1
|
Wyndham-Thomas C, Newbern EC, Mira-Iglesias A, Dwivedi A, Orrico Sánchez A, Antón A, Martin C, Icardi G, Casas I, Ngew KY, Drikite L, de Munter L, Ten Kate GL, Vroom N, Baumgartner S, Otero-Romero S, Holemans X, Bollaerts K, Praet N. Vaccine effectiveness of JCOVDEN single-dose against COVID-19 hospitalisation in Europe: An id.DRIVE test-negative case-control study. J Infect Public Health 2025; 18:102700. [PMID: 40014936 DOI: 10.1016/j.jiph.2025.102700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND JCOVDEN (Ad26.COV2.S), a viral-vector vaccine, was granted conditional marketing authorisation in the European Union for the prevention of COVID-19 in early 2021. We present JCOVDEN single-dose vaccine effectiveness (VE) estimates against COVID-19 hospitalisation. METHODS The id.DRIVE (previously COVIDRIVE) COVID-19 VE study is an ongoing European non-interventional, multi-centre study with a test-negative case-control design. Study participants were adults ≥ 18 years old, hospitalised with severe acute respiratory infection between 1 May 2021 and 28 February 2023. Estimated as a single measure over the entire study period, VE was stratified by risk group, time since vaccination intervals (14 days-12 weeks, 12-to-25 weeks, 25-to-52 weeks, >52 weeks), SARS-CoV-2 variant and calendar time categories. All estimates were adjusted for symptom-onset date, age, sex, and number of pre-defined chronic conditions. RESULTS Overall, VE was 55.6 % (95 % CI 23.6; 74.2) for a median time since vaccination of 146 days. For 18- to 49-year-olds, VE was 61.6 % (95 % CI 16.2; 82.4), 57.7 % (95 % CI 3.4; 81.5) for 50- to-64-years-olds, and 40.8 % (95 % CI -6.0; 66.9) for ≥ 65-year-olds. Most precise estimates were obtained for time since vaccination 12-to- 25-week interval (59.2 % [95 % CI 25.0; 77.8]) and for the calendar time period 1 Aug 2021 -30 Nov 2021 (Delta predominant; 51.2 % [95 % CI 21.7; 69.6]). CONCLUSION The JCOVDEN single-dose protected against COVID-19 hospitalisation. It is effective for at least six months, with VE estimates comparatively lower in the older age groups. Results had low to medium levels of certainty and are to be interpreted with caution.
Collapse
Affiliation(s)
| | - E Claire Newbern
- Janssen Research & Development, LLC, 200 Tournament Dr, Horsham, PA 19044, United States
| | - Ainara Mira-Iglesias
- Valencia Hospital Network for the Study of Infectious Diseases, Fundación Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Av. de Catalunya 21, Valencia 46020, Spain; Vaccine Research Department of Fisabio-Public Health, Valencia, Spain; CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - Akshat Dwivedi
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | - Alejandro Orrico Sánchez
- Valencia Hospital Network for the Study of Infectious Diseases, Fundación Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana, Av. de Catalunya 21, Valencia 46020, Spain; Vaccine Research Department of Fisabio-Public Health, Valencia, Spain; CIBER de Epidemiología y Salud Pública, Instituto de Salud Carlos III, Madrid, Spain
| | - Andrés Antón
- Hospital Universitari Vall d'Hebron, Paseo Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Charlotte Martin
- Le Centre Hospitalier Universitaire St Pierre, Rue Haute 322, Brussels 1000, Belgium
| | - Giancarlo Icardi
- Centro Interuniversitario per la Ricerca sull'Influenza e le altre Infezioni Trasmissibili Largo R. Benzi 10, Genoa 16132, Italy
| | - Irma Casas
- Hospital Universitari Germans Trias i Pujol, Carretera del Canyet, Badalona, Barcelona 08916, Spain
| | - Kok Yew Ngew
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | - Laura Drikite
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | - Leonie de Munter
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | | | - Nikki Vroom
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | | | - Susana Otero-Romero
- Hospital Universitari Vall d'Hebron, Paseo Vall d'Hebron 119-129, Barcelona 08035, Spain
| | - Xavier Holemans
- Grand Hôpital de Charleroi, Grand Rue 3, Charleroi B-6000, Belgium
| | - Kaatje Bollaerts
- P95 Clinical and Epidemiology Services, Diestsevest 125, Leuven 3000, Belgium
| | - Nicolas Praet
- Janssen Research & Development, LLC, 200 Tournament Dr, Horsham, PA 19044, United States; Janssen Research & Development, LLC, 2340, Turnhoutseweg 30, Beerse 2340, Belgium.
| |
Collapse
|
2
|
Dai ZX, Lan HJ, Hai N, Wang JY, Wang HH. Balancing fairness and efficiency in dynamic vaccine allocation during major infectious disease outbreaks. Sci Rep 2025; 15:1371. [PMID: 39779729 PMCID: PMC11711769 DOI: 10.1038/s41598-024-84027-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
The outbreak of novel infectious diseases presents major public health challenges, highlighting the urgency of accelerating vaccination efforts to reduce morbidity and mortality. Vaccine allocation has become a crucial societal concern. This paper introduces a dynamic vaccine allocation model that considers demand uncertainty and vaccination willingness, focusing on the trade-off between fairness and efficiency. We develop a multi-period dynamic vaccine allocation model, evaluating optimal strategies over different periods. The model addresses structural differences among vaccination groups, strategy selection, dynamic demand, and vaccination willingness. Our findings suggest that prioritizing efficiency in the initial outbreak stages may lead to inequitable distribution, causing adverse social impacts, while overemphasizing fairness can undermine overall utility. Therefore, we propose a dynamic optimization-based strategy balancing fairness and efficiency at different pandemic stages. Our results indicate that allocation strategies should shift from efficiency to fairness as the pandemic evolves to enhance vaccine utility. Additionally, macro-level interventions like reducing free-rider behavior and increasing vaccination convenience can improve total vaccine utility. This study offers new perspectives and methodologies for dynamic vaccine allocation, highlighting the trade-off between fairness and efficiency, providing crucial insights for policy formulation and pandemic response.
Collapse
Affiliation(s)
- Zi-Xuan Dai
- School of Economics and Management, Beijing Jiaotong University, No.3 Shangyuan Cun, Haidian District, Beijing, 100044, People's Republic of China
| | - Hong-Jie Lan
- School of Economics and Management, Beijing Jiaotong University, No.3 Shangyuan Cun, Haidian District, Beijing, 100044, People's Republic of China.
| | - Nan Hai
- School of Economics and Management, Beijing Jiaotong University, No.3 Shangyuan Cun, Haidian District, Beijing, 100044, People's Republic of China
| | - Jia-Yuan Wang
- School of Economics and Management, Beijing Jiaotong University, No.3 Shangyuan Cun, Haidian District, Beijing, 100044, People's Republic of China
| | - Huan-Huan Wang
- School of Economics and Management, Beijing Jiaotong University, No.3 Shangyuan Cun, Haidian District, Beijing, 100044, People's Republic of China
| |
Collapse
|
3
|
Patel DA, Marcum ZA, Chansakul A, Toyip A, Nerney K, Panozzo CA, St Laurent S, Mehta D, Ghaswalla P. Economic burden of cardiorespiratory hospitalizations associated with respiratory syncytial virus among United States adults in 2017-2019. Hum Vaccin Immunother 2024; 20:2364493. [PMID: 38982719 PMCID: PMC11238920 DOI: 10.1080/21645515.2024.2364493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/02/2024] [Indexed: 07/11/2024] Open
Abstract
Morbidity and mortality caused by respiratory syncytial virus (RSV) in older adults and those with underlying health conditions can be potentially alleviated through vaccination. To assist vaccine policy decision-makers and payers, we estimated the annual economic burden of RSV-associated cardiorespiratory hospitalizations among insured US adults aged ≥18 y in the Merative MarketScan claims database from September through August of 2017-2018 and 2018-2019. Negative binomial regression models were used to estimate the number of RSV-associated cardiorespiratory hospitalizations using MarketScan-identified cardiorespiratory diagnosis codes in the presence or absence of RSV circulation per weekly laboratory test positivity percentages from the Centers for Disease Control and Prevention. This number was multiplied by mean cardiorespiratory hospitalization costs to estimate total costs for RSV-associated cardiorespiratory hospitalizations. Number and cost for International Classification of Diseases (ICD)-coded RSV hospitalizations were quantified from MarketScan. In 2017-2018 and 2018-2019, respectively, 18,515,878 and 16,462,120 adults with commercial or Medicare supplemental benefits were assessed. In 2017-2018, 301,248 cardiorespiratory hospitalizations were observed; 0.32% had RSV-specific ICD codes, costing $44,916,324, and 5.52% were RSV-associated cardiorespiratory hospitalizations, costing $734,078,602 (95% CI: $460,826,580-$1,103,358,799). In 2018-2019, 215,525 cardiorespiratory hospitalizations were observed; 0.34% had RSV-specific ICD codes, costing $33,053,105, and 3.14% were RSV-associated cardiorespiratory hospitalizations, costing $287,549,472 (95% CI: $173,377,778-$421,884,259). RSV contributes to substantial economic burden of cardiorespiratory hospitalizations among US adults. Modeling excess risk using viral positivity data provides a comprehensive estimation of RSV hospitalization burden and associated costs, compared with relying on ICD diagnosis codes alone.
Collapse
Affiliation(s)
| | | | | | | | | | - Catherine A Panozzo
- Department of Health Economics and Outcomes Research, Moderna, Inc ., Cambridge, MA, USA
| | - Samantha St Laurent
- Department of Health Economics and Outcomes Research, Moderna, Inc ., Cambridge, MA, USA
| | - Darshan Mehta
- Department of Health Economics and Outcomes Research, Moderna, Inc ., Cambridge, MA, USA
| | - Parinaz Ghaswalla
- Department of Health Economics and Outcomes Research, Moderna, Inc ., Cambridge, MA, USA
| |
Collapse
|
4
|
Boehme A, Harvey RA, Madsen A, Rubens L, Toyip A, Batech M, Ricci D, Nyaku M. An observational post-authorization study to assess the effectiveness of a single dose Ad26.COV2.S for the prevention of COVID-19 using real-world data. Front Public Health 2024; 12:1501919. [PMID: 39697294 PMCID: PMC11652512 DOI: 10.3389/fpubh.2024.1501919] [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: 09/25/2024] [Accepted: 11/21/2024] [Indexed: 12/20/2024] Open
Abstract
Background The goal of this FDA-committed, post authorization study was to assess the real-world effectiveness of Ad26.COV2.S in preventing observed COVID-19 disease in individuals in the United States interacting with the healthcare system who were vaccinated according to the national immunization recommendations. Methods The study cohort consisted of individuals ≥18 years in the U.S. between March 1, 2021 and July 31, 2022. Two exposure groups were considered: those who received a single dose of COVID-19 Ad26.COV2.S vaccine and individuals who were unvaccinated. Individuals eligible for the referent group, defined as those who were unvaccinated, were identified through exact matching on age, sex, location, and Gagne comorbidity score. Propensity-score (PS) matched Cox proportional hazards models were used to evaluate COVID-19 related outcomes. Results A total of 478,162 vaccinated, and 1,897,759 risk set sampled (RSS) and PS-matched unvaccinated referent individuals were included. The vaccine effectiveness (VE) against any observed COVID-19 disease was 20% (95% CI, 19 to 21%). VE increased as the outcome severity increased. The VE against COVID-19 related hospitalizations was 43% (95% CI, 40 to 45%). VE was highest, 53% (95% CI, 42 to 61%), against all-cause mortality temporally associated with COVID-19. The results of subgroup analyses generally showed a similar pattern as the main analyses with VE increasing in parallel with seriousness of outcomes, albeit with lower VE in groups thought to be at higher risk of COVID-19. Discussion This population-representative cohort study in U.S. clinical practice showed that a single dose of Ad26.COV2.S is effective for at least 12 months against several COVID-19 related outcomes. Individuals who were vaccinated with a single dose of Ad26.COV2.S were at lower risk for developing COVID-19, for being hospitalized for COVID-19, and for all-cause mortality temporally associated with COVID-19 compared to unvaccinated individuals in the U.S. during Alpha, Delta, and Omicron BA.1, BA.2, BA.212.1, and BA.5 variants circulation.
Collapse
Affiliation(s)
| | - Raymond A. Harvey
- Johnson & Johnson Innovative Medicine, Spring House, PA, United States
| | - Ann Madsen
- Aetion Inc., New York, NY, United States
| | | | | | | | - Deborah Ricci
- Johnson & Johnson Innovative Medicine, Spring House, PA, United States
| | - Mawuli Nyaku
- Johnson & Johnson Innovative Medicine, Spring House, PA, United States
| |
Collapse
|
5
|
Jain P, Embry A, Arakaki B, Estevez I, Marcum ZA, Viscidi E. Prevalence of Genital Herpes and Antiviral Treatment. Sex Transm Dis 2024; 51:686-693. [PMID: 38691406 DOI: 10.1097/olq.0000000000001990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
BACKGROUND Genital herpes is a common sexually transmitted infection caused by the herpes simplex virus. Contemporary US population-based epidemiologic data on genital herpes are limited. This study aimed to provide nationally representative estimates of genital herpes prevalence and treatment using a large US health insurance claims database. METHODS This observational cohort study used administrative claims data from HealthVerity. Crude and age- and sex-standardized prevalence rates of genital herpes and recurrent genital herpes were calculated for the years 2019 to 2021. The distribution of patients with prevalent genital herpes who received episodic or suppressive antiviral therapy was also estimated. RESULTS From 2019 to 2021, the standardized prevalence of genital herpes and recurrent genital herpes ranged from 236 to 280 cases per 100,000 person-years and 81 to 98 cases per 100,000 person-years, respectively. The prevalence of genital herpes was highest among those aged 25 to 29 years (prevalence range, 497-582 years), female patients (prevalence range, 348-404 years), and those with a history of HIV infection (prevalence range, 1608-2080 years). The prevalence of recurrent genital herpes was also highest in these groups. From 2019 to 2021, two-thirds of patients (65%-68%) with prevalent genital herpes received antiviral medications; the majority received episodic therapy (80%) rather than suppressive therapy (20%). CONCLUSIONS The burden of genital herpes and recurrent genital herpes in the United States is substantial, with the highest rates observed in young adults, women, and immunocompromised individuals. About two-thirds receive antiviral treatment each year.
