1751
|
Eckman MH, Powers-Fletcher MV, Forrester JW, Fichtenbaum CJ, Lofgren R, Smulian AG. Take Your Best Shot: Which SARS-CoV-2 Vaccine Should I Get? MDM Policy Pract 2021; 6:23814683211031226. [PMID: 34621992 PMCID: PMC8492082 DOI: 10.1177/23814683211031226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 06/18/2021] [Indexed: 11/16/2022] Open
Abstract
Background. Three vaccines against SARS-CoV-2 (severe acute respiratory syndrome coronavirus-2) have now received emergency use authorization by the US Food and Drug Administration. Patients may have the opportunity to make a choice about which vaccine they prefer to receive. Vaccine hesitancy is a hurdle to the development of widespread immunity, with many patients struggling to decide whether to get vaccinated at all. Objective. Develop a decision model exploring the question, "Should I get vaccinated with mRNA or adenovirus vector vaccine (AVV) if either is available now?"Design. Markov state transition model with lifetime time horizon. Data Sources. MEDLINE searches, bibliographies from relevant English-language articles. Setting. United States, ambulatory clinical setting. Participants. Previously uninfected, nonimmunized adults in the United States. Interventions. 1) Do Not Vaccinate, 2) Vaccination with mRNA Vaccine, 3) Vaccination with Adenovirus Vector Vaccine. Main Measures. Quality-adjusted life years (QALYs). Key Results. Base case-for a healthy 65-year-old patient, both vaccines yield virtually equivalent results (difference of 0.0028 QALYs). In sensitivity analyses, receiving the AVV is preferred if the short-term morbidity associated with each vaccine dose exceeds 1.8 days. Both vaccines afford an even greater benefit compared with Do Not Vaccinate if the pandemic is in a surge phase with a rising incidence of infection or if the current 7-day incidence is greater than the base case estimate of 105 cases per 100,000. Conclusions. Preferred vaccination strategies change under differing assumptions, but differences in outcomes are negligible. The best advice for patients is to get vaccinated against COVID-19 disease with whatever vaccine is available first. Providing mRNA vaccine to the remaining eligible US population would result in an aggregate gain of 3.92 million QALYs.
Collapse
Affiliation(s)
- Mark H Eckman
- Division of General Internal Medicine and the Center for Clinical Effectiveness, University of Cincinnati Medical Center, Cincinnati, Ohio
| | | | - Jennifer W Forrester
- Division of Infectious Diseases, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Richard Lofgren
- Division of General Internal Medicine and the Center for Clinical Effectiveness, University of Cincinnati Medical Center, Cincinnati, Ohio
| | - Alan George Smulian
- Division of Infectious Diseases, University of Cincinnati Medical Center, Cincinnati, Ohio
| |
Collapse
|
1752
|
Kasapçopur Ö. A Big Problem and Debate in COVID-19 Pandemics: Equitable and Effective Access of the COVID-19 Vaccines and Waiver of Intellectual Property. Turk Arch Pediatr 2021; 56:283-284. [PMID: 35005720 PMCID: PMC8655952 DOI: 10.5152/turkarchpediatr.2021.180621] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Özgür Kasapçopur
- Department of Pediatric Rheumatology, Istanbul University Cerrahpasa School of Medicine, Istanbul, Turkey
| |
Collapse
|
1753
|
Deplanque D, Launay O. Efficacy of COVID-19 vaccines: From clinical trials to real life. Therapie 2021; 76:277-283. [PMID: 34049688 PMCID: PMC8114590 DOI: 10.1016/j.therap.2021.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/12/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly spread around the globe leading to the COVID-19 pandemic. To mitigate the effects of the virus on public health and the global economy, vaccines were rapidly developed. In less than one year, with respect to usual clinical development rules, several vaccines have been put on the market and mass vaccination campaigns have been deployed. During the phase I to phase III clinical trials, most of these vaccines have demonstrated both their safety and efficacy. Despite questions remain about the impact of virus variants and the duration of the immune response, messenger RNA (mRNA)-based and adenoviral vectored vaccines have demonstrated an overall efficacy from 70 to 95% in both phase III trials and real life. In addition, all these vaccines also reduce the severe forms of the disease and might strongly impact the mortality which could change the course of the pandemic.
