of symptomatic infections, COVID-19-related hospitalizations and deaths, and costs and quality-adjusted life-years (QALYs) gained were calculated using a decision tree model. The incremental cost-effectiveness ratio (ICER) of a Moderna updated mRNA fall 2023 vaccine (Moderna Fall Campaign) was compared to no additional vaccination. Potential differences between the Moderna and the Pfizer-BioNTech fall 2023 vaccines were also examined. RESULTS Base case results suggest that the Moderna Fall Campaign would decrease the expected 64.2 million symptomatic infections by 7.2 million (11%) to 57.0 million. COVID-19-related hospitalizations and deaths are expected to decline by 343,000 (-29%) and 50,500 (-33%), respectively. The Moderna Fall Campaign would increase QALYs by 740,880 and healthcare costs by $5.7 billion relative to no vaccine, yielding an ICER of $7700 per QALY gained. Using a societal cost perspective, the ICER is $2100. Sensitivity analyses suggest that vaccine effectiveness, COVID-19 incidence, hospitalization rates, and costs drive cost-effectiveness. With a relative vaccine effectiveness of 5.1% for infection and 9.8% for hospitalization for the Moderna vaccine versus the Pfizer-BioNTech vaccine, use of the Moderna vaccine is expected to prevent 24,000 more hospitalizations and 3300 more deaths than the Pfizer-BioNTech vaccine. LIMITATIONS AND CONCLUSIONS As COVID-19 becomes endemic, future incidence, including patterns of infection, are highly uncertain. The effectiveness of fall 2023 vaccines is unknown, and it is unclear when a new variant that evades natural or vaccine immunity will emerge. Despite these limitations, our model predicts the Moderna Fall Campaign vaccine is highly cost-effective across all sensitivity analyses.
Collapse
Affiliation(s)
| | | | | | - Amy Lee
- Quadrant Health Economics Inc., Cambridge, ON, Canada
| | - Kelly Fust
- Quadrant Health Economics Inc., Cambridge, ON, Canada
| | | | | | | |
Collapse
|
40
|
Li Y, Liu Y, Duo M, Wu R, Jiang T, Li P, Wang Y, Cheng Z. Bioinformatic analysis and preliminary validation of potential therapeutic targets for COVID-19 infection in asthma patients. Cell Commun Signal 2022; 20:201. [PMID: 36575422 PMCID: PMC9793391 DOI: 10.1186/s12964-022-01010-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/03/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 causes coronavirus disease 19 (COVID-19). The number of confirmed cases of COVID-19 is also rapidly increasing worldwide, posing a significant challenge to human safety. Asthma is a risk factor for COVID-19, but the underlying molecular mechanisms of the asthma-COVID-19 interaction remain unclear. METHODS We used transcriptome analysis to discover molecular biomarkers common to asthma and COVID-19. Gene Expression Omnibus database RNA-seq datasets (GSE195599 and GSE196822) were used to identify differentially expressed genes (DEGs) in asthma and COVID-19 patients. After intersecting the differentially expressed mRNAs, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed to identify the common pathogenic molecular mechanism. Bioinformatic methods were used to construct protein-protein interaction (PPI) networks and identify key genes from the networks. An online database was used to predict interactions between transcription factors and key genes. The differentially expressed long noncoding RNAs (lncRNAs) in the GSE195599 and GSE196822 datasets were intersected to construct a competing endogenous RNA (ceRNA) regulatory network. Interaction networks were constructed for key genes with RNA-binding proteins (RBPs) and oxidative stress-related proteins. The diagnostic efficacy of key genes in COVID-19 was verified with the GSE171110 dataset. The differential expression of key genes in asthma was verified with the GSE69683 dataset. An asthma cell model was established with interleukins (IL-4, IL-13 and IL-17A) and transfected with siRNA-CXCR1. The role of CXCR1 in asthma development was preliminarily confirmed. RESULTS By intersecting the differentially expressed genes for COVID-19 and asthma, 393 common DEGs were obtained. GO and KEGG enrichment analyses of the DEGs showed that they mainly affected inflammation-, cytokine- and immune-related functions and inflammation-related signaling pathways. By analyzing the PPI network, we obtained 10 key genes: TLR4, TLR2, MMP9, EGF, HCK, FCGR2A, SELP, NFKBIA, CXCR1, and SELL. By intersecting the differentially expressed lncRNAs for COVID-19 and asthma, 13 common differentially expressed lncRNAs were obtained. LncRNAs that regulated microRNAs (miRNAs) were mainly concentrated in intercellular signal transduction, apoptosis, immunity and other related functional pathways. The ceRNA network suggested that there were a variety of regulatory miRNAs and lncRNAs upstream of the key genes. The key genes could also bind a variety of RBPs and oxidative stress-related genes. The key genes also had good diagnostic value in the verification set. In the validation set, the expression of key genes was statistically significant in both the COVID-19 group and the asthma group compared with the healthy control group. CXCR1 expression was upregulated in asthma cell models, and interference with CXCR1 expression significantly reduced cell viability. CONCLUSIONS Key genes may become diagnostic and predictive biomarkers of outcomes in COVID-19 and asthma. Video Abstract.