Collapse
Affiliation(s)
- Purva Jain
- From the Aetion, Inc., New York City, NY
| | | | | | | | | | | |
Collapse
|
6
|
Marcum ZA, Jain P, Embry A, Arakaki B, Estevez I, Viscidi E. Incidence of Herpes Zoster and Postherpetic Neuralgia and Herpes Zoster Vaccination Uptake in a US Administrative Claims Database. Open Forum Infect Dis 2024; 11:ofae211. [PMID: 38737423 PMCID: PMC11083623 DOI: 10.1093/ofid/ofae211] [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: 12/15/2023] [Accepted: 04/11/2024] [Indexed: 05/14/2024] Open
Abstract
Background The objective of this study was to estimate the annual incidence rates of herpes zoster (HZ) and postherpetic neuralgia (PHN) among individuals aged ≥19 years and the proportion who received HZ vaccination among those aged ≥50 years. Methods This observational cohort study was conducted with administrative claims data from HealthVerity and included insured individuals across the US. Crude and US age- and sex-standardized incidence rates of HZ and PHN were calculated from 1 January 2019 to 31 May 2022 by calendar year in persons aged ≥19 years. Outcomes were defined as ≥1 ICD-10 diagnosis code for HZ or PHN. Analyses were stratified by age, sex, and immunocompromised status. Among those aged ≥50 years, the proportion who received 1 or 2 doses of recombinant zoster vaccine (Shingrix) or 1 dose of Zostavax was calculated. Results Standardized annual incidence rates from 2019 to 2021 were 542 to 685 per 100 000 person-years for HZ and 35 to 38 per 100 000 person-years for PHN. Rates were highest among females, older adults, and individuals who were immunocompromised. From 1 January 2019 to 31 May 2022, 4.3% and 9.0% of persons aged ≥50 years received 1 and 2 doses of Shingrix, respectively, and 0.2% received 1 dose of Zostavax. Conclusions In this US claims database analysis, HZ and PHN were more frequent among older adults, females, and individuals who were immunocompromised. Between 1 January 2019 and 31 May 2022, 9% of persons aged ≥50 years received 2 doses of the Shingrix vaccine. Greater efforts are needed to increase vaccine uptake against HZ, especially for those at highest risk.
Collapse
Affiliation(s)
| | - Purva Jain
- Science, Aetion, Inc, New York, New York, USA
| | - Alan Embry
- Clinical Development–Infectious Diseases, Moderna, Inc, Cambridge, Massachusetts, USA
| | | | | | - Emma Viscidi
- Clinical Development–Infectious Diseases, Moderna, Inc, Cambridge, Massachusetts, USA
| |
Collapse
|
7
|
Zhu C, Pang S, Liu J, Duan Q. Current Progress, Challenges and Prospects in the Development of COVID-19 Vaccines. Drugs 2024; 84:403-423. [PMID: 38652356 DOI: 10.1007/s40265-024-02013-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2024] [Indexed: 04/25/2024]
Abstract
The COVID-19 pandemic has resulted in over 772 million confirmed cases, including nearly 7 million deaths, according to the World Health Organization (WHO). Leveraging rapid development, accelerated vaccine approval processes, and large-scale production of various COVID-19 vaccines using different technical platforms, the WHO declared an end to the global health emergency of COVID-19 on May 5, 2023. Current COVID-19 vaccines encompass inactivated, live attenuated, viral vector, protein subunit, nucleic acid (DNA and RNA), and virus-like particle (VLP) vaccines. However, the efficacy of these vaccines is diminishing due to the constant mutation of SARS-CoV-2 and the heightened immune evasion abilities of emerging variants. This review examines the impact of the COVID-19 pandemic, the biological characteristics of the virus, and its diverse variants. Moreover, the review underscores the effectiveness, advantages, and disadvantages of authorized COVID-19 vaccines. Additionally, it analyzes the challenges, strategies, and future prospects of developing a safe, broad-spectrum vaccine that confers sufficient and sustainable immune protection against new variants of SARS-CoV-2. These discussions not only offer insight for the development of next-generation COVID-19 vaccines but also summarize experiences for combating future emerging viruses.
Collapse
Affiliation(s)
- Congrui Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture, Guangdong Provincial Key Laboratory of Animal Nutrition Control, State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510000, China
| | - Shengmei Pang
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jiaqi Liu
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Qiangde Duan
- Department of Veterinary Microbiology, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
- Jiangsu Joint Laboratory for International Cooperation in Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
8
|
Gardner BJ, Kilpatrick AM. Predicting Vaccine Effectiveness for Hospitalization and Symptomatic Disease for Novel SARS-CoV-2 Variants Using Neutralizing Antibody Titers. Viruses 2024; 16:479. [PMID: 38543844 PMCID: PMC10975673 DOI: 10.3390/v16030479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 05/23/2024] Open
Abstract
The emergence of new virus variants, including the Omicron variant (B.1.1.529) of SARS-CoV-2, can lead to reduced vaccine effectiveness (VE) and the need for new vaccines or vaccine doses if the extent of immune evasion is severe. Neutralizing antibody titers have been shown to be a correlate of protection for SARS-CoV-2 and other pathogens, and could be used to quickly estimate vaccine effectiveness for new variants. However, no model currently exists to provide precise VE estimates for a new variant against severe disease for SARS-CoV-2 using robust datasets from several populations. We developed predictive models for VE against COVID-19 symptomatic disease and hospitalization across a 54-fold range of mean neutralizing antibody titers. For two mRNA vaccines (mRNA-1273, BNT162b2), models fit without Omicron data predicted that infection with the BA.1 Omicron variant increased the risk of hospitalization 2.8-4.4-fold and increased the risk of symptomatic disease 1.7-4.2-fold compared to the Delta variant. Out-of-sample validation showed that model predictions were accurate; all predictions were within 10% of observed VE estimates and fell within the model prediction intervals. Predictive models using neutralizing antibody titers can provide rapid VE estimates, which can inform vaccine booster timing, vaccine design, and vaccine selection for new virus variants.
Collapse
Affiliation(s)
- Billy J. Gardner
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| | - A. Marm Kilpatrick
- Department of Ecology and Evolutionary Biology, University of California, Santa Cruz, CA 95060, USA
| |
Collapse
|
9
|
Kemerley A, Gupta A, Thirunavukkarasu M, Maloney M, Burgwardt S, Maulik N. COVID-19 Associated Cardiovascular Disease-Risks, Prevention and Management: Heart at Risk Due to COVID-19. Curr Issues Mol Biol 2024; 46:1904-1920. [PMID: 38534740 PMCID: PMC10969474 DOI: 10.3390/cimb46030124] [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: 11/28/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 03/28/2024] Open
Abstract
The SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus-2) virus and the resulting COVID-19 pandemic have had devastating and lasting impact on the global population. Although the main target of the disease is the respiratory tract, clinical outcomes, and research have also shown significant effects of infection on other organ systems. Of interest in this review is the effect of the virus on the cardiovascular system. Complications, including hyperinflammatory syndrome, myocarditis, and cardiac failure, have been documented in the context of COVID-19 infection. These complications ultimately contribute to worse patient outcomes, especially in patients with pre-existing conditions such as hypertension, diabetes, or cardiovascular disease (CVD). Importantly and interestingly, reports have demonstrated that COVID-19 also causes myocardial injury in adults without pre-existing conditions and contributes to systemic complications in pediatric populations, such as the development of multisystem inflammatory syndrome in children (MIS-C). Although there is still a debate over the exact mechanisms by which such complications arise, understanding the potential paths by which the virus can influence the cardiovascular system to create an inflammatory environment may clarify how SARS-CoV-2 interacts with human physiology. In addition to describing the mechanisms of disease propagation and patient presentation, this review discusses the diagnostic findings and treatment strategies and the evolution of management for patients presenting with cardiovascular complications, focusing on disease treatment and prevention.
Collapse
Affiliation(s)
| | | | | | | | | | - Nilanjana Maulik
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut School of Medicine, UConn Health, 263 Farmington Avenue, Farmington, CT 06030, USA; (A.K.); (A.G.); (M.T.); (S.B.)
| |
Collapse
|
10
|
McMahan K, Wegmann F, Aid M, Sciacca M, Liu J, Hachmann NP, Miller J, Jacob-Dolan C, Powers O, Hope D, Wu C, Pereira J, Murdza T, Mazurek CR, Hoyt A, Boon ACM, Davis-Gardner M, Suthar MS, Martinot AJ, Boursiquot M, Cook A, Pessaint L, Lewis MG, Andersen H, Tolboom J, Serroyen J, Solforosi L, Costes LMM, Zahn RC, Barouch DH. Mucosal boosting enhances vaccine protection against SARS-CoV-2 in macaques. Nature 2024; 626:385-391. [PMID: 38096903 PMCID: PMC10849944 DOI: 10.1038/s41586-023-06951-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 12/07/2023] [Indexed: 01/26/2024]
Abstract
A limitation of current SARS-CoV-2 vaccines is that they provide minimal protection against infection with current Omicron subvariants1,2, although they still provide protection against severe disease. Enhanced mucosal immunity may be required to block infection and onward transmission. Intranasal administration of current vaccines has proven inconsistent3-7, suggesting that alternative immunization strategies may be required. Here we show that intratracheal boosting with a bivalent Ad26-based SARS-CoV-2 vaccine results in substantial induction of mucosal humoral and cellular immunity and near-complete protection against SARS-CoV-2 BQ.1.1 challenge. A total of 40 previously immunized rhesus macaques were boosted with a bivalent Ad26 vaccine by the intramuscular, intranasal and intratracheal routes, or with a bivalent mRNA vaccine by the intranasal route. Ad26 boosting by the intratracheal route led to a substantial expansion of mucosal neutralizing antibodies, IgG and IgA binding antibodies, and CD8+ and CD4+ T cell responses, which exceeded those induced by Ad26 boosting by the intramuscular and intranasal routes. Intratracheal Ad26 boosting also led to robust upregulation of cytokine, natural killer, and T and B cell pathways in the lungs. After challenge with a high dose of SARS-CoV-2 BQ.1.1, intratracheal Ad26 boosting provided near-complete protection, whereas the other boosting strategies proved less effective. Protective efficacy correlated best with mucosal humoral and cellular immune responses. These data demonstrate that these immunization strategies induce robust mucosal immunity, suggesting the feasibility of developing vaccines that block respiratory viral infections.
Collapse
Affiliation(s)
- Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Frank Wegmann
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Michaela Sciacca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Nicole P Hachmann
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jessica Miller
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Olivia Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - David Hope
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Cindy Wu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Juliana Pereira
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tetyana Murdza
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Camille R Mazurek
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Amelia Hoyt
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | | | - Amanda J Martinot
- Tufts University Cummings School of Veterinary Medicine, Grafton, MA, USA
| | | | | | | | | | | | | | - Jan Serroyen
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | | | | | - Roland C Zahn
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
11
|
Lundstrom K. COVID-19 Vaccines: Where Did We Stand at the End of 2023? Viruses 2024; 16:203. [PMID: 38399979 PMCID: PMC10893040 DOI: 10.3390/v16020203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/23/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Vaccine development against SARS-CoV-2 has been highly successful in slowing down the COVID-19 pandemic. A wide spectrum of approaches including vaccines based on whole viruses, protein subunits and peptides, viral vectors, and nucleic acids has been developed in parallel. For all types of COVID-19 vaccines, good safety and efficacy have been obtained in both preclinical animal studies and in clinical trials in humans. Moreover, emergency use authorization has been granted for the major types of COVID-19 vaccines. Although high safety has been demonstrated, rare cases of severe adverse events have been detected after global mass vaccinations. Emerging SARS-CoV-2 variants possessing enhanced infectivity have affected vaccine protection efficacy requiring re-design and re-engineering of novel COVID-19 vaccine candidates. Furthermore, insight is given into preparedness against emerging SARS-CoV-2 variants.
Collapse
|
12
|
Mehta D, Sun T, Wang J, Situ A, Park Y. Comparison of healthcare resource use and cost between influenza and COVID-19 vaccine coadministration and influenza vaccination only. J Med Econ 2024; 27:1190-1196. [PMID: 39231068 DOI: 10.1080/13696998.2024.2400852] [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: 07/31/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To compare healthcare resource utilization (HCRU) and all-cause medical costs among individuals aged ≥50 years who received influenza and COVID-19 vaccines on the same day and those who received influenza vaccine only. METHODS We conducted a retrospective cohort study leveraging Optum's de-identified Clinformatics DataMart from 8/31/2021 to 7/31/2023. Individuals aged ≥50 years continuously enrolled in health plans for 1 year prior and until 7/31/2023 were included. Two cohorts were formed based on vaccination status between 8/31/2022 and 1/31/2023: co-administered influenza and COVID-19 vaccines (co-admin cohort) and influenza vaccine only (influenza cohort). Associations between vaccination status and all-cause, influenza-related, COVID-related, pneumonia-related, and cardiorespiratory-related hospitalization, outpatient or emergency room visits and all-cause medical costs were estimated by weighted generalized linear models, adjusting for confounding by stabilized inverse probability of treatment weighting. RESULTS 613,156 (mean age: 71) and 1,340,011 (mean age: 72) individuals were included in the co-admin and influenza cohorts, respectively. After weighting, the baseline characteristics were balanced between cohorts. The co-admin cohort was at statistically significant lower risk of all-cause (RR: 0.95, 95% CI: 0.93-0.96), COVID-19-related (RR: 0.59, 95% CI: 0.56-0.63), cardiorespiratory-related (RR: 0.94, 95% CI: 0.93-0.96) and pneumonia-related (RR: 0.86, 95% CI: 0.83-0.90) hospitalization but not influenza-related hospitalizations (RR: 0.91, 95% CI: 0.81, 1.04) compared with the influenza cohort. Co-administration was associated with 3% lower all-cause medical cost (cost ratio: 0.974, 95% CI: 0.968, 0.979) during the follow-up period compared to receiving influenza vaccine only. LIMITATIONS Limitations include the potential residual confounding bias in observational data, measurement errors from claims data, and that the cohort was followed for a single season. CONCLUSION Receiving co-administered COVID-19 and influenza vaccines versus only receiving influenza vaccination reduced the risk of HCRU, especially COVID-19-related hospitalization and all-cause medical costs. Increasing vaccine coverage, particularly for COVID-19, might have public health and economic benefits.