Collapse
Affiliation(s)
- Dominique Deplanque
- Université de Lille, Inserm, CHU Lille, CIC 1403 - Clinical Investigation Center, 59000 Lille, France.; F-CRIN IREIVAC/COVIREIVAC, 75679 Paris, France.
| | - Odile Launay
- F-CRIN IREIVAC/COVIREIVAC, 75679 Paris, France; Université de Paris, Inserm CIC 1417, Assistance publique - Hôpitaux de Paris, hôpital Cochin, 75679 Paris, France
| |
Collapse
|
1754
|
Differences in the Protection Motivation Theory Constructs between People with Various Latent Classes of Motivation for Vaccination and Preventive Behaviors against COVID-19 in Taiwan. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18137042. [PMID: 34280979 PMCID: PMC8297011 DOI: 10.3390/ijerph18137042] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023]
Abstract
The present study aimed to identify the distinct classes of motivations to get vaccinated and to adopt preventive behaviors against the coronavirus disease 2019 (COVID-19) among people in Taiwan and to examine the roles of Protection Motivation Theory (PMT) cognitive constructs in these unique classes of motivations to get vaccinated and to adopt preventive behaviors. We recruited 1047 participants by using a Facebook advertisement. Participants’ motivations to get vaccinated and to adopt preventive behaviors against COVID-19, PMT constructs of threat appraisal (perceived severity and perceived vulnerability), and PMT constructs of coping appraisal (self-efficacy, response efficacy, response cost, knowledge, and previous vaccination for seasonal influenza) were determined. We analyzed participants’ motivations to get vaccinated and to adopt preventive behaviors against COVID-19 by using latent profile analysis. The present study identified three latent classes, including the participants with high motivation for vaccination and preventive behaviors (the class of Both High), those with low motivation for vaccination and preventive behaviors (the class of Both Low), and those with high motivation for vaccination but low motivation for preventive behaviors (the class of High Vaccination but Low Preventive Behaviors). Compared with the participants in the class of Both High, participants in the class of Both Low had lower levels of perceived vulnerability, perceived severity, self-efficacy to have vaccination, response efficacy of vaccination, knowledge about vaccination, and previous vaccination for seasonal influenza; participants in the class of High Vaccination but Low Preventive Behaviors had lower levels of perceived vulnerability and perceived severity but higher levels of response cost of vaccination. We concluded that varieties of motivations, threat, and coping appraisals should be considered in intervention programs aiming to increase motivation to adopt recommended protective behaviors against COVID-19.
Collapse
|
1755
|
Castonguay N, Zhang W, Langlois MA. Meta-Analysis and Structural Dynamics of the Emergence of Genetic Variants of SARS-CoV-2. Front Microbiol 2021; 12:676314. [PMID: 34267735 PMCID: PMC8276313 DOI: 10.3389/fmicb.2021.676314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/27/2021] [Indexed: 12/18/2022] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in late December 2019 in Wuhan, China, and is the causative agent for the worldwide COVID-19 pandemic. SARS-CoV-2 is a positive-sense single-stranded RNA virus belonging to the betacoronavirus genus. Due to the error-prone nature of the viral RNA-dependent polymerase complex, coronaviruses are known to acquire new mutations at each cycle of genome replication. This constitutes one of the main factors driving the evolution of its relatively large genome and the emergence of new genetic variants. In the past few months, the identification of new B.1.1.7 (United Kingdom), B.1.351 (South Africa), and P.1 (Brazil) variants of concern (VOC) has highlighted the importance of tracking the emergence of mutations in the SARS-CoV-2 genome that impact transmissibility, virulence, and immune and neutralizing antibody escape. Here we analyzed the appearance and prevalence trajectory over time of mutations that appeared in all SARS-CoV-2 genes from December 2019 to April 2021. The goal of the study was to identify which genetic modifications are the most frequent and study the dynamics of their propagation, their incorporation into the consensus sequence, and their impact on virus biology. We also analyzed the structural properties of the spike glycoprotein of the B.1.1.7, B.1.351, and P.1 variants for its binding to the host receptor ACE2. This study offers an integrative view of the emergence, disappearance, and consensus sequence integration of successful mutations that constitute new SARS-CoV-2 variants and their impact on neutralizing antibody therapeutics and vaccines.