Collapse
Affiliation(s)
- Yue Li
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Ye Liu
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Mengjie Duo
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Ruhao Wu
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Tianci Jiang
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Pengfei Li
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yu Wang
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Zhe Cheng
- grid.412633.10000 0004 1799 0733Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
41
|
Brook B, Fatou B, Kumar Checkervarty A, Barman S, Sweitzer C, Bosco AN, Sherman AC, Baden LR, Morrocchi E, Sanchez-Schmitz G, Palma P, Nanishi E, O'Meara TR, McGrath ME, Frieman MB, Soni D, van Haren SD, Ozonoff A, Diray-Arce J, Steen H, Dowling DJ, Levy O. The mRNA vaccine BNT162b2 demonstrates impaired T H1 immunogenicity in human elders in vitro and aged mice in vivo. RESEARCH SQUARE 2022:rs.3.rs-2395118. [PMID: 36597547 PMCID: PMC9810224 DOI: 10.21203/rs.3.rs-2395118/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
mRNA vaccines have been key to addressing the SARS-CoV-2 pandemic but have impaired immunogenicity and durability in vulnerable older populations. We evaluated the mRNA vaccine BNT162b2 in human in vitro whole blood assays with supernatants from adult (18-50 years) and elder (≥60 years) participants measured by mass spectrometry and proximity extension assay proteomics. BNT162b2 induced increased expression of soluble proteins in adult blood (e.g., C1S, PSMC6, CPN1), but demonstrated reduced proteins in elder blood (e.g., TPM4, APOF, APOC2, CPN1, and PI16), including 30-85% lower induction of TH1-polarizing cytokines and chemokines (e.g., IFNγ, and CXCL10). Elder TH1 impairment was validated in mice in vivo and associated with impaired humoral and cellular immunogenicity. Our study demonstrates the utility of a human in vitro platform to model age-specific mRNA vaccine activity, highlights impaired TH1 immunogenicity in older adults, and provides rationale for developing enhanced mRNA vaccines with greater immunogenicity in vulnerable populations.
Collapse
Affiliation(s)
- Byron Brook
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Benoit Fatou
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Abhinav Kumar Checkervarty
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Prevention of Organ Failure (PROOF) Centre of Excellence, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada
- UBC Centre for Heart Lung Innovation, Providence Research, St Paul's Hospital, Vancouver, BC, Canada
| | - Soumik Barman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Cali Sweitzer
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Anna-Nicole Bosco
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Amy C Sherman
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Lindsey R Baden
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Elena Morrocchi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Guzman Sanchez-Schmitz
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Bambino Gesù Children's Hospital, Rome, Italy
- Chair of Pediatrics, University of Rome, Tor Vergata, Italy
| | - Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Timothy R O'Meara
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Marisa E McGrath
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Matthew B Frieman
- Center for Pathogen Research, Department of Microbiology and Immunology, The University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dheeraj Soni
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Joann Diray-Arce
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
42
|
Mollura M, Sarti R, Levi R, Pozzi C, Azzolini E, Politi LS, Mantovani A, Barbieri R, Rescigno M. Antibody Titer Correlates with Omicron Infection in Vaccinated Healthcare Workers. Viruses 2022; 14:2605. [PMID: 36560609 PMCID: PMC9782630 DOI: 10.3390/v14122605] [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: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
The advent of vaccines against SARS-CoV-2 has drastically reduced the level of hospitalization with severe COVID-19 disease in infected individuals. However, the diffusion of variants of concern still challenge the protection conferred by vaccines raised against the wild-type form of the virus. Here, we have characterized the antibody response to the BNT162b2 (Comirnaty) mRNA vaccine in patients infected with the Omicron variant. We analyzed a population of 4354 vaccinated healthcare workers (HCW) from 7 different hospitals in Italy and monitored infection with SARS-CoV-2 Omicron. We correlated infection with the antibody response after vaccination. We found that a lower level of IgG, younger age, and the presence of allergies correlate with increased infection during the Omicron wave, and that infections correlate with wild-type spike protein antibody titers below 350 BAU/mL. These results support the necessity of a fourth booster dose, particularly for individuals with lower levels of antibodies.