Collapse
Affiliation(s)
- Darshan Mehta
- Health Economics and Outcomes Research, Moderna, Inc, Cambridge, MA, USA
| | - Tianyu Sun
- Real-World Evidence Analytics, Moderna, Inc, Cambridge, MA, USA
| | - Jane Wang
- Real-World Evidence Analytics, Moderna, Inc, Cambridge, MA, USA
| | - Aaron Situ
- Real-World Evidence Analytics, Moderna, Inc, Cambridge, MA, USA
| | - Yoonyoung Park
- Infectious Disease, Epidemiology, Moderna, Inc, Cambridge, MA, USA
| |
Collapse
|
13
|
Youssef A, Ulloa L. Ethical and Legal Debates on Vaccine Infodemics. Cureus 2024; 16:e52566. [PMID: 38371094 PMCID: PMC10874613 DOI: 10.7759/cureus.52566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2024] [Indexed: 02/20/2024] Open
Abstract
Over the course of three and a half years, the global toll of coronavirus disease 2019 (COVID-19) has claimed the lives of millions of individuals. Scientific breakthroughs, exemplified by mRNA vaccines, have emerged as crucial tools in saving numerous lives and fortifying our defenses against future pandemics. However, the battle against the virus has been complicated by the dissemination of misleading political and ethical information, resulting in avoidable fatalities. Recognizing this phenomenon, the term 'infodemics' has been coined to denote the proliferation of false or misleading information that hinders effective social responses. Given the historical prevalence of infodemics surrounding vaccinations, this discussion delves into the ongoing ethical and legal deliberations concerning vaccination mandates, an indispensable health intervention in the face of pandemics. Governments bear the responsibility of safeguarding their citizens, acknowledging the social requirements imposed by the collective well-being. The protection of both citizens and healthcare workers becomes paramount, considering the potential risks of infection and mortality associated with individuals refusing vaccination. Historically, governments have played a pivotal role in eradicating pandemics through the implementation of vaccine mandates. However, the contemporary landscape is marked by the infusion of political and misleading misinformation, presenting new challenges. Governments are now confronted with an ethical duty to ensure that citizens possess the necessary information to make informed decisions and safeguard their well-being. While grappling with the realization that extraordinary circumstances demand extraordinary responses, the lessons from past pandemics underscore the imperative of prioritizing public health, especially in the context of the high numbers of casualties worldwide. This discourse explores the ethical and legal dimensions surrounding vaccine mandates, with particular emphasis on their relevance to healthcare workers.
Collapse
Affiliation(s)
- Ayman Youssef
- Anesthesiology and Perioperative Medicine, Duke University Medical Center, Durham, USA
| | - Luis Ulloa
- Anesthesiology and Perioperative Medicine, Duke University Medical Center, Durham, USA
| |
Collapse
|
14
|
Islas-Vazquez L, Alvarado-Alvarado YC, Cruz-Aguilar M, Velazquez-Soto H, Villalobos-Gonzalez E, Ornelas-Hall G, Perez-Tapia SM, Jimenez-Martinez MC. Evaluation of the Abdala Vaccine: Antibody and Cellular Response to the RBD Domain of SARS-CoV-2. Vaccines (Basel) 2023; 11:1787. [PMID: 38140191 PMCID: PMC10748004 DOI: 10.3390/vaccines11121787] [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: 10/17/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/24/2023] Open
Abstract
Abdala is a recently released RBD protein subunit vaccine against SARS-CoV-2. A few countries, including Mexico, have adopted Abdala as a booster dose in their COVID-19 vaccination schemes. Despite that, most of the Mexican population has received full-scheme vaccination with platforms other than Abdala; little is known regarding Abdala's immunological features, such as its antibody production and T- and B-cell-specific response induction. This work aimed to study antibody production and the adaptive cellular response in the Mexican population that received the Abdala vaccine as a booster. We recruited 25 volunteers and evaluated their RBD-specific antibody production, T- and B-cell-activating profiles, and cytokine production. Our results showed that the Abdala vaccine increases the concentration of RBD IgG-specific antibodies. Regarding the cellular response, after challenging peripheral blood cultures with RBD, the plasmablast (CD19+CD27+CD38High) and transitional B-cell (CD19+CD21+CD38High) percentages increased significantly, while T cells showed an increased activated phenotype (CD3+CD4+CD25+CD69+ and CD3+CD4+CD25+HLA-DR+). Also, IL-2 and IFN-γ increased significantly in the supernatant of the RBD-stimulated cells. Our results suggest that Abdala vaccination, used as a booster, evokes antibody production and the activation of previously generated memory against the SARS-CoV-2 RBD domain.
Collapse
Affiliation(s)
- Lorenzo Islas-Vazquez
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Yan Carlos Alvarado-Alvarado
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Marisa Cruz-Aguilar
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Henry Velazquez-Soto
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
| | - Eduardo Villalobos-Gonzalez
- Unidad de Vigilancia Epidemiológica Hospitalaria, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico
| | - Gloria Ornelas-Hall
- Unidad de Vigilancia Epidemiológica Hospitalaria, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico
| | - Sonia Mayra Perez-Tapia
- Unidad de Desarrollo e Investigación en Bioterapéuticos (UDIBI), Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Mexico
- Laboratorio Nacional para Servicios Especializados de Investigación, Desarrollo e Innovación (I+D+i) para Farmoquímicos y Biotecnológicos, LANSEIDI-FarBiotec-CONACyT, Mexico City 11340, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (ENCB-IPN), Mexico City 11340, Mexico
| | - Maria C. Jimenez-Martinez
- Department of Immunology and Research Unit, Institute of Ophthalmology “Conde de Valenciana Foundation”, Mexico City 06800, Mexico; (L.I.-V.)
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City 04510, Mexico
| |
Collapse
|
15
|
Jain P, Basnet S, Syed S, Arakaki B, Mues KE, Marcum ZA, Diaz-Decaro J. Testing for Cytomegalovirus Among Individuals Who Were Immunocompromised, 2018-2022. JAMA Netw Open 2023; 6:e2345126. [PMID: 38010654 PMCID: PMC10682830 DOI: 10.1001/jamanetworkopen.2023.45126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/14/2023] [Indexed: 11/29/2023] Open
Abstract
This cohort study investigates rates of cytomegalovirus testing before and during the COVID-19 pandemic among individuals who were immunocompromised.
Collapse
|
16
|
Buzas D, Bunzel AH, Staufer O, Milodowski EJ, Edmunds GL, Bufton JC, Vidana Mateo BV, Yadav SKN, Gupta K, Fletcher C, Williamson MK, Harrison A, Borucu U, Capin J, Francis O, Balchin G, Hall S, Vega MV, Durbesson F, Lingappa S, Vincentelli R, Roe J, Wooldridge L, Burt R, Anderson RJL, Mulholland AJ, Bristol UNCOVER Group, Hare J, Bailey M, Davidson AD, Finn A, Morgan D, Mann J, Spatz J, Garzoni F, Schaffitzel C, Berger I. In vitro generated antibodies guide thermostable ADDomer nanoparticle design for nasal vaccination and passive immunization against SARS-CoV-2. Antib Ther 2023; 6:277-297. [PMID: 38075238 PMCID: PMC10702856 DOI: 10.1093/abt/tbad024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/12/2023] [Accepted: 10/14/2023] [Indexed: 01/10/2024] Open
Abstract
Background Due to COVID-19, pandemic preparedness emerges as a key imperative, necessitating new approaches to accelerate development of reagents against infectious pathogens. Methods Here, we developed an integrated approach combining synthetic, computational and structural methods with in vitro antibody selection and in vivo immunization to design, produce and validate nature-inspired nanoparticle-based reagents against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Results Our approach resulted in two innovations: (i) a thermostable nasal vaccine called ADDoCoV, displaying multiple copies of a SARS-CoV-2 receptor binding motif derived epitope and (ii) a multivalent nanoparticle superbinder, called Gigabody, against SARS-CoV-2 including immune-evasive variants of concern (VOCs). In vitro generated neutralizing nanobodies and electron cryo-microscopy established authenticity and accessibility of epitopes displayed by ADDoCoV. Gigabody comprising multimerized nanobodies prevented SARS-CoV-2 virion attachment with picomolar EC50. Vaccinating mice resulted in antibodies cross-reacting with VOCs including Delta and Omicron. Conclusion Our study elucidates Adenovirus-derived dodecamer (ADDomer)-based nanoparticles for use in active and passive immunization and provides a blueprint for crafting reagents to combat respiratory viral infections.
Collapse
Affiliation(s)
- Dora Buzas
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Adrian H Bunzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Oskar Staufer
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Leibniz Institute for New Materials, Helmholtz Institute for Pharmaceutical Research and Center for Biophysics, Saarland University, Saarbrücken 66123, Germany
| | | | - Grace L Edmunds
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joshua C Bufton
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | | | | | - Kapil Gupta
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | | | - Maia K Williamson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | - Ufuk Borucu
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Julien Capin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Ore Francis
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Georgia Balchin
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Sophie Hall
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Mirella V Vega
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
| | - Fabien Durbesson
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | | | - Renaud Vincentelli
- Architecture et Fonction des Macromolécules Biologiques, UMR 7257, CNRS, Aix-Marseille Université, Marseille, France
| | - Joe Roe
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Linda Wooldridge
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Rachel Burt
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | | | | | | | - Jonathan Hare
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Mick Bailey
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Andrew D Davidson
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Adam Finn
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
- Children's Vaccine Centre, Bristol Medical School, Bristol BS2 8EF UK
| | - David Morgan
- Imophoron Ltd, Science Creates Old Market, Midland Rd, Bristol BS2 0JZ UK
| | - Jamie Mann
- Bristol Veterinary School, University of Bristol, Bristol BS40 5DU UK
| | - Joachim Spatz
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Frederic Garzoni
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Christiane Schaffitzel
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| | - Imre Berger
- Max Planck Bristol Centre for Minimal Biology, University of Bristol, Bristol BS8 1TS, UK
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK
- School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
- Bristol University COVID-19 Emergency Research Group, Bristol BS8 1TH, UK
| |
Collapse
|
17
|
Prajapati G, Das A, Sun Y, Fonseca E. Hospitalization Among Patients Treated With Molnupiravir: A Retrospective Study of Administrative Data. Clin Ther 2023; 45:957-964. [PMID: 37598055 DOI: 10.1016/j.clinthera.2023.07.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/21/2023]
Abstract
PURPOSE Molnupiravir is an oral antiviral agent authorized for emergency use to treat mild to moderate cases of coronavirus disease 2019 (COVID-19) in adults at high risk for progression to severe clinical outcomes. This study aimed to describe patient characteristics and health outcomes in a cohort of adult patients treated with molnupiravir in an outpatient setting in the United States. METHODS This was a retrospective cohort study of adults identified in the HealthVerity database with a pharmacy claim for molnupiravir between December 24, 2021, and April 14, 2022. Hospitalization and health care use were assessed over the 28 days after the molnupiravir pharmacy claim. FINDINGS Among 26,554 patients identified, 71.1% were aged ≥50 years and 58.9% were female. A total of 8794 patients (33.1%) had received at least 1 dose of the COVID-19 vaccine. The most prevalent risk factors for severe COVID-19 identified were hypertension (45.1%), steroid and/or immunosuppressant use (39.6%), and being obese or overweight (24.6%), with 79.1% of patients having ≥1 risk factor. The majority (61.0%) of patients were prescribed comedications contraindicated with or had the potential for major drug-drug interactions with ritonavir-containing regimens. Within 28 days after initiating molnupiravir, 3.3% of patients were hospitalized for any cause and 1.7% for COVID-19-related reasons. Among all hospitalized patients, 9.2% were admitted to an intensive care unit, 3.9% received oxygen, and 3.8% required mechanical ventilation. IMPLICATIONS The majority of patients treated with molnupiravir in early 2022 had at least one risk factor for severe COVID-19 and had comedications that could require treatment modification or monitoring if given a ritonavir-containing regimen. Hospitalization was uncommon after treatment with molnupiravir, with COVID-19-related inpatient admission in <2% of patients. Among those hospitalized, patient use of intensive care and oxygen-based resources was infrequent. The study design, however, does not permit any conclusions regarding the effectiveness of molnupiravir.