Collapse
Affiliation(s)
- Nicolas Castonguay
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wandong Zhang
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- uOttawa Center for Infection, Immunity and Inflammation (CI3), Ottawa, ON, Canada
| |
Collapse
|
1756
|
FDA-authorized mRNA COVID-19 vaccines are effective per real-world evidence synthesized across a multi-state health system. MED 2021; 2:979-992.e8. [PMID: 34223401 PMCID: PMC8238652 DOI: 10.1016/j.medj.2021.06.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/05/2021] [Accepted: 06/17/2021] [Indexed: 02/01/2023]
Abstract
Background Two US Food and Drug Administration (FDA)-authorized coronavirus disease 2019 (COVID-19) mRNA vaccines, BNT162b2 (Pfizer/BioNTech) and mRNA-1273 (Moderna), have demonstrated high efficacy in large phase 3 randomized clinical trials. It is important to assess their effectiveness in a real-world setting. Methods This is a retrospective analysis of 136,532 individuals in the Mayo Clinic health system (Arizona, Florida, Iowa, Minnesota, and Wisconsin) with PCR testing data between December 1, 2020 and April 20, 2021. We compared clinical outcomes for a vaccinated cohort of 68,266 individuals who received at least one dose of either vaccine (nBNT162b2 = 51,795; nmRNA-1273 = 16,471) and an unvaccinated control cohort of 68,266 individuals propensity matched based on relevant demographic, clinical, and geographic features. We estimated real-world vaccine effectiveness by comparing incidence rates of positive severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) PCR testing and COVID-19-associated hospitalization and intensive care unit (ICU) admission starting 7 days after the second vaccine dose. Findings The real-world vaccine effectiveness of preventing SARS-CoV-2 infection was 86.1% (95% confidence interval [CI]: 82.4%-89.1%) for BNT162b2 and 93.3% (95% CI: 85.7%-97.4%) for mRNA-1273. BNT162b2 and mRNA-1273 were 88.8% (95% CI: 75.5%-95.7%) and 86.0% (95% CI: 71.6%-93.9%) effective in preventing COVID-19-associated hospitalization. Both vaccines were 100% effective (95% CIBNT162b2: 51.4%-100%; 95% CImRNA-1273: 43.3%-100%) in preventing COVID-19-associated ICU admission. Conclusions BNT162b2 and mRNA-1273 are effective in a real-world setting and are associated with reduced rates of SARS-CoV-2 infection and decreased burden of COVID-19 on the healthcare system. Funding This study was funded by nference.
Collapse
|
1757
|
Impaired humoral responses to COVID-19 vaccination in patients with lymphoma receiving B-cell directed therapies. Blood 2021; 138:811-814. [PMID: 34189565 PMCID: PMC8245303 DOI: 10.1182/blood.2021012443] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022] Open
|
1758
|
Lyubavina N, Saltsev S, Menkov N, Tyurikova L, Plastinina S, Shonia M, Tulichev A, Milyutina M, Makarova E. Immunological Approaches to the Treatment of New Coronavirus Infection (Review). Sovrem Tekhnologii Med 2021; 13:81-99. [PMID: 34603758 PMCID: PMC8482822 DOI: 10.17691/stm2021.13.3.09] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Indexed: 01/08/2023] Open
Abstract
The pandemic of the new coronavirus infection (COVID-19) caused by the SARS-CoV-2 virus has spread all over the world. The large amount of information that appears every day requires comprehension and systematization. The immunological aspects of the virus-host interaction are the core issues in the effective treatment and prevention of COVID-19' development. The review analyzes the known pathways of the viral invasion and evasion, the mechanisms of the cytokine storm, endothelial damage, and hypercoagulability associated with SARS-CoV-2 infection. Clinical data from previous SARS and MERS epidemics is discussed here. We also address the therapeutic approaches based on the basic knowledge of immune response and the blood cells' immune functions, as well as the ways to reduce their hyperactivation. The use of interferon therapy, anti-inflammatory therapy, anti-cytokine therapy, neutralizing antibodies, convalescent plasma, and mesenchymal stem cells, as well as prophylactic vaccines, is discussed.
Collapse
Affiliation(s)
- N.A. Lyubavina
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - S.G. Saltsev
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - N.V. Menkov
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - L.V. Tyurikova
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - S.S. Plastinina
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M.L. Shonia
- Associate Professor, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - A.A. Tulichev
- Assistant, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - M.Yu. Milyutina
- Assistant, Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| | - E.V. Makarova
- Associate Professor, Head of the Department of Propedeutics of Internal Diseases; Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Square, Nizhny Novgorod, 603005, Russia
| |
Collapse
|
1759
|
Duerr R, Crosse KM, Valero-Jimenez AM, Dittmann M. SARS-CoV-2 Portrayed against HIV: Contrary Viral Strategies in Similar Disguise. Microorganisms 2021; 9:1389. [PMID: 34198973 PMCID: PMC8307803 DOI: 10.3390/microorganisms9071389] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 and HIV are zoonotic viruses that rapidly reached pandemic scale, causing global losses and fear. The COVID-19 and AIDS pandemics ignited massive efforts worldwide to develop antiviral strategies and characterize viral architectures, biological and immunological properties, and clinical outcomes. Although both viruses have a comparable appearance as enveloped viruses with positive-stranded RNA and envelope spikes mediating cellular entry, the entry process, downstream biological and immunological pathways, clinical outcomes, and disease courses are strikingly different. This review provides a systemic comparison of both viruses' structural and functional characteristics, delineating their distinct strategies for efficient spread.