Collapse
Affiliation(s)
- Maximiliano Mollura
- Department of Electronic, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Riccardo Sarti
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Riccardo Levi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Chiara Pozzi
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Elena Azzolini
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Letterio S. Politi
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Alberto Mantovani
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London E1 4NS, UK
| | - Riccardo Barbieri
- Department of Electronic, Information and Bioengineering, Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Maria Rescigno
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| |
Collapse
|
43
|
Li Y, Guo T, Zhong J, Fang C, Xiong H, Hu Z, Zhu Y, Tan J, Liu S, Jing Q, Zhang D. Effect of Vaccination Time Intervals on SARS-COV-2 Omicron Variant Strain Infection in Guangzhou: A Real-World Matched Case–Control Study. Vaccines (Basel) 2022; 10:vaccines10111855. [PMID: 36366363 PMCID: PMC9693306 DOI: 10.3390/vaccines10111855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
In April 2022, a COVID-19 outbreak caused by the Omicron variant emerged in Guangzhou. A case–control study was conducted to explore the relationship between vaccination intervals and SARS-CoV-2 infection in the real world. According to the vaccination dose and age information of the cases, a 1:4 matched case–control sample was established, finally including n = 242 for the case group and n = 968 for the control group. The results indicated that among the participants who received three vaccine doses, those with an interval of more than 300 days between the receipt of the first vaccine dose and infection (or the first contact with a confirmed case) were less likely to be infected with SARS-CoV-2 than those with an interval of less than 300 days (OR = 0.67, 95% CI = 0.46–0.99). After age-stratified analysis, among participants aged 18–40 years who received two doses of vaccine, those who received the second dose more than 30 days after the first dose were less likely to be infected with SARS-CoV-2 (OR = 0.53, 95% CI = 0.30–0.96). Our findings suggest that we need to extend the interval between the first dose and the second dose and further explore the optimal interval between the first and second and between the second and third doses in order to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yufen Li
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tong Guo
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
- Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
- Guangzhou Center for Disease Control and Prevention, Institute of Public Health, Guangzhou Medical University, Guangzhou 510180, China
| | - Jiayi Zhong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Chuanjun Fang
- Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
- Guangzhou Center for Disease Control and Prevention, Institute of Public Health, Guangzhou Medical University, Guangzhou 510180, China
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Husheng Xiong
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Zengyun Hu
- State Key Laboratory of desert and Oasis Ecology, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Urumqi 830011, China
| | - Yajuan Zhu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jinlin Tan
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuang Liu
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qinlong Jing
- Guangzhou Center for Disease Control and Prevention, Guangzhou 510440, China
- Guangzhou Center for Disease Control and Prevention, Institute of Public Health, Guangzhou Medical University, Guangzhou 510180, China
- Correspondence: (Q.J.); (D.Z.)
| | - Dingmei Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-Sen University, Guangzhou 510080, China
- NMPA Key Laboratory for Quality Monitoring and Evaluation of Vaccines and Biological Products, Guangzhou 510080, China
- Correspondence: (Q.J.); (D.Z.)
| |
Collapse
|
44
|
Cardoso CW, Souza MMDS, Teles ACVDS, Argibay HD, Reis OBD, Pereira FM, Giovanetti M, Magalhaes T, Ribeiro GS. COVID-19 outbreaks among crew members in non-cruise vessels anchoring in Salvador, Brazil, 2021. Mem Inst Oswaldo Cruz 2022; 117:e220114. [DOI: 10.1590/0074-02760220114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 10/10/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - Tereza Magalhaes
- Universidade Federal da Bahia, Brazil; Colorado State University, USA
| | | |
Collapse
|