Collapse
Affiliation(s)
| | - Amar Das
- Merck & Co, Inc, Rahway, New Jersey, USA
| | - Yezhou Sun
- Merck & Co, Inc, Rahway, New Jersey, USA
| | | |
Collapse
|
18
|
Jing X, Han M, Wang X, Zhou L. SARS-CoV-2 vaccine breakthrough infection in the older adults: a meta-analysis and systematic review. BMC Infect Dis 2023; 23:577. [PMID: 37667195 PMCID: PMC10478381 DOI: 10.1186/s12879-023-08553-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Corona Virus Disease 2019 (COVID-19) mRNA vaccine effectiveness (VE) has recently declined, and reports about COVID-19 breakthrough infection have increased. We aimed to conduct a meta-analysis on population-based studies of the prevalence and incidence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infection amongst older adults worldwide. METHODS Studies from PubMed, Embase, Cochrane Library, and Web of Science were systematically screened to determine the prevalence and incidence of SARS-CoV-2 breakthrough infection in older adults from inception to November 2, 2022. Our meta-analysis included 30 studies, all published in English. Pooled estimates were calculated using a random-effect model through the inverse variance method. Publication bias was tested through funnel plots and Egger's regression test, and sensitivity analyses were performed to confirm the robustness of the results. This research was performed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. RESULTS Thirty publications were included in this meta-analysis (17 on prevalence, 17 on incidence, and 4 on both). The pooled prevalence of COVID-19 breakthrough infection among older adults was 7.7 per 1,000 persons (95% confidence interval [95%CI] 4.0-15.0). At the same time, the pooled incidence was 29.1 per 1000 person-years (95%CI 15.2-55.7). CONCLUSIONS This meta-analysis provides estimates of prevalence and incidence in older adults. We concluded that the prevalence and incidence of SARS-CoV-19 breakthrough infection in older people was low. The prevalence and incidence of breakthrough infection admitted to hospital, severe-critical, and deathly was significantly lower. Otherwise, there was considerable heterogeneity among estimates in this study, which should be considered when interpreting the results.
Collapse
Affiliation(s)
- Xiaohui Jing
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Tianjin, 301617, P.R. China.
| | - Menglin Han
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Tianjin, 301617, P.R. China
| | - Xiaoxuan Wang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Tianjin, 301617, P.R. China
| | - Li Zhou
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, Tianjin, 301617, P.R. China
| |
Collapse
|
19
|
Feng JL, Wang WJ, Jin PF, Zheng H, Jin LR, Xia X, Zhang XY, Li ZP, Li JX, Zhu FC. Comparison of antibody persistency through one year between one-dose and two-dose regimens of Ad5-nCoV vaccine for COVID-19. Hum Vaccin Immunother 2023; 19:2230760. [PMID: 37428653 DOI: 10.1080/21645515.2023.2230760] [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: 01/31/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 07/12/2023] Open
Abstract
This post-hoc analysis compared the receptor-binding domain (RBD)-specific and pseudovirus neutralizing antibodies against the wild-type SARS-CoV-2 strain elicited by one or two doses (56-d interval) of Ad5-nCoV vaccine regimen (NCT04341389 and NCT04566770). Both trials had low-dose and high-dose groups. Propensity score matching was used to adjust the baseline between one- and two-dose regimens. To predict the decrease in antibody titers 1 y after vaccination, half-lives of RBD-binding antibodies and pseudovirus neutralizing antibodies were computed. We obtained 34 and 29 pairs of participants in the low- and high-dose groups based on the propensity score matching. The two-dose regimen of Ad5-nCoV increased the peaking level of neutralizing antibodies compared to the one-dose regimen at day 28, but the responses of the neutralizing antibodies were not consistent with those of the RBD antibodies. Half-lives of the RBD-binding antibodies in the two-dose Ad5-nCoV regimen (202-209 days) were longer than those in the one-dose regimen (136-137 d); half-lives of the pseudovirus neutralizing antibody in the one-dose Ad5-nCoV regimen (177 d) were longer than those in the two-dose regimen (116-131 d). The predicted positive rates of RBD-binding antibodies in the one-dose regimen (34.1%-38.3%) would be lower than those in the two-dose Ad5-nCoV regimen (67.0%-84.0%), while the positive rates of pseudovirus neutralizing antibodies in the one-dose regimen (65.4%-66.7%) would be higher than those in the two-dose regimen (48.3%-58.0%). The two-dose Ad5-nCoV regimen with a 56-d interval had no effect on the persistence of neutralizing antibodies but slowed decay trend of RBD-binding antibodies.
Collapse
Affiliation(s)
- Jia-Lu Feng
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R China
| | - Wen-Juan Wang
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R China
| | - Peng-Fei Jin
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R China
| | - Hui Zheng
- School of Public Health, Southeast University, Nanjing, P.R China
| | - Lai-Run Jin
- School of Public Health, Southeast University, Nanjing, P.R China
| | - Xin Xia
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R China
| | - Xiao-Yin Zhang
- School of Public Health, Southeast University, Nanjing, P.R China
| | - Zhuo-Pei Li
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R China
| | - Jing-Xin Li
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R China
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R China
- School of Public Health, Southeast University, Nanjing, P.R China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P.R China
| | - Feng-Cai Zhu
- School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, P.R China
- National Health Commission (NHC) Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing, Jiangsu, P.R China
- School of Public Health, Southeast University, Nanjing, P.R China
- Institute of Global Public Health and Emergency Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, P.R China
| |
Collapse
|
20
|
Miteva D, Kitanova M, Batselova H, Lazova S, Chervenkov L, Peshevska-Sekulovska M, Sekulovski M, Gulinac M, Vasilev GV, Tomov L, Velikova T. The End or a New Era of Development of SARS-CoV-2 Virus: Genetic Variants Responsible for Severe COVID-19 and Clinical Efficacy of the Most Commonly Used Vaccines in Clinical Practice. Vaccines (Basel) 2023; 11:1181. [PMID: 37514997 PMCID: PMC10385722 DOI: 10.3390/vaccines11071181] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/22/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
Although the chief of the World Health Organization (WHO) has declared the end of the coronavirus disease 2019 (COVID-19) as a global health emergency, the disease is still a global threat. To be able to manage such pandemics in the future, it is necessary to develop proper strategies and opportunities to protect human life. The data on the SARS-CoV-2 virus must be continuously analyzed, and the possibilities of mutation and the emergence of new, more infectious variants must be anticipated, as well as the options of using different preventive and therapeutic techniques. This is because the fast development of severe acute coronavirus 2 syndrome (SARS-CoV-2) variants of concern have posed a significant problem for COVID-19 pandemic control using the presently available vaccinations. This review summarizes data on the SARS-CoV-2 variants that are responsible for severe COVID-19 and the clinical efficacy of the most commonly used vaccines in clinical practice. The consequences after the disease (long COVID or post-COVID conditions) continue to be the subject of studies and research, and affect social and economic life worldwide.
Collapse
Affiliation(s)
- Dimitrina Miteva
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov str., 1164 Sofia, Bulgaria; (D.M.); (M.K.)
| | - Meglena Kitanova
- Department of Genetics, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 8 Dragan Tzankov str., 1164 Sofia, Bulgaria; (D.M.); (M.K.)
| | - Hristiana Batselova
- Department of Epidemiology and Disaster Medicine, University Hospital “Saint George”, Medical University, 6000 Plovdiv, Bulgaria;
| | - Snezhina Lazova
- Pediatric Department, University Hospital “N. I. Pirogov,” 21 “General Eduard I. Totleben” Blvd, 1606 Sofia, Bulgaria;
- Department of Healthcare, Faculty of Public Health “Prof. Tsekomir Vodenicharov, MD, DSc”, Medical University of Sofia, Bialo More 8 str., 1527 Sofia, Bulgaria
| | - Lyubomir Chervenkov
- Department of Diagnostic Imaging, Medical University Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria;
| | - Monika Peshevska-Sekulovska
- Department of Gastroenterology, University Hospital Lozenetz, 1407 Sofia, Bulgaria;
- Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria;
| | - Metodija Sekulovski
- Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria;
- Department of Anesthesiology and Intensive Care, University Hospital Lozenetz, 1 Kozyak str., 1407 Sofia, Bulgaria
| | - Milena Gulinac
- Department of General and Clinical Pathology, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria;
| | - Georgi V. Vasilev
- Clinic of Endocrinology and Metabolic Disorders, UMHAT “Sv. Georgi”, 4000 Plovdiv, Bulgaria;
| | - Luchesar Tomov
- Department of Informatics, New Bulgarian University, Montevideo 21 str., 1618 Sofia, Bulgaria;
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, 1407 Sofia, Bulgaria;
| |
Collapse
|
21
|
Ikeokwu AE, Adeniran OF, Marwizi FM, Kolade-Ernest OJ, Solomon RO, Ogedengbe W, Onyemarin-Henry P, Okpo NC, Onyinye O. A Meta-Analysis To Ascertain the Effectiveness of COVID-19 Vaccines on Clinical Outcomes in Patients With COVID-19 Infection in North America. Cureus 2023; 15:e41053. [PMID: 37519527 PMCID: PMC10374409 DOI: 10.7759/cureus.41053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
The challenges in controlling the pandemic have been exacerbated by the disease's morbidity and the emergence of additional COVID-19 variants. The use of emergency vaccines to circumvent these challenges has sparked mixed opinions on their effectiveness. Therefore, we conducted a meta-analysis to assess the efficacy of COVID-19 vaccines on clinical outcomes such as incidence, hospitalization, and ventilation rates in both vaccinated and unvaccinated patients. PubMed, Google Scholar, and Cochrane Central Register of Clinical Trials were searched on April 21, 2022, to extract published articles comparing vaccinated COVID-19 patients versus unvaccinated COVID-19 patients and their clinical outcomes. The clinical outcomes studied were incidence rate, intensive care unit (ICU) admission, mechanical ventilation, and hospitalization rates. The analysis was performed with Review Manager (RevMan) software. Random-effect models were used to calculate pooled odds ratio and corresponding 95% confidence interval (CI). In our meta-analysis, we have identified a total of 250 published findings, encompassing 15 studies that involved a cumulative count of 24,164,227 individuals diagnosed with COVID-19. Being unvaccinated had a significant association with severe clinical outcomes in patients infected with COVID-19. Unvaccinated individuals were 2.36 times more likely to be infected, with a 95% CI ranging from 1.13 to 4.94 (p = 0.02). Unvaccinated subjects with COVID-19 infection were 6.93 times more likely to be admitted to the ICU than their vaccinated counterparts, with a 95% CI ranging from 3.57 to 13.46 (p < 0.0001). The hospitalization rate was 3.37 higher among the unvaccinated compared to those vaccinated, with a 95% CI ranging from 1.92 to 5.93 (p < 0.0001). In addition, patients with COVID-19 infection who are unvaccinated were 6.44 times more likely to be mechanically ventilated than those vaccinated, with a 95% CI ranging from 3.13 to 13.23 (p < 0.0001). Overall, our study revealed that vaccination against COVID-19 disease is beneficial and effective in mitigating the spread of the infection and associated clinical outcomes. However, more awareness and proper education must be made to increase vaccine acceptance. We, therefore, recommend and urge all stakeholders involved in COVID-19 prevention, management, and control to strengthen awareness and educate the people on the effectiveness of COVID-19 vaccination.
Collapse
Affiliation(s)
| | | | - Farirai M Marwizi
- General Medicine, Universitatea de Medicină şi Farmacie, Timisoara, ROU
| | - Oreoluwa J Kolade-Ernest
- Pediatrics, SUNY (State University of New York) Downstate Health Sciences University, New York City, USA
| | - Rebecca O Solomon
- Community Medicine, Lagos State Primary Health Care Board, Lagos, NGA
| | - William Ogedengbe
- Medicine and Surgery, Lagos University Teaching Hospital (LUTH), Lagos, NGA
| | | | | | - Okam Onyinye
- Internal Medicine, Alimosho General Hospital, Lagos, NGA
| |
Collapse
|
22
|
Igho-Osagie E, Puenpatom A, Williams MG, Song Y, Yi D, Wang J, Berman R, Gu M, He C. Prevalence of potential drug-drug interactions with ritonavir-containing COVID-19 therapy. J Manag Care Spec Pharm 2023; 29:509-518. [PMID: 36989455 PMCID: PMC10394216 DOI: 10.18553/jmcp.2023.22366] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
BACKGROUND: Patients with COVID-19 receiving ritonavir-containing therapies are at risk of potential drug-drug interactions (pDDIs) because of ritonavir's effects on cytochrome P450 3A4. OBJECTIVE: To assess the prevalence of pDDIs with ritonavir-containing COVID-19 therapy in adults with COVID-19 using the Optum Clinformatics Data Mart database. METHODS: In this retrospective, observational cohort study, patients with COVID-19 aged 18 years or older prescribed cytochrome P450 3A4-mediated medications with supply days overlapping the date of COVID-19 diagnosis between January 1, 2020, and June 30, 2021, were classified as having pDDIs. pDDI was classified as contraindicated, major, moderate, or mild using established drug interaction resources. Prevalence of pDDIs with ritonavir-containing COVID-19 therapy was estimated for the entire cohort and in patient groups with high risk of severe COVID-19 progression or pDDIs. Actual COVID-19 treatments received by the patients, if any, were not considered. Outcomes were presented descriptively without adjusted comparisons. RESULTS: A total of 718,387 patients with COVID-19 were identified. The age-sex standardized national prevalence of pDDIs of any severity was estimated at 52.2%. Approximately 34.5% were at risk of contraindicated or major pDDIs. Compared with patients without pDDI, patients exposed to pDDIs were older and more likely to be female, reside in long-term care facilities, and have risk factors for progression to severe COVID-19. Higher prevalence of major/contraindicated pDDIs was observed in older patients (76.1%), female patients (65.0%), and patients with multiple morbidities (84.6%). CONCLUSIONS: Study findings demonstrate that more than one-third of patients with COVID-19 were at risk of significant pDDIs if treated with ritonavir-containing COVID-19 therapy and highlight the need to assess all patients with COVID-19 for pDDIs. Ritonavir-based therapies may not be appropriate for certain patient groups, and alternative therapies should be considered. DISCLOSURES: Drs Igho-Osagie, Puenpatom, and Grifasi Williams are employees of Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc. Dr Song and Ms He are employees of Analysis Group, Inc., and served as paid consultants for Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc. Drs Yi and Wang, and Mr Berman, and Ms Gu were employees of Analysis Group, Inc., at the time of study conduct. Financial support for this study was provided by Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc. The study sponsor was involved in the design and conduct of the study; collection, management, analysis, interpretation of data; preparation, review, or approval of the manuscript; and decision to submit the manuscript for publication.