Collapse
Affiliation(s)
- Ralf Duerr
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA; (K.M.C.); (A.M.V.-J.); (M.D.)
| | | | | | | |
Collapse
|
1760
|
Manuel O. COVID-19 vaccination in solid-organ transplant recipients: generating new data as fast as possible, but taking clinical decisions as slow as necessary. Clin Microbiol Infect 2021; 27:1070-1071. [PMID: 34153456 PMCID: PMC8213513 DOI: 10.1016/j.cmi.2021.06.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/07/2021] [Accepted: 06/11/2021] [Indexed: 01/06/2023]
Affiliation(s)
- Oriol Manuel
- Infectious Diseases Service and Transplantation Centre, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
1761
|
|
1762
|
Flacco ME, Soldato G, Acuti Martellucci C, Carota R, Di Luzio R, Caponetti A, Manzoli L. Interim Estimates of COVID-19 Vaccine Effectiveness in a Mass Vaccination Setting: Data from an Italian Province. Vaccines (Basel) 2021; 9:628. [PMID: 34200538 PMCID: PMC8227269 DOI: 10.3390/vaccines9060628] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
This retrospective cohort study compared the rates of virologically-confirmed SARS-CoV-2 infections, symptomatic or lethal COVID-19 among the residents of the Italian province of Pescara who received one or two doses of COVID-19 vaccines, versus the unvaccinated. The official data of the National Health System were used, and a total of 69,539 vaccinated adults were compared with 175,687 unvaccinated. Among the subjects who received at least one vaccine dose, 85 infections (0.12%), 18 severe and 3 lethal COVID-19 cases were recorded after an average follow-up of 38 days. Among the unvaccinated, the numbers were 6948 (4.00%), 933 (0.53%) and 241 (0.14%), respectively. The serious adverse event reports-yet unconfirmed-were 24 out of 102,394 administered doses. In a Cox model, adjusting for age, gender, and selected comorbidities, the effectiveness of either BNT162b2, ChAdOx1 nCoV-19 or mRNA-1273 vaccines was higher than 95% in preventing infections (mostly due to B.1.1.7 variant), symptomatic or lethal COVID-19. No differences were observed across genders, and among the 691 subjects who received the second dose of vaccine later than the recommended date. Although preliminary, these findings support current immunization policies and may help reducing vaccine hesitancy.
Collapse
Affiliation(s)
- Maria Elena Flacco
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.E.F.); (C.A.M.)
| | - Graziella Soldato
- Local Health Unit of Pescara, 65124 Pescara, Italy; (G.S.); (R.C.); (R.D.L.); (A.C.)
| | | | - Roberto Carota
- Local Health Unit of Pescara, 65124 Pescara, Italy; (G.S.); (R.C.); (R.D.L.); (A.C.)
| | - Rossano Di Luzio
- Local Health Unit of Pescara, 65124 Pescara, Italy; (G.S.); (R.C.); (R.D.L.); (A.C.)
| | - Antonio Caponetti
- Local Health Unit of Pescara, 65124 Pescara, Italy; (G.S.); (R.C.); (R.D.L.); (A.C.)
| | - Lamberto Manzoli
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy; (M.E.F.); (C.A.M.)
| |
Collapse
|
1763
|
Rubbert-Roth A, Vuilleumier N, Ludewig B, Schmiedeberg K, Haller C, von Kempis J. Anti-SARS-CoV-2 mRNA vaccine in patients with rheumatoid arthritis. LANCET RHEUMATOLOGY 2021; 3:e470-e472. [PMID: 34124693 PMCID: PMC8186851 DOI: 10.1016/s2665-9913(21)00186-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
| | - Nicolas Vuilleumier
- Laboratory Medicine Division, Geneva University Hospitals, Geneva, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St Gallen, St Gallen, Switzerland
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Christoph Haller
- Division of Rheumatology, Kantonsspital St Gallen, St Gallen, Switzerland
| | | |
Collapse
|
1764
|
Feraoun Y, Maisonnasse P, Le Grand R, Beignon AS. [COVID-19: Warp Speed vaccines]. Med Sci (Paris) 2021; 37:759-772. [PMID: 34080537 DOI: 10.1051/medsci/2021094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
A vaccine is required to effectively control the COVID-19 pandemic in the mid and long term. The development of vaccines against SARS-CoV-2 was initiated as soon as the genetic sequence of the virus was published, and has evolved at an unprecedented speed, with a first clinical trial launched in March 2020. One year later, more than a dozen of vaccines based on different concepts, with some having been evaluated only in clinical trials so far, are authorized under emergency procedures. Here, we review these vaccines, compare their properties and discuss the challenges they face, including the emergence of viral variants of concern.