Collapse
Affiliation(s)
| | | | | | - Yan Song
- Analysis Group, Inc., Boston, MA
| | | | | | | | - Miley Gu
- Analysis Group, Inc., Boston, MA
| | | |
Collapse
|
23
|
Swart M, van der Lubbe J, Schmit-Tillemans S, van Huizen E, Verspuij J, Gil AI, Choi Y, Daal C, Perkasa A, de Wilde A, Claassen E, de Jong R, Wiese KE, Cornelissen L, van Es M, van Heerden M, Kourkouta E, Tahiri I, Mulders M, Vreugdenhil J, Feddes-de Boer K, Muchene L, Tolboom J, Dekking L, Juraszek J, Vellinga J, Custers J, Bos R, Schuitemaker H, Wegmann F, Roozendaal R, Kuipers H, Zahn R. Booster vaccination with Ad26.COV2.S or an Omicron-adapted vaccine in pre-immune hamsters protects against Omicron BA.2. NPJ Vaccines 2023; 8:40. [PMID: 36927774 PMCID: PMC10018069 DOI: 10.1038/s41541-023-00633-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
Since the original outbreak of the SARS-CoV-2 virus, several rapidly spreading SARS-CoV-2 variants of concern (VOC) have emerged. Here, we show that a single dose of Ad26.COV2.S (based on the Wuhan-Hu-1 spike variant) protects against the Gamma and Delta variants in naive hamsters, supporting the observed maintained vaccine efficacy in humans against these VOC. Adapted spike-based booster vaccines targeting Omicron variants have now been authorized in the absence of human efficacy data. We evaluated the immunogenicity and efficacy of Ad26.COV2.S.529 (encoding a stabilized Omicron BA.1 spike) in naive mice and in hamsters with pre-existing immunity to the Wuhan-Hu-1 spike. In naive mice, Ad26.COV2.S.529 elicited higher neutralizing antibody titers against SARS-CoV-2 Omicron BA.1 and BA.2, compared with Ad26.COV2.S. However, neutralizing titers against the SARS-CoV-2 B.1 (D614G) and Delta variants were lower after primary vaccination with Ad26.COV2.S.529 compared with Ad26.COV2.S. In contrast, we found comparable Omicron BA.1 and BA.2 neutralizing titers in hamsters with pre-existing Wuhan-Hu-1 spike immunity after vaccination with Ad26.COV2.S, Ad26.COV2.S.529 or a combination of the two vaccines. Moreover, all three vaccine modalities induced equivalent protection against Omicron BA.2 challenge in these animals. Overall, our data suggest that an Omicron BA.1-based booster in rodents does not improve immunogenicity and efficacy against Omicron BA.2 over an Ad26.COV2.S booster in a setting of pre-existing immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Maarten Swart
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | | | | | | | - Ying Choi
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | | | - Erwin Claassen
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Rineke de Jong
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Katrin E Wiese
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Lisette Cornelissen
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Marieke van Es
- Wageningen Bioveterinary Research, Wageningen University & Research, Lelystad, The Netherlands
| | - Marjolein van Heerden
- Janssen Research and Development, Preclinical Sciences and Translational Safety, Beerse, Belgium
| | | | - Issam Tahiri
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | | | | | | | | | | | - Jort Vellinga
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | - Rinke Bos
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | - Frank Wegmann
- Janssen Vaccines & Prevention, Leiden, The Netherlands
| | | | | | - Roland Zahn
- Janssen Vaccines & Prevention, Leiden, The Netherlands.
| |
Collapse
|
24
|
Ortiz-Millán G. COVID-19 Health Passes: Practical and Ethical Issues. JOURNAL OF BIOETHICAL INQUIRY 2023; 20:125-138. [PMID: 36630062 PMCID: PMC9832398 DOI: 10.1007/s11673-022-10227-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/28/2022] [Indexed: 05/04/2023]
Abstract
Several countries have implemented COVID-19 health passes or certificates to promote a safer return to in-person social activities. These passes have been proposed as a way to prove that someone has been vaccinated, has recovered from the disease, or has negative results on a diagnostic test. However, many people have questioned their ethical justification. This article presents some practical and ethical problems to consider in the event of wishing to implement these passes. Among the former, it is questioned how accurate diagnostic tests are as a means of ensuring that a person is not contagious, whether vaccination guarantees immunity, the fact that health passes can be forged, whether they encourage vaccination, and the problem that there is no universally recognized health pass. Among the ethical issues, it is discussed whether health passes promote discrimination and inequality and whether they violate rights to privacy and freedom. It is concluded that health passes have enough ethical justification to be implemented.
Collapse
Affiliation(s)
- Gustavo Ortiz-Millán
- Instituto de Investigaciones Filosóficas, Universidad Nacional Autónoma de México (UNAM), Circuito Mario de la Cueva s/n, Ciudad Universitaria, Coyoacán, 04510, Mexico City, Mexico.
| |
Collapse
|
25
|
Dari A, Boulton M, Neyens M, Le Gars M, Valenzuela B, Shukarev G, Cárdenas V, Ruiz-Guiñazú J, Sadoff J, Hoetelmans RMW, Ruixo JJP. Quantifying Antibody Persistence After a Single Dose of COVID-19 Vaccine Ad26.COV2.S in Humans Using a Mechanistic Modeling and Simulation Approach. Clin Pharmacol Ther 2023; 113:380-389. [PMID: 36377532 PMCID: PMC10107600 DOI: 10.1002/cpt.2796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
Understanding persistence of humoral immune responses elicited by vaccination against coronavirus disease 2019 (COVID-19) is critical for informing the duration of protection and appropriate booster timing. We developed a mechanistic model to characterize the time course of humoral immune responses in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-seronegative adults after primary vaccination with the Janssen COVID-19 vaccine, Ad26.COV2.S. The persistence of antibody responses was quantified through mechanistic modeling-based simulations. Two biomarkers of humoral immune responses were examined: SARS-CoV-2 neutralizing antibodies determined by wild-type virus neutralization assay (wtVNA) and spike protein-binding antibodies determined by indirect spike protein enzyme-linked immunosorbent assay (S-ELISA). The persistence of antibody responses was defined as the period of time during which wtVNA and S-ELISA titers remained above the lower limit of quantification. A total of 442 wtVNA and 1,185 S-ELISA titers from 82 and 220 participants, respectively, were analyzed following administration of a single dose of Ad26.COV2.S (5 × 1010 viral particles). The mechanistic model adequately described the time course of observed wtVNA and S-ELISA serum titers and its associated variability up to 8 months following vaccination. Mechanistic model-based simulations show that single-dose Ad26.COV2.S elicits durable but waning antibody responses up to 24 months following immunization. Of the estimated model parameters, the production rate of memory B cells was decreased in older adults relative to younger adults, and the antibody production rate mediated by long-lived plasma cells was increased in women relative to men. A steeper waning of antibody responses was predicted in men and in older adults.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research and Development, Beerse, Belgium
| | | | | | | | - Belén Valenzuela
- Janssen-Cilag Spain, Part of Janssen Pharmaceutical Companies, Madrid, Spain
| | | | - Vicky Cárdenas
- Janssen Research and Development, Spring House, Pennsylvania, USA
| | | | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, The Netherlands
| | | | | |
Collapse
|
26
|
Evaluating Johnson and Johnson COVID-19 Vaccination Outcomes in a Low-Income Hispanic Population. Vaccines (Basel) 2023; 11:vaccines11010148. [PMID: 36679993 PMCID: PMC9865710 DOI: 10.3390/vaccines11010148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND A pilot study was performed at a low-income emergency care clinic to assess the humoral immune response to the Johnson & Johnson (J&J) COVID-19 vaccine (Ad26.COV2.S) to better understand how to evaluate the COVID-19 health status of its Hispanic patient population following vaccination. METHODS This study used the Clungene® SARS-CoV-2 IgG/IgM Rapid Test Cassette to determine the presence of binding antibodies resulting from the J&J COVID-19 vaccine. The Clungene test principle is based on the receptor-binding domain (RBD) of the spike protein. Antibodies targeting the spike protein are considered an appropriate measure of humoral response from spike-based vaccines. RESULTS The study confirmed previous research that antibodies wane over time, and results are consistent with reported vaccine efficacy. There was a statistically significant relationship between the humoral immune response and demographic and health status variables. CONCLUSIONS COVID-19 negative patients can be easily and efficiently monitored to determine the success and durability of COVID-19 vaccines in low-income minority populations. The use of simple low-cost spike targeted COVID-19 antibody lateral flow devices may serve as a useful adjunct to assist community-based physicians on the COVID-19 health status of its patients. Further research is needed to confirm the utility of this approach.
Collapse
|
27
|
Pierre V, Draica F, Di Fusco M, Yang J, Nunez-Gonzalez S, Kamar J, Lopez S, Moran MM, Nguyen J, Alvarez P, Cha-Silva A, Gavaghan M, Yehoshua A, Stapleton N, Burnett H. The impact of vaccination and outpatient treatment on the economic burden of Covid-19 in the United States omicron era: a systematic literature review. J Med Econ 2023; 26:1519-1531. [PMID: 37964554 DOI: 10.1080/13696998.2023.2281882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/07/2023] [Indexed: 11/16/2023]
Abstract
AIMS To identify and synthesize evidence regarding how coronavirus disease 2019 (COVID-19) interventions, including vaccines and outpatient treatments, have impacted healthcare resource use (HCRU) and costs in the United States (US) during the Omicron era. MATERIALS AND METHODS A systematic literature review (SLR) was performed to identify articles published between 1 January 2021 and 10 March 2023 that assessed the impact of vaccination and outpatient treatment on costs and HCRU outcomes associated with COVID-19. Screening was performed by two independent researchers using predefined inclusion/exclusion criteria. RESULTS Fifty-eight unique studies were included in the SLR, of which all reported HCRU outcomes, and one reported costs. Overall, there was a significant reduction in the risk of COVID-19-related hospitalization for patients who received an original monovalent primary series vaccine plus booster dose vs. no vaccination. Moreover, receipt of a booster vaccine was associated with a lower risk of hospitalization vs. primary series vaccination. Evidence also indicated a significantly reduced risk of hospitalizations among recipients of nirmatrelvir/ritonavir (NMV/r), remdesivir, sotrovimab, and molnupiravir compared to non-recipients. Treated and/or vaccinated patients also experienced reductions in intensive care unit (ICU) admissions, length of stay, and emergency department (ED)/urgent care clinic encounters. LIMITATIONS The identified studies may not represent unique patient populations as many utilized the same regional/national data sources. Synthesis of the evidence was also limited by differences in populations, outcome definitions, and varying duration of follow-up across studies. Additionally, significant gaps, including HCRU associated with long COVID and various high-risk populations and cost data, were observed. CONCLUSIONS Despite evidence gaps, findings from the SLR highlight the significant positive impact that vaccination and outpatient treatment have had on HCRU in the US, including periods of Omicron predominance. Continued research is needed to inform clinical and policy decision-making in the US as COVID-19 continues to evolve as an endemic disease.
Collapse
Affiliation(s)
- Vicki Pierre
- Evidence Synthesis, Modeling & Communication, Evidera Inc., Bethesda, MD, USA
| | - Florin Draica
- Vaccine Clinical Research, Pfizer Inc., New York, NY, USA
| | | | - Jingyan Yang
- Vaccine Clinical Research, Pfizer Inc., New York, NY, USA
| | | | - Joanna Kamar
- Evidence Synthesis, Modeling & Communication, Evidera Inc., Bethesda, MD, USA
| | - Santiago Lopez
- Vaccine Clinical Research, Pfizer Inc., New York, NY, USA
| | - Mary M Moran
- Vaccine Clinical Research, Pfizer Inc., New York, NY, USA
| | | | - Piedad Alvarez
- Evidence Synthesis, Modeling & Communication, Evidera Inc., Bethesda, MD, USA
| | | | | | - Alon Yehoshua
- Vaccine Clinical Research, Pfizer Inc., New York, NY, USA
| | - Naomi Stapleton
- Evidence Synthesis, Modeling & Communication, Evidera Inc., Bethesda, MD, USA
| | - Heather Burnett
- Evidence Synthesis, Modeling & Communication, Evidera Inc., Bethesda, MD, USA
| |
Collapse
|
28
|
Khawaja F, Papanicolaou G, Dadwal S, Pergam SA, Wingard JR, Boghdadly ZE, Abidi MZ, Waghmare A, Shahid Z, Michaels L, Hill JA, Kamboj M, Boeckh M, Auletta JJ, Chemaly RF. Frequently Asked Questions on Coronavirus Disease 2019 Vaccination for Hematopoietic Cell Transplantation and Chimeric Antigen Receptor T-Cell Recipients From the American Society for Transplantation and Cellular Therapy and the American Society of Hematology. Transplant Cell Ther 2023; 29:10-18. [PMID: 36273782 PMCID: PMC9584756 DOI: 10.1016/j.jtct.2022.10.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/13/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), disproportionately affects immunocompromised and elderly patients. Not only are hematopoietic cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cell recipients at greater risk for severe COVID-19 and COVID-19-related complications, but they also may experience suboptimal immune responses to currently available COVID-19 vaccines. Optimizing the use, timing, and number of doses of the COVID-19 vaccines in these patients may provide better protection against SARS-CoV-2 infection and better outcomes after infection. To this end, current guidelines for COVID-19 vaccination in HCT and CAR T-cell recipients from the American Society of Transplantation and Cellular Therapy Transplant Infectious Disease Special Interest Group and the American Society of Hematology are provided in a frequently asked questions format.
Collapse
Affiliation(s)
- Fareed Khawaja
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Genovefa Papanicolaou
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sanjeet Dadwal
- Division of Infectious Diseases, City of Hope, Duarte, California
| | - Steven A Pergam
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington
| | - John R Wingard
- Division of Hematology/Oncology, University of Florida, Gainesville, Florida
| | - Zeinab El Boghdadly
- Division of Infectious Diseases, The Ohio State University College of Medicine, Columbus, Ohio
| | - Maheen Z Abidi
- Division of Infectious Diseases, University of Colorado, Boulder, Colorado
| | - Alpana Waghmare
- Division of Infectious Diseases, Seattle Children's Hospital, Seattle, Washington
| | - Zainab Shahid
- Division of Infectious Diseases, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Laura Michaels
- Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Joshua A Hill
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Mini Kamboj
- Division of Infectious Diseases, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael Boeckh
- Vaccine and Infectious Diseases, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jeffery J Auletta
- National Marrow Donor Program/Center for International Blood and Marrow Transplant Research, Minneapolis, Minnesota; Divisions of Hematology/Oncology/BMT and Infectious Diseases, Nationwide Children's Hospital, Columbus, Ohio
| | - Roy F Chemaly
- Department of Infectious Diseases, Infection Control, and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
29
|
Hardt K, Vandebosch A, Sadoff J, Le Gars M, Truyers C, Lowson D, Van Dromme I, Vingerhoets J, Kamphuis T, Scheper G, Ruiz-Guiñazú J, Faust SN, Spinner CD, Schuitemaker H, Van Hoof J, Douoguih M, Struyf F, Albertson TE, Sandrock C, Lee JS, Looney MR, Tapson VF, Wiysonge CS, Velarde LHA, Backenroth D, Bhushanan J, Brandenburg B, Cárdenas V, Chen B, Chen F, Chetty P, Chu PL, Cooper K, Custers J, Delanghe H, Duca A, Henrick T, Juraszek J, Nalpas C, Peeters M, Pinheiro J, Roels S, Ryser MF, Salas J, Santoro Matias S, Scheys I, Shetty P, Shukarev G, Stoddard J, Talloen W, Tran N, Vaissiere N, van Son-Palmen E, Xu J, Goecker EA, Greninger AL, Jerome KR, Roychoudhury P, Takuva SG, Accini Mendoza JL, Achtyes E, Ahsan H, Alhatemi A, Allen N, Arribas JR, Bahrami G, Bailon L, Bajwa A, Baker J, Baron M, Benet S, Berdaï D, Berger P, Bertoch T, Bethune C, Bevilacqua S, Biagioni Santos MS, Binnian I, Bisnauthsing K, Boivin JM, Bollen H, Bonnet S, Borobia AM, Botelho-Nevers E, Bright P, Britten V, Brown C, Buadi A, Buntinx E, Burgess L, Bush L, Capeding MR, Carr QO, Carrasco Mas A, Catala H, Cathie K, Caudill TS, Cereto Castro F, Chau K, et alHardt K, Vandebosch A, Sadoff J, Le Gars M, Truyers C, Lowson D, Van Dromme I, Vingerhoets J, Kamphuis T, Scheper G, Ruiz-Guiñazú J, Faust SN, Spinner CD, Schuitemaker H, Van Hoof J, Douoguih M, Struyf F, Albertson TE, Sandrock C, Lee JS, Looney MR, Tapson VF, Wiysonge CS, Velarde LHA, Backenroth D, Bhushanan J, Brandenburg B, Cárdenas V, Chen B, Chen F, Chetty P, Chu PL, Cooper K, Custers J, Delanghe H, Duca A, Henrick T, Juraszek J, Nalpas C, Peeters M, Pinheiro J, Roels S, Ryser MF, Salas J, Santoro Matias S, Scheys I, Shetty P, Shukarev G, Stoddard J, Talloen W, Tran N, Vaissiere N, van Son-Palmen E, Xu J, Goecker EA, Greninger AL, Jerome KR, Roychoudhury P, Takuva SG, Accini Mendoza JL, Achtyes E, Ahsan H, Alhatemi A, Allen N, Arribas JR, Bahrami G, Bailon L, Bajwa A, Baker J, Baron M, Benet S, Berdaï D, Berger P, Bertoch T, Bethune C, Bevilacqua S, Biagioni Santos MS, Binnian I, Bisnauthsing K, Boivin JM, Bollen H, Bonnet S, Borobia AM, Botelho-Nevers E, Bright P, Britten V, Brown C, Buadi A, Buntinx E, Burgess L, Bush L, Capeding MR, Carr QO, Carrasco Mas A, Catala H, Cathie K, Caudill TS, Cereto Castro F, Chau K, Chavoustie S, Chowdhury M, Chronos N, Cicconi P, Cifuentes L, Cobo SM, Collins H, Colton H, Cuaño CRG, D'Onofrio V, Dargan P, Darton T, Deane P, Del Pozo JL, Derdelinckx I, Desai A, Dever M, Díaz-Pollán B, DiBuono M, Doust M, Duncan C, Echave-Sustaeta JM, Eder F, Ellis K, Elzi S, Emmett S, Engelbrecht J, Evans M, Farah T, Felton T, Ferreira JP, Floutier C, Flume P, Ford S, Fragoso V, Freedman A, Frentiu E, Galloway C, Galtier F, Garcia Diaz J, García García I, Garcia A, Gardener Z, Gauteul P, Geller S, Gibson A, Gillet C, Girerd N, Girodet PO, Gler MT, Glover R, Go HDD, Gokani K, Gonthier D, Green C, Greenberg R, Griffin C, Grobbelaar C, Guancia A, Hakkarainen G, Harris J, Hassman M, Heimer D, Hellstrom-Louw E, Herades Y, Holroyd C, Hussen N, Isidro MGD, Jackson Y, Jain M, João Filho EC, Johnson D, Jones B, Joseph N, Jumeras A, Junquera P, Kellett-Wright J, Kennedy P, Kilgore PE, Kim K, Kimmel M, Konis G, Kutner M, Lacombe K, Launay O, Lazarus R, Lederman S, Lefebvre G, Lennon Collins K, Leroux-Roels I, Lim KWO, Lins M, Liu E, Llewelyn M, Mahomed A, Maia BP, Marín-Candon A, Martínez-Gómez X, Martinot JB, Mazzella A, McCaughan F, McCormack L, McGettigan J, Mehra P, Mejeur R, Miller V, Mills A, Molto Marhuenda J, Moodley P, Mora-Rillo M, Mothe B, Mullan D, Munro A, Myers P, Nell J, Newman Lobato Souza T, O'Halloran JA, Ochoa Mazarro MD, Oliver A, Onate Gutierrez JM, Ortega J, Oshita M, Otero Romero S, Overcash JS, Owens D, Packham A, Paiva de Sousa L, Palfreeman A, Pallares CJ, Patel R, Patel S, Pelkey L, Peluso D, Penciu F, Pinto SJ, Pounds K, Pouzar J, Pragalos A, Presti R, Price D, Qureshi E, Ramalho Madruga JV, Ramesh M, Rankin B, Razat B, Riegel Santos B, Riesenberg R, Riffer E, Roche S, Rose K, Rosellini P, Rossignol P, Safirstein B, Salazar H, Sanchez Vallejo G, Santhosh S, Seco-Meseguer E, Seep M, Sherry E, Short P, Soentjens P, Solis J, Soriano Viladomiu A, Sorli C, Spangenthal S, Spence N, Stephenson E, Strout C, Surowitz R, Taladua KM, Tellalian D, Thalamas C, Thiriphoo N, Thomas J, Thomas N, Trout G, Urroz M, Veekmans B, Veekmans L, Villalobos REM, Webster B, White A, Williams G, Williams H, Wilson B, Winston A, Wiselka M, Zervos M. Efficacy, safety, and immunogenicity of a booster regimen of Ad26.COV2.S vaccine against COVID-19 (ENSEMBLE2): results of a randomised, double-blind, placebo-controlled, phase 3 trial. THE LANCET. INFECTIOUS DISEASES 2022; 22:1703-1715. [PMID: 36113538 PMCID: PMC9639796 DOI: 10.1016/s1473-3099(22)00506-0] [Show More Authors] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND Despite the availability of effective vaccines against COVID-19, booster vaccinations are needed to maintain vaccine-induced protection against variant strains and breakthrough infections. This study aimed to investigate the efficacy, safety, and immunogenicity of the Ad26.COV2.S vaccine (Janssen) as primary vaccination plus a booster dose. METHODS ENSEMBLE2 is a randomised, double-blind, placebo-controlled, phase 3 trial including crossover vaccination after emergency authorisation of COVID-19 vaccines. Adults aged at least 18 years without previous COVID-19 vaccination at public and private medical practices and hospitals in Belgium, Brazil, Colombia, France, Germany, the Philippines, South Africa, Spain, the UK, and the USA were randomly assigned 1:1 via a computer algorithm to receive intramuscularly administered Ad26.COV2.S as a primary dose plus a booster dose at 2 months or two placebo injections 2 months apart. The primary endpoint was vaccine efficacy against the first occurrence of molecularly confirmed moderate to severe-critical COVID-19 with onset at least 14 days after booster vaccination, which was assessed in participants who received two doses of vaccine or placebo, were negative for SARS-CoV-2 by PCR at baseline and on serology at baseline and day 71, had no major protocol deviations, and were at risk of COVID-19 (ie, had no PCR-positive result or discontinued the study before day 71). Safety was assessed in all participants; reactogenicity, in terms of solicited local and systemic adverse events, was assessed as a secondary endpoint in a safety subset (approximately 6000 randomly selected participants). The trial is registered with ClinicalTrials.gov, NCT04614948, and is ongoing. FINDINGS Enrolment began on Nov 16, 2020, and the primary analysis data cutoff was June 25, 2021. From 34 571 participants screened, the double-blind phase enrolled 31 300 participants, 14 492 of whom received two doses (7484 in the Ad26.COV2.S group and 7008 in the placebo group) and 11 639 of whom were eligible for inclusion in the assessment of the primary endpoint (6024 in the Ad26.COV2.S group and 5615 in the placebo group). The median (IQR) follow-up post-booster vaccination was 36·0 (15·0-62·0) days. Vaccine efficacy was 75·2% (adjusted 95% CI 54·6-87·3) against moderate to severe-critical COVID-19 (14 cases in the Ad26.COV2.S group and 52 cases in the placebo group). Most cases were due to the variants alpha (B.1.1.7) and mu (B.1.621); endpoints for the primary analysis accrued from Nov 16, 2020, to June 25, 2021, before the global dominance of delta (B.1.617.2) or omicron (B.1.1.529). The booster vaccine exhibited an acceptable safety profile. The overall frequencies of solicited local and systemic adverse events (evaluated in the safety subset, n=6067) were higher among vaccine recipients than placebo recipients after the primary and booster doses. The frequency of solicited adverse events in the Ad26.COV2.S group were similar following the primary and booster vaccinations (local adverse events, 1676 [55·6%] of 3015 vs 896 [57·5%] of 1559, respectively; systemic adverse events, 1764 [58·5%] of 3015 vs 821 [52·7%] of 1559, respectively). Solicited adverse events were transient and mostly grade 1-2 in severity. INTERPRETATION A homologous Ad26.COV2.S booster administered 2 months after primary single-dose vaccination in adults had an acceptable safety profile and was efficacious against moderate to severe-critical COVID-19. Studies assessing efficacy against newer variants and with longer follow-up are needed. FUNDING Janssen Research & Development.
Collapse
Affiliation(s)
- Karin Hardt
- Janssen Research & Development, Beerse, Belgium
| | | | | | | | | | - David Lowson
- Janssen Research & Development, High Wycombe, UK
| | | | | | | | - Gert Scheper
- Janssen Vaccines & Prevention, Leiden, Netherlands
| | | | - Saul N Faust
- NIHR Southampton Clinical Research Facility and Biomedical Research Centre, Southampton, UK; Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, UK
| | | | | | | | | | - Frank Struyf
- Janssen Research & Development, Beerse, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Dhawan M, Sharma A, Priyanka, Thakur N, Rajkhowa TK, Choudhary OP. Delta variant (B.1.617.2) of SARS-CoV-2: Mutations, impact, challenges and possible solutions. Hum Vaccin Immunother 2022; 18:2068883. [PMID: 35507895 PMCID: PMC9359381 DOI: 10.1080/21645515.2022.2068883] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/04/2022] [Accepted: 04/18/2022] [Indexed: 12/24/2022] Open
Abstract
Since commencement of COVID-19 pandemic, several SARS-CoV-2 variants have emerged amid containment efforts via vaccination. The Delta variant (B.1.617.2), discovered in October 2020, was designated as a VOC by the WHO on May 11, 2021. The enhanced transmissibility of Delta variant has been associated with critical mutations such as D614G, L452R, P681R, and T478K in the S-protein. The increased affinity of the S-protein and ACE2 has been postulated as a key reason for decreased vaccine efficacy. As per evidence, the Delta variant possesses increased transmissibility and decreased vaccine efficacy compared to other VOCs like Alpha and Beta. This has led to concerns regarding the acquisition of novel mutations in the Delta variant and outbreaks in vulnerable communities, including vaccinated people. In this mini-review of Delta variant, we have explained its evolution and characteristics, the impact of spike mutations on infectivity and immune evasion, and measures to combat future outbreaks.