Collapse
Affiliation(s)
- Yanis Feraoun
- Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB), Département IDMIT (Infectious Disease Models and Innovative Therapies), UMR 1184, Université Paris-Saclay, Unité Inserm 1184, CEA, 18 route du Panorama, 92265 Fontenay-aux-Roses, France
| | - Pauline Maisonnasse
- Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB), Département IDMIT (Infectious Disease Models and Innovative Therapies), UMR 1184, Université Paris-Saclay, Unité Inserm 1184, CEA, 18 route du Panorama, 92265 Fontenay-aux-Roses, France
| | - Roger Le Grand
- Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB), Département IDMIT (Infectious Disease Models and Innovative Therapies), UMR 1184, Université Paris-Saclay, Unité Inserm 1184, CEA, 18 route du Panorama, 92265 Fontenay-aux-Roses, France
| | - Anne-Sophie Beignon
- Immunologie des maladies virales, auto-immunes, hématologiques et bactériennes (IMVA-HB), Département IDMIT (Infectious Disease Models and Innovative Therapies), UMR 1184, Université Paris-Saclay, Unité Inserm 1184, CEA, 18 route du Panorama, 92265 Fontenay-aux-Roses, France
| |
Collapse
|
1765
|
Diefenbach C, Caro J, Koide A, Grossbard M, Goldberg JD, Raphael B, Hymes K, Moskovits T, Kreditor M, Kaminetzky D, Fleur-Lominy SS, Choi J, Thannickal SA, Stapleford KA, Koide S. Impaired Humoral Immunity to SARS-CoV-2 Vaccination in Non-Hodgkin Lymphoma and CLL Patients. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34100025 DOI: 10.1101/2021.06.02.21257804] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Patients with hematologic malignancies are a high priority for SARS-CoV-2 vaccination, yet the benefit they will derive is uncertain. We investigated the humoral response to vaccination in 53 non-Hodgkin lymphoma (NHL), Hodgkin lymphoma (HL), or CLL patients. Peripheral blood was obtained 2 weeks after first vaccination and 6 weeks after second vaccination for antibody profiling using the multiplex bead-binding assay. Serum IgG, IgA, and IgM antibody levels to the spike specific receptor binding domain (RBD) were evaluated as a measure of response. Subsequently, antibody-positive serum were assayed for neutralization capacity against authentic SARS-CoV-2. Histology was 68% lymphoma and 32% CLL; groups were: patients receiving anti-CD20-based therapy (45%), monitored with disease (28%), receiving BTK inhibitors (19%), or chemotherapy (all HL) (8%). SARS-CoV-2 specific RBD IgG antibody response was decreased across all NHL and CLL groups: 25%, 73%, and 40%, respectively. Antibody IgG titers were significantly reduced (p < 0.001) for CD20 treated and targeted therapy patients, and (p = 0.003) for monitored patients. In 94% of patients evaluated after first and second vaccination, antibody titers did not significantly boost after second vaccination. Only 13% of CD20 treated and 13% of monitored patients generated neutralizing antibodies to SARS-CoV-2 with ICD50s 135 to 1767, and 445 and > 10240. This data has profound implications given the current guidance relaxing masking restrictions and for timing of vaccinations. Unless immunity is confirmed with laboratory testing, these patients should continue to mask, socially distance, and to avoid close contact with non-vaccinated individuals. Statement of Translational Relevance Non Hodgkin lymphoma (NHL) and Chronic Lymphocytic leukemia (CLL) patients who are treated with anti-CD20 antibody therapy, BTK inhibitor therapy, or who are monitored with active disease, have decreased antibody response to SARS-CoV-2 vaccination and decreased antibody titers compared to healthy controls. Antibody titers do not boost following second vaccination, and very few patients generate neutralizing antibodies against SARS-CoV-2. This data is of particular importance, given the recent guidance from the CDC that vaccinated patients no longer need to be masked indoors as well as outdoors. Patients with NHL or CLL who fall into these categories should not consider their immunity from vaccination to be assured. If infected with SARS-CoV-2, they should be a high priority for monoclonal antibody directed therapy. Unless immune response to vaccination is confirmed with laboratory testing, they should continue to mask, socially distance, and to avoid close contact with non-vaccinated individuals.
Collapse
|
1766
|
Comparison and Analysis of Neutralizing Antibody Levels in Serum after Inoculating with SARS-CoV-2, MERS-CoV, or SARS-CoV Vaccines in Humans. Vaccines (Basel) 2021; 9:vaccines9060588. [PMID: 34199384 PMCID: PMC8229804 DOI: 10.3390/vaccines9060588] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/24/2021] [Accepted: 05/28/2021] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Middle East respiratory syndrome coronavirus (MERS-CoV), and severe acute respiratory syndrome coronavirus (SARS-CoV) pose a great threat to humanity. Every pandemic involving these coronaviruses has seriously affected human health and economic development. Currently, there are no approved therapeutic drugs against their infections. Therefore, the development of vaccines is particularly important to combat these coronaviruses. In this review, we summarized and analyzed the progress of vaccines against SARS-CoV, MERS-CoV, and SARS-CoV-2, including inactivated vaccines, live attenuated vaccines, subunit vaccines, nucleic acid vaccines, and viral vector vaccines. In addition, we compared the levels of neutralizing antibodies in the serum of patients with these three kinds of coronaviruses at different stages, and their ability and effects against SARS-CoV-2, MERS-CoV, and SARS-CoV. This review provides useful information for vaccine evaluation and analysis.