Collapse
Affiliation(s)
- Manish Dhawan
- Department of Microbiology, Punjab Agricultural University, Ludhiana, India
- Trafford College, Altrincham, Manchester, UK
| | - Abhilasha Sharma
- Department of Life Science, University School of Sciences, Gujarat University, Ahmedabad, India
| | - Priyanka
- Independent Researcher, 07, Type IV Quarter, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| | - Nanamika Thakur
- Department of Medical Lab Technology, Faculty of Allied and Healthcare Sciences, GNA University, Phagwara, Punjab, India
| | - Tridib Kumar Rajkhowa
- Department of Veterinary Pathology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| | - Om Prakash Choudhary
- Department of Veterinary Anatomy and Histology, College of Veterinary Sciences and Animal Husbandry, Central Agricultural University (I), Selesih, Aizawl, Mizoram, India
| |
Collapse
|
31
|
Chun JY, Jeong H, Kim Y. Identifying susceptibility of children and adolescents to the Omicron variant (B.1.1.529). BMC Med 2022; 20:451. [PMID: 36419108 PMCID: PMC9684890 DOI: 10.1186/s12916-022-02655-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND The Omicron variant (B.1.1.529) is estimated to be more transmissible than previous strains of SARS-CoV-2 especially among children, potentially resulting in croup which is a characteristic disease in children. Current coronavirus disease 2019 (COVID-19) cases among children might be higher because (i) school-aged children have higher contact rates and (ii) the COVID-19 vaccination strategy prioritizes the elderly in most countries. However, there have been no reports confirming the age-varying susceptibility to the Omicron variant to date. METHODS We developed an age-structured compartmental model, combining age-specific contact matrix in South Korea and observed distribution of periods between each stage of infection in the national epidemiological investigation. A Bayesian inference method was used to estimate the age-specific force of infection and, accordingly, age-specific susceptibility, given epidemic data during the third (pre-Delta), fourth (Delta driven), and fifth (Omicron driven) waves in South Korea. As vaccine uptake increased, individuals who were vaccinated were excluded from the susceptible population in accordance with vaccine effectiveness against the Delta and Omicron variants, respectively. RESULTS A significant difference between the age-specific susceptibility to the Omicron and that to the pre-Omicron variants was found in the younger age group. The rise in susceptibility to the Omicron/pre-Delta variant was highest in the 10-15 years age group (5.28 times [95% CI, 4.94-5.60]), and the rise in susceptibility to the Omicron/Delta variant was highest in the 15-19 years age group (3.21 times [95% CI, 3.12-3.31]), whereas in those aged 50 years or more, the susceptibility to the Omicron/pre-Omicron remained stable at approximately twofold. CONCLUSIONS Even after adjusting for contact pattern, vaccination status, and waning of vaccine effectiveness, the Omicron variant of SARS-CoV-2 tends to propagate more easily among children than the pre-Omicron strains.
Collapse
Affiliation(s)
- June Young Chun
- Department of Internal Medicine, National Cancer Center, Goyang, South Korea.
| | - Hwichang Jeong
- Department of Statistics, Seoul National University, 56-1 Mountain, Sillim-dong, Gwanak-gu, Seoul, 08826, South Korea
| | - Yongdai Kim
- Department of Statistics, Seoul National University, 56-1 Mountain, Sillim-dong, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
32
|
Mues KE, Kirk B, Patel DA, Gelman A, Chavers LS, Talarico CA, Esposito DB, Martin D, Mansi J, Chen X, Gatto NM, Van de Velde N. Real-world comparative effectiveness of mRNA-1273 and BNT162b2 vaccines among immunocompromised adults identified in administrative claims data in the United States. Vaccine 2022; 40:6730-6739. [PMID: 36163093 PMCID: PMC9507810 DOI: 10.1016/j.vaccine.2022.09.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Head-to-head studies comparing COVID-19 mRNA vaccine effectiveness in immunocompromised individuals, who are vulnerable to severe disease are lacking, as large sample sizes are required to make meaningful inferences. METHODS This observational comparative effectiveness study was conducted in closed administrative claims data from the US HealthVerity database (December 11, 2020-January 10, 2022, before omicron). A 2-dose mRNA-1273 versus BNT162b2 regimen was assessed for preventing medically-attended breakthrough COVID-19 diagnosis and hospitalizations among immunocompromised adults. Inverse probability of treatment weighting was applied to balance baseline characteristics between vaccine groups. Incidence rates from patient-level data and hazard ratios (HRs) using weighted Cox proportional hazards models were calculated. RESULTS Overall, 57,898 and 66,981 individuals received a 2-dose regimen of mRNA-1273 or BNT161b2, respectively. Among the weighted population, mean age was 51 years, 53 % were female, and baseline immunodeficiencies included prior blood transplant (8%-9%), prior organ transplant (7%), active cancer (12%-13%), primary immunodeficiency (5-6%), HIV (20%-21%), and immunosuppressive therapy use (60%-61%). Rates per 1,000 person-years (PYs; 95% confidence intervals [CI]s) of breakthrough medically-attended COVID-19 were 25.82 (23.83-27.97) with mRNA-1273 and 30.98 (28.93, 33.18) with BNT162b2 (HR, 0.83; 95% CI, 0.75-0.93). When requiring evidence of an antigen or polymerase chain reaction test before COVID-19 diagnosis, the HR for medically-attended COVID-19 was 0.78 (0.67-0.92). Breakthrough COVID-19 hospitalization rates per 1,000 PYs (95% CI) were 3.66 (2.96-4.51) for mRNA-1273 and 4.68 (3.91-5.59) for BNT162b2 (HR, 0.78; 0.59-1.03). Utilizing open and closed claims for outcome capture only, or both cohort entry/outcome capture, produced HRs (95% CIs) for COVID-19 hospitalization of 0.72 (0.57-0.92) and 0.66 (0.58-0.76), respectively. CONCLUSIONS Among immunocompromised adults, a 2-dose mRNA-1273 regimen was more effective in preventing medically-attended COVID-19 in any setting (inpatient and outpatient) than 2-dose BNT162b2. Results were similar for COVID-19 hospitalization, although statistical power was limited when using closed claims only. STUDY REGISTRATION NCT05366322.
Collapse
Affiliation(s)
- Katherine E. Mues
- Aetion, Inc, 5 Pennsylvania Plaza, New York, NY 10001, USA,Corresponding author
| | - Brenna Kirk
- Aetion, Inc, 5 Pennsylvania Plaza, New York, NY 10001, USA
| | | | - Alice Gelman
- Aetion, Inc, 5 Pennsylvania Plaza, New York, NY 10001, USA
| | | | | | | | - David Martin
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, USA
| | - James Mansi
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, USA
| | - Xing Chen
- Moderna, Inc, 200 Technology Square, Cambridge, MA 02139, USA
| | | | | |
Collapse
|
33
|
Chavda VP, Yao Q, Vora LK, Apostolopoulos V, Patel CA, Bezbaruah R, Patel AB, Chen ZS. Fast-track development of vaccines for SARS-CoV-2: The shots that saved the world. Front Immunol 2022; 13:961198. [PMID: 36263030 PMCID: PMC9574046 DOI: 10.3389/fimmu.2022.961198] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
In December 2019, an outbreak emerged of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) which leads to coronavirus disease 2019 (COVID-19). The World Health Organisation announced the outbreak a global health emergency on 30 January 2020 and by 11 March 2020 it was declared a pandemic. The spread and severity of the outbreak took a heavy toll and overburdening of the global health system, particularly since there were no available drugs against SARS-CoV-2. With an immediate worldwide effort, communication, and sharing of data, large amounts of funding, researchers and pharmaceutical companies immediately fast-tracked vaccine development in order to prevent severe disease, hospitalizations and death. A number of vaccines were quickly approved for emergency use, and worldwide vaccination rollouts were immediately put in place. However, due to several individuals being hesitant to vaccinations and many poorer countries not having access to vaccines, multiple SARS-CoV-2 variants quickly emerged that were distinct from the original variant. Uncertainties related to the effectiveness of the various vaccines against the new variants as well as vaccine specific-side effects have remained a concern. Despite these uncertainties, fast-track vaccine approval, manufacturing at large scale, and the effective distribution of COVID-19 vaccines remain the topmost priorities around the world. Unprecedented efforts made by vaccine developers/researchers as well as healthcare staff, played a major role in distributing vaccine shots that provided protection and/or reduced disease severity, and deaths, even with the delta and omicron variants. Fortunately, even for those who become infected, vaccination appears to protect against major disease, hospitalisation, and fatality from COVID-19. Herein, we analyse ongoing vaccination studies and vaccine platforms that have saved many deaths from the pandemic.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, LM College of Pharmacy, Ahmedabad, Gujarat, India
| | - Qian Yao
- Graduate School, University of St. La Salle, Bacolod City, Philippines
| | | | | | - Chirag A. Patel
- Department of Pharmacology, LM College of Pharmacy, Ahmedabad, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam, India
| | - Aayushi B. Patel
- Pharmacy Section, LM. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| |
Collapse
|
34
|
Negroni D, Carriero S, Passarella I, Siani A, Biondetti P, Pizzolante A, Saba L, Guzzardi G. Preliminary Analysis of the Effects of Ad26.COV2.S Vaccination on CT Findings and High Intensive Care Admission Rates of COVID-19 Patients. Tomography 2022; 8:2403-2410. [PMID: 36287798 PMCID: PMC9611738 DOI: 10.3390/tomography8050199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
On 27 February 2021, the Food and Drug Administration(FDA) authorized the administration of the adenovirus-based Ad26.COV2-S vaccine (J&J-Janssen) for the prevention of COVID-19, a viral pandemic that, to date, has killed more than 5.5 million people. Performed during the early phase of the COVID-19 4th wave, this retrospective observational study aims to report the computerized tomography (CT) findings and intensive care unit admission rates of Ad26.COV2-S-vaccinated vs. unvaccinated COVID-19 patients. From the 1st to the 23rd of December 2021, all confirmed COVID-19 patients that had been subjected to chest non-contrast CT scan analysis were enrolled in the study. These were divided into Ad26.COV2.S-vaccinated (group 1) and unvaccinated patients (group 2). The RSNA severity score was calculated for each patient and correlated to CT findings and type of admission to a healthcare setting after CT-i.e., home care, ordinary hospitalization, sub-intensive care, and intensive care. Descriptive and inference statistical analyses were performed by comparing the data from the two groups. Data from a total of 71 patients were collected: 10 patients in group 1 (4M, 6F, mean age 63.5 years, SD ± 4.2) and 61 patients in group 2 (32M, 29F, mean age 64.7 years, SD ± 3.7). Statistical analysis showed lower values of RSNA severity in group 1 compared to group 2 (mean value 14.1 vs. 15.7, p = 0.009, respectively). Furthermore, vaccinated patients were less frequently admitted to both sub-intensive and high-intensive care units than group 2, with an odds ratio of 0.45 [95%CI (0.01; 3.92)]. Ad26.COV2.S vaccination protects from severe COVID-19 based on CT severity scores. As a result, Ad26.COV2.S-vaccinated COVID-19 patients are more frequently admitted to home in comparison with unvaccinated patients.
Collapse
Affiliation(s)
- Davide Negroni
- Department of Radiology, University of Piemonte Orientale, Piedmont, 28100 Novara, Italy
| | - Serena Carriero
- Department of Radiology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Ilaria Passarella
- Departement of Internal Medicine, ASST Ovest Milanese Ospedale Fornaroli, 20013 Magenta, Italy
| | - Agnese Siani
- Department of Radiology, University of Piemonte Orientale, Piedmont, 28100 Novara, Italy
| | - Pierpaolo Biondetti
- Department of Radiology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Antonio Pizzolante
- Department of Radiology, University of Piemonte Orientale, Piedmont, 28100 Novara, Italy
| | - Luca Saba
- Department of Radiology, Azienda Ospedaliero Universitaria Cagliari, 09124 Polo di Monserrato, Italy
| | - Giuseppe Guzzardi
- Department of Radiology, University of Piemonte Orientale, Piedmont, 28100 Novara, Italy
| |
Collapse
|
35
|
Affiliation(s)
- Dan H Barouch
- From Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, and the Ragon Institute of MGH, MIT, and Harvard, Cambridge - both in Massachusetts
| |
Collapse
|
36
|
Zheutlin A, Ott M, Sun R, Zemlianskaia N, Meyer CS, Rubel M, Hayden J, Neri B, Kamath T, Khan N, Schneeweiss S, Sarsour K. Durability of Protection Post-Primary COVID-19 Vaccination in the United States. Vaccines (Basel) 2022; 10:vaccines10091458. [PMID: 36146536 PMCID: PMC9505933 DOI: 10.3390/vaccines10091458] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 12/27/2022] Open
Abstract
The durability of immune responses after COVID-19 vaccination will drive long-term vaccine effectiveness across settings and may differ by vaccine type. To determine durability of protection of COVID-19 vaccines (BNT162b2, mRNA-1273, and Ad26.COV2.S) following primary vaccination in the United States, a matched case-control study was conducted in three cohorts between 1 January and 7 September 2021 using de-identified data from a database covering 168 million lives. Odds ratios (ORs) for developing outcomes of interest (breakthrough SARS-CoV-2 infection, hospitalization, or intensive care unit admission) were determined for each vaccine (no direct comparisons). In total, 17,017,435 individuals were identified. Relative to the baseline, stable protection was observed for Ad26.COV2.S against infections (OR [95% confidence interval (CI)], 1.31 [1.18–1.47]) and hospitalizations (OR [95% CI], 1.25 [0.86–1.80]). Relative to the baseline, protection waned over time against infections for BNT162b2 (OR [95% CI], 2.20 [2.01–2.40]) and mRNA-1273 (OR [95% CI], 2.07 [1.87–2.29]) and against hospitalizations for BNT162b2 (OR [95% CI], 2.38 [1.79–3.17]). Baseline protection remained stable for intensive care unit admissions for all three vaccines. Calculated baseline VE was consistent with published literature. This study suggests that the three vaccines in three separate populations may have different durability profiles.