Collapse
|
1767
|
Iftikhar H, Noor SMU, Masood M, Bashir K. Bell's Palsy After 24 Hours of mRNA-1273 SARS-CoV-2 Vaccine. Cureus 2021; 13:e15935. [PMID: 34336436 PMCID: PMC8312995 DOI: 10.7759/cureus.15935] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 02/06/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) has become the fastest-spreading pandemic of the 21st century. Various vaccines have been made available via emergency use authorization. Currently, two mRNA vaccines are being offered internationally, BNT162b2 and mRNA-1273. In randomized trials of these vaccines, the incidence of Bell's palsy in the vaccinated group does not statistically exceed the placebo group. The FDA recommends increased surveillance for Bell's palsy as a potential side effect with the administration of the vaccines among larger populations globally. There have been a few case reports of Bell's palsy associated with mRNA vaccines. Type I interferons have been proposed as the potential mechanism linking mRNA COVID-19 vaccines to Bell's palsy. Here, we report the case of a 36-year-old previously healthy patient who developed symptoms of Bell's palsy along with left-arm numbness, tingling, and subjective weakness masquerading as a subacute stroke after receiving the second dose of the mRNA-1273 vaccine. CT and MRI of the brain were unremarkable. He was discharged home with a diagnosis of Bell's palsy and improved on follow-up. mRNA COVID-19 vaccines may be considered a risk factor for Bell's palsy.
Collapse
Affiliation(s)
- Haris Iftikhar
- Emergency Medicine, Hamad Medical Corporation, Doha, QAT
| | | | - Maarij Masood
- Emergency Medicine, Hamad Medical Corporation, Doha, QAT
| | - Khalid Bashir
- Emergency Medicine, Hamad Medical Corporation, Doha, QAT
| |
Collapse
|
1768
|
Cromer D, Juno JA, Khoury D, Reynaldi A, Wheatley AK, Kent SJ, Davenport MP. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat Rev Immunol 2021; 21:395-404. [PMID: 33927374 PMCID: PMC8082486 DOI: 10.1038/s41577-021-00550-x] [Citation(s) in RCA: 186] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is central to long-term control of the current pandemic. Despite our rapidly advancing knowledge of immune memory to SARS-CoV-2, understanding how these responses translate into protection against reinfection at both the individual and population levels remains a major challenge. An ideal outcome following infection or after vaccination would be a highly protective and durable immunity that allows for the establishment of high levels of population immunity. However, current studies suggest a decay of neutralizing antibody responses in convalescent patients, and documented cases of SARS-CoV-2 reinfection are increasing. Understanding the dynamics of memory responses to SARS-CoV-2 and the mechanisms of immune control are crucial for the rational design and deployment of vaccines and for understanding the possible future trajectories of the pandemic. Here, we summarize our current understanding of immune responses to and immune control of SARS-CoV-2 and the implications for prevention of reinfection.
Collapse
Affiliation(s)
- Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| | | |
Collapse
|
1769
|
Caniels TG, Bontjer I, van der Straten K, Poniman M, Burger JA, Appelman B, Lavell AHA, Oomen M, Godeke GJ, Valle C, Mögling R, van Willigen HDG, Wynberg E, Schinkel M, van Vught LA, Guerra D, Snitselaar JL, Chaturbhuj DN, Martin IC, Moore JP, de Jong MD, Reusken C, Sikkens JJ, Bomers MK, de Bree GJ, van Gils MJ, Eggink D, Sanders RW. Emerging SARS-CoV-2 variants of concern evade humoral immune responses from infection and vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 34100023 DOI: 10.1101/2021.05.26.21257441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Emerging SARS-CoV-2 variants pose a threat to human immunity induced by natural infection and vaccination. We assessed the recognition of three variants of concern (B.1.1.7, B.1.351 and P.1) in cohorts of COVID-19 patients ranging in disease severity (n = 69) and recipients of the Pfizer/BioNTech vaccine (n = 50). Spike binding and neutralization against all three VOC was substantially reduced in the majority of samples, with the largest 4-7-fold reduction in neutralization being observed against B.1.351. While hospitalized COVID-19 patients and vaccinees maintained sufficient neutralizing titers against all three VOC, 39% of non-hospitalized patients did not neutralize B.1.351. Moreover, monoclonal neutralizing antibodies (NAbs) show sharp reductions in their binding kinetics and neutralizing potential to B.1.351 and P.1, but not to B.1.1.7. These data have implications for the degree to which pre-existing immunity can protect against subsequent infection with VOC and informs policy makers of susceptibility to globally circulating SARS-CoV-2 VOC.
Collapse
|
1770
|
Abstract
Initial optimism regarding the development of COVID-19 vaccines has been tempered by the emergence of new variants of SARS-CoV-2. Will vaccination be able to contain the pandemic? How can we best optimize the limited supplies of vaccines available and what should future COVID-19 vaccines look like? This Comment discusses how the emerging SARS-CoV-2 variants of concern could impact on the hopes of long-term pandemic control through vaccination and the mutations that might be relevant to the design of modified vaccines.
Collapse
Affiliation(s)
- Ravindra K Gupta
- Cambridge Institute of Therapeutic Immunology & Infectious Disease, University of Cambridge, Cambridge, UK.
| |
Collapse
|
1771
|
[Autoimmune diseases and vaccines against COVID-19. Decision making in uncertain scenarios]. Med Clin (Barc) 2021; 157:247-252. [PMID: 34140165 PMCID: PMC8162708 DOI: 10.1016/j.medcli.2021.05.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 02/02/2023]
|
1772
|
COVID-19 Vaccine Breakthrough Infections Reported to CDC - United States, January 1-April 30, 2021. MMWR-MORBIDITY AND MORTALITY WEEKLY REPORT 2021; 70:792-793. [PMID: 34043615 PMCID: PMC8158893 DOI: 10.15585/mmwr.mm7021e3] [Citation(s) in RCA: 271] [Impact Index Per Article: 67.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
1773
|
Lipsitch M, Kahn R. Interpreting vaccine efficacy trial results for infection and transmission. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.25.21252415. [PMID: 33655276 PMCID: PMC7924301 DOI: 10.1101/2021.02.25.21252415] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Randomized controlled trials (RCTs) have shown high efficacy of multiple vaccines against SARS-CoV-2 disease (COVID-19), and recent studies have shown the vaccines are also effective against infection. Evidence for the effect of each of these vaccines on ability to transmit the virus is also beginning to emerge. We describe an approach to estimate these vaccines' effects on viral positivity, a prevalence measure which under the reasonable assumption that vaccinated individuals who become infected are no more infectious than unvaccinated individuals forms a lower bound on efficacy against transmission. Specifically, we recommend separate analysis of positive tests triggered by symptoms (usually the primary outcome) and cross-sectional prevalence of positive tests obtained regardless of symptoms. The odds ratio of carriage for vaccine vs. placebo provides an unbiased estimate of vaccine effectiveness against viral positivity, under certain assumptions, and we show through simulations that likely departures from these assumptions will only modestly bias this estimate. Applying this approach to published data from the RCT of the Moderna vaccine, we estimate that one dose of vaccine reduces the potential for transmission by at least 61%, possibly considerably more. We describe how these approaches can be translated into observational studies of vaccine effectiveness.
Collapse
Affiliation(s)
- Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Rebecca Kahn
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| |
Collapse
|
1774
|
Thrombotic Thrombocytopenia after COVID-19 Vaccination: In Search of the Underlying Mechanism. Vaccines (Basel) 2021; 9:vaccines9060559. [PMID: 34071883 PMCID: PMC8227748 DOI: 10.3390/vaccines9060559] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
The rollout of COVID-19 vaccines brings hope for successful pandemic mitigation and getting the transmission of SARS-CoV-2 under control. The vaccines authorized in Europe displayed a good safety profile in the clinical trials. However, during their post-authorization use, unusual thrombotic events associated with thrombocytopenia have rarely been reported for vector vaccines. This led to the temporary suspension of the AZD1222 vaccine (Oxford/AstraZeneca) in various European countries and the Ad26.COV2 vaccine (Janssen/Johnson&Johnson) in the United States, with regulatory bodies launching investigations into potential causal associations. The thromboembolic reactions were also rarely reported after mRNA vaccines. The exact cause of these adverse effects remains to be elucidated. The present paper outlines the hypotheses on the mechanisms behind the very rare thrombotic thrombocytopenia reported after the COVID-19 vaccination, along with currently existing evidence and future research prospects. The following are discussed: (i) the role of antibodies against platelet factor 4 (PF4), (ii) the direct interaction between adenoviral vector and platelets, (iii) the cross-reactivity of antibodies against SARS-CoV-2 spike protein with PF4, (iv) cross-reactivity of anti-adenovirus antibodies and PF4, (v) interaction between spike protein and platelets, (vi) the platelet expression of spike protein and subsequent immune response, and (vii) the platelet expression of other adenoviral proteins and subsequent reactions. It is also plausible that thrombotic thrombocytopenia after the COVID-19 vaccine is multifactorial. The elucidation of the causes of these adverse events is pivotal in taking precautionary measures and managing vaccine hesitancy. It needs to be stressed, however, that the reported cases are currently sporadic and that the benefits of COVID-19 vaccines vastly outweigh their potential risks.
Collapse
|
1775
|
Corbett KS, Werner AP, O' Connell S, Gagne M, Lai L, Moliva JI, Flynn B, Choi A, Koch M, Foulds KE, Andrew SF, Flebbe DR, Lamb E, Nurmukhambetova ST, Provost SJ, Bock KW, Minai M, Nagata BM, Van Ry A, Flinchbaugh Z, Johnston TS, Mokhtari EB, Mudvari P, Henry AR, Laboune F, Chang B, Porto M, Wear J, Alvarado GS, Boyoglu-Barnum S, Todd JPM, Bart B, Cook A, Dodson A, Pessaint L, Steingrebe K, Elbashir S, Andersen H, Wu K, Edwards DK, Kar S, Lewis MG, Bortiz E, Moore IN, Carfi A, Suthar MS, McDermott A, Roederer M, Nason MC, Sullivan NJ, Douek DC, Graham BS, Seder RA. Evaluation of mRNA-1273 against SARS-CoV-2 B.1.351 Infection in Nonhuman Primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 34075375 DOI: 10.1101/2021.05.21.445189] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Background Vaccine efficacy against the B.1.351 variant following mRNA-1273 vaccination in humans has not been determined. Nonhuman primates (NHP) are a useful model for demonstrating whether mRNA-1273 mediates protection against B.1.351. Methods Nonhuman primates received 30 or 100 µg of mRNA-1273 as a prime-boost vaccine at 0 and 4 weeks, a single immunization of 30 µg at week 0, or no vaccine. Antibody and T cell responses were assessed in blood, bronchioalveolar lavages (BAL), and nasal washes. Viral replication in BAL and nasal swabs were determined by qRT-PCR for sgRNA, and histopathology and viral antigen quantification were performed on lung tissue post-challenge. Results Eight weeks post-boost, 100 µg x2 of mRNA-1273 induced reciprocal ID 50 neutralizing geometric mean titers against live SARS-CoV-2 D614G and B.1.351 of 3300 and 240, respectively, and 430 and 84 for the 30 µg x2 group. There were no detectable neutralizing antibodies against B.1351 after the single immunization of 30 µg. On day 2 following B.1.351 challenge, sgRNA in BAL was undetectable in 6 of 8 NHP that received 100 µg x2 of mRNA-1273, and there was a ∼2-log reduction in sgRNA in NHP that received two doses of 30 µg compared to controls. In nasal swabs, there was a 1-log 10 reduction observed in the 100 µg x2 group. There was limited inflammation or viral antigen in lungs of vaccinated NHP post-challenge. Conclusions Immunization with two doses of mRNA-1273 achieves effective immunity that rapidly controls lower and upper airway viral replication against the B.1.351 variant in NHP.
Collapse
|
1776
|
|
1777
|
Park JH, Lee HK. Delivery Routes for COVID-19 Vaccines. Vaccines (Basel) 2021; 9:524. [PMID: 34069359 PMCID: PMC8158705 DOI: 10.3390/vaccines9050524] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
The novel coronavirus, SARS-CoV-2, which causes COVID-19, has resulted in a pandemic with millions of deaths. To eradicate SARS-CoV-2 and prevent further infections, many vaccine candidates have been developed. These vaccines include not only traditional subunit vaccines and attenuated or inactivated viral vaccines but also nucleic acid and viral vector vaccines. In contrast to the diversity in the platform technology, the delivery of vaccines is limited to intramuscular vaccination. Although intramuscular vaccination is safe and effective, mucosal vaccination could improve the local immune responses that block the spread of pathogens. However, a lack of understanding of mucosal immunity combined with the urgent need for a COVID-19 vaccine has resulted in only intramuscular vaccinations. In this review, we summarize the history of vaccines, current progress in COVID-19 vaccine technology, and the status of intranasal COVID-19 vaccines. Future research should determine the most effective route for vaccine delivery based on the platform and determine the mechanisms that underlie the efficacy of different delivery routes.
Collapse
Affiliation(s)
| | - Heung Kyu Lee
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea;
| |
Collapse
|
1778
|
Muena NA, García-Salum T, Pardo-Roa C, Serrano EF, Levican J, Avendaño MJ, Almonacid LI, Valenzuela G