Collapse
Affiliation(s)
- Amanda Zheutlin
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Miles Ott
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Ran Sun
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Natalia Zemlianskaia
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Craig S. Meyer
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Meagan Rubel
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Jennifer Hayden
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Breno Neri
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Tripthi Kamath
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Najat Khan
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02120, USA
| | - Khaled Sarsour
- Data Sciences, Research & Development, Janssen Pharmaceuticals, Titusville, NJ 08560, USA
- Correspondence: ; Tel.: +1-650-296-0719
| |
Collapse
|
37
|
Chanda D, Hines JZ, Itoh M, Fwoloshi S, Minchella PA, Zyambo KD, Sivile S, Kampamba D, Chirwa B, Chanda R, Mutengo K, Kayembe MF, Chewe W, Chipimo P, Mweemba A, Agolory S, Mulenga LB. COVID-19 Vaccine Effectiveness Against Progression to In-Hospital Mortality in Zambia, 2021-2022. Open Forum Infect Dis 2022; 9:ofac469. [PMID: 36196297 PMCID: PMC9522674 DOI: 10.1093/ofid/ofac469] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/08/2022] [Indexed: 09/24/2023] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) vaccines are highly effective for reducing severe disease and mortality. However, vaccine effectiveness data are limited from Sub-Saharan Africa. We report COVID-19 vaccine effectiveness against progression to in-hospital mortality in Zambia. Methods We conducted a retrospective cohort study among admitted patients at 8 COVID-19 treatment centers across Zambia during April 2021 through March 2022, when the Delta and Omicron variants were circulating. Patient demographic and clinical information including vaccination status and hospitalization outcome (discharged or died) were collected. Multivariable logistic regression was used to assess the odds of in-hospital mortality by vaccination status, adjusted for age, sex, number of comorbid conditions, disease severity, hospitalization month, and COVID-19 treatment center. Vaccine effectiveness of ≥1 vaccine dose was calculated from the adjusted odds ratio. Results Among 1653 patients with data on their vaccination status and hospitalization outcome, 365 (22.1%) died. Overall, 236 (14.3%) patients had received ≥1 vaccine dose before hospital admission. Of the patients who had received ≥1 vaccine dose, 22 (9.3%) died compared with 343 (24.2%) among unvaccinated patients (P < .01). The median time since receipt of a first vaccine dose (interquartile range) was 52.5 (28-107) days. Vaccine effectiveness for progression to in-hospital mortality among hospitalized patients was 64.8% (95% CI, 42.3%-79.4%). Conclusions Among patients admitted to COVID-19 treatment centers in Zambia, COVID-19 vaccination was associated with lower progression to in-hospital mortality. These data are consistent with evidence from other countries demonstrating the benefit of COVID-19 vaccination against severe complications. Vaccination is a critical tool for reducing the consequences of COVID-19 in Zambia.
Collapse
Affiliation(s)
- Duncan Chanda
- University Teaching Hospital, Lusaka, Zambia
- Ministry of Health, Lusaka, Zambia
| | - Jonas Z Hines
- US Centers for Disease Control and Prevention, Lusaka, Zambia
| | - Megumi Itoh
- US Centers for Disease Control and Prevention, Lusaka, Zambia
| | - Sombo Fwoloshi
- University Teaching Hospital, Lusaka, Zambia
- Ministry of Health, Lusaka, Zambia
- University of Zambia School of Medicine, Lusaka, Zambia
| | | | - Khozya D Zyambo
- University Teaching Hospital, Lusaka, Zambia
- Ministry of Health, Lusaka, Zambia
| | - Suilanji Sivile
- University Teaching Hospital, Lusaka, Zambia
- Ministry of Health, Lusaka, Zambia
| | | | | | | | | | | | | | - Peter Chipimo
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Aggrey Mweemba
- Levy Mwanawasa University Teaching Hospital, Lusaka, Zambia
| | - Simon Agolory
- US Centers for Disease Control and Prevention, Lusaka, Zambia
| | | |
Collapse
|
38
|
Le Gars M, Hendriks J, Sadoff J, Ryser M, Struyf F, Douoguih M, Schuitemaker H. Immunogenicity and efficacy of Ad26.COV2.S: An adenoviral vector-based COVID-19 vaccine. Immunol Rev 2022; 310:47-60. [PMID: 35689434 PMCID: PMC9349621 DOI: 10.1111/imr.13088] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/02/2022] [Indexed: 12/26/2022]
Abstract
Since its emergence in late 2019, the coronavirus disease 2019 (COVID-19) pandemic has caused substantial morbidity and mortality. Despite the availability of efficacious vaccines, new variants with reduced sensitivity to vaccine-induced protection are a troubling new reality. The Ad26.COV2.S vaccine is a recombinant, replication-incompetent human adenovirus type 26 vector encoding a full-length, membrane-bound severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein in a prefusion-stabilized conformation. This review discusses the immunogenicity and efficacy of Ad26.COV2.S as a single-dose primary vaccination and as a homologous or heterologous booster vaccination. Ad26.COV2.S elicits broad humoral and cellular immune responses, which are associated with protective efficacy/effectiveness against SARS-CoV-2 infection, moderate to severe/critical COVID-19, and COVID-19-related hospitalization and death, including against emerging SARS-CoV-2 variants. The humoral immune responses elicited by Ad26.COV2.S vaccination are durable, continue to increase for at least 2-3 months postvaccination, and involve a range of functional antibodies. Ad26.COV2.S given as a heterologous booster to mRNA vaccine-primed individuals markedly increases humoral and cellular immune responses. The use of Ad26.COV2.S as primary vaccination and as part of booster regimens is supporting the ongoing efforts to control and mitigate the COVID-19 pandemic.
Collapse
Affiliation(s)
| | - Jenny Hendriks
- Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, The Netherlands
| | - Martin Ryser
- Janssen Research and Development, Beerse, Belgium
| | - Frank Struyf
- Janssen Research and Development, Beerse, Belgium
| | | | | |
Collapse
|
39
|
Patel RS, Agrawal B. Heterologous immunity induced by 1 st generation COVID-19 vaccines and its role in developing a pan-coronavirus vaccine. Front Immunol 2022; 13:952229. [PMID: 36045689 PMCID: PMC9420909 DOI: 10.3389/fimmu.2022.952229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 07/19/2022] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome virus-2 (SARS-CoV-2), the causative infectious agent of the COVID-19 pandemic, has led to multiple (4-6) waves of infections worldwide during the past two years. The development of vaccines against SARS-CoV-2 has led to successful mass immunizations worldwide, mitigating the worldwide mortality due the pandemic to a great extent. Yet the evolution of new variants highlights a need to develop a universal vaccine which can prevent infections from all virulent SARS-CoV-2. Most of the current first generation COVID-19 vaccines are based on the Spike protein from the original Wuhan-hu-1 virus strain. It is encouraging that they still protect from serious illnesses, hospitalizations and mortality against a number of mutated viral strains, to varying degrees. Understanding the mechanisms by which these vaccines provide heterologous protection against multiple highly mutated variants can reveal strategies to develop a universal vaccine. In addition, many unexposed individuals have been found to harbor T cells that are cross-reactive against SARS-CoV-2 antigens, with a possible protective role. In this review, we will discuss various aspects of natural or vaccine-induced heterologous (cross-reactive) adaptive immunity against SARS-CoV-2 and other coronaviruses, and their role in achieving the concept of a pan-coronavirus vaccine.
Collapse
Affiliation(s)
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
40
|
Tan CS, Collier ARY, Yu J, Liu J, Chandrashekar A, McMahan K, Jacob-Dolan C, He X, Roy V, Hauser BM, Munt JE, Mallory ML, Mattocks M, Powers JM, Meganck RM, Rowe M, Hemond R, Bondzie EA, Jaegle KH, Baric RS, Schmidt AG, Alter G, Le Gars M, Sadoff J, Barouch DH. Durability of Heterologous and Homologous COVID-19 Vaccine Boosts. JAMA Netw Open 2022; 5:e2226335. [PMID: 35947380 PMCID: PMC9366542 DOI: 10.1001/jamanetworkopen.2022.26335] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
IMPORTANCE Antibody responses elicited by current messenger RNA (mRNA) COVID-19 vaccines decline rapidly and require repeated boosting. OBJECTIVE To evaluate the immunogenicity and durability of heterologous and homologous prime-boost regimens involving the adenovirus vector vaccine Ad26.COV2.S and the mRNA vaccine BNT162b2. DESIGN, SETTING, AND PARTICIPANTS In this cohort study at a single clinical site in Boston, Massachusetts, 68 individuals who were vaccinated at least 6 months previously with 2 immunizations of BNT162b2 were boosted with either Ad26.COV2.S or BNT162b2. Enrollment of participants occurred from August 12, 2021, to October 25, 2021, and this study involved 4 months of follow-up. Data analysis was performed from November 2021 to February 2022. EXPOSURES Participants who were previously vaccinated with BNT162b2 received a boost with either Ad26.COV2.S or BNT162b2. MAIN OUTCOMES AND MEASURES Humoral immune responses were assessed by neutralizing, binding, and functional antibody responses for 16 weeks following the boost. CD8+ and CD4+ T-cell responses were evaluated by intracellular cytokine staining assays. RESULTS Among 68 participants who were originally vaccinated with BNT162b2 and boosted with Ad26.COV2.S (41 participants; median [range] age, 36 [23-84] years) or BNT162b2 (27 participants; median [range] age, 35 [23-76] years), 56 participants (82%) were female, 7 (10%) were Asian, 4 (6%) were Black, 4 (6%) were Hispanic or Latino, 3 (4%) were more than 1 race, and 53 (78%) were White. Both vaccines were found to be associated with increased humoral and cellular immune responses, including against SARS-CoV-2 variants of concern. BNT162b2 boosting was associated with a rapid increase of Omicron neutralizing antibodies that peaked at a median (IQR) titer of 1018 (699-1646) at week 2 and declined by 6.9-fold to a median (IQR) titer of 148 (95-266) by week 16. Ad26.COV2.S boosting was associated with increased Omicron neutralizing antibodies titers that peaked at a median (IQR) of 859 (467-1838) week 4 and declined by 2.1-fold to a median (IQR) of 403 (208-1130) by week 16. CONCLUSIONS AND RELEVANCE Heterologous Ad26.COV2.S boosting was associated with durable humoral and cellular immune responses in individuals who originally received the BNT162b2 vaccine. These data suggest potential benefits of heterologous prime-boost vaccine regimens for SARS-CoV-2.
Collapse
Affiliation(s)
- C. Sabrina Tan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Ai-ris Y. Collier
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Jinyan Liu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Abishek Chandrashekar
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Katherine McMahan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Xuan He
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Vicky Roy
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge
| | - Blake M. Hauser
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge
| | | | | | | | | | | | - Marjorie Rowe
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Rachel Hemond
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Esther A. Bondzie
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Kate H. Jaegle
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | | | - Aaron G. Schmidt
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge
| | - Galit Alter
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard, Cambridge
| | | | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, Netherlands
| | - Dan H. Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| |
Collapse
|
41
|
The Role of Cellular Immunity in the Protective Efficacy of the SARS-CoV-2 Vaccines. Vaccines (Basel) 2022; 10:vaccines10071103. [PMID: 35891267 PMCID: PMC9324880 DOI: 10.3390/vaccines10071103] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/30/2022] Open
Abstract
Multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines have been approved for clinical use. SARS-CoV-2 neutralizing antibody titers after immunization are widely used as an evaluation indicator, and the roles of cellular immune responses in the protective efficacy of vaccines are rarely mentioned. However, therapeutic monoclonal neutralizing antibodies have shown limited efficacy in improving the outcomes of hospitalized patients with coronavirus disease 2019 (COVID-19), suggesting a passive role of cellular immunity in SARS-CoV-2 vaccines. The synergistic effect of virus-specific humoral and cellular immune responses helps the host to fight against viral infection. In fact, it has been observed that the early appearance of specific T-cell responses is strongly correlated with mild symptoms of COVID-19 patients and that individuals with pre-existing SARS-CoV-2 nonstructural-protein-specific T cells are more resistant to SARS-CoV-2 infection. These findings suggest the important contribution of the cellular immune response to the fight against SARS-CoV-2 infection and severe COVID-19. Nowadays, new SARS-CoV-2 variants that can escape from the neutralization of antibodies are rapidly increasing. However, the epitopes of these variants recognized by T cells are largely preserved. Paying more attention to cellular immune responses may provide new instructions for designing effective vaccines for the prevention of severe disease induced by the break-through infection of new variants and the sequelae caused by virus latency. In this review, we deliberate on the role of cellular immunity against COVID-19 and summarize recent advances in the development of SARS-CoV-2 vaccines and the immune responses induced by vaccines to improve the design of new vaccines and immunization strategies.
Collapse
|
42
|
Shafqat A, Arabi TZ, Sabbah BN, Abdulkader HS, Shafqat S, Razak A, Kashir J, Alkattan K, Yaqinuddin A. Understanding COVID-19 Vaccines Today: Are T-cells Key Players? Vaccines (Basel) 2022; 10:904. [PMID: 35746512 PMCID: PMC9227180 DOI: 10.3390/vaccines10060904] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 02/05/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has heavily mutated since the beginning of the coronavirus-2019 (COVID-19) pandemic. In this regard, the so-called variants of concern (VOCs) feature mutations that confer increased transmissibility and evasion of antibody responses. The VOCs have caused significant spikes in COVID-19 cases, raising significant concerns about whether COVID-19 vaccines will protect against current and future variants. In this context, whereas the protection COVID-19 vaccines offer against the acquisition of infection appears compromised, the protection against severe COVID-19 is maintained. From an immunologic standpoint, this is likely underpinned by the maintenance of T-cell responses against VOCs. Therefore, the role of T-cells is essential to understanding the broader adaptive immune response to COVID-19, which has the potential to shape public policies on vaccine protocols and inform future vaccine design. In this review, we survey the literature on the immunology of T-cell responses upon SARS-CoV-2 vaccination with the current FDA-approved and Emergency Use Authorized COVID-19 vaccines.
Collapse
Affiliation(s)
- Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Tarek Z. Arabi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Belal N. Sabbah
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Humzah S. Abdulkader
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Shameel Shafqat
- Medical College, Aga Khan University, P.O. Box 3500, Karachi, Pakistan;
| | - Adhil Razak
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Junaid Kashir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Khaled Alkattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (T.Z.A.); (B.N.S.); (H.S.A.); (A.R.); (J.K.); (K.A.); (A.Y.)
| |
Collapse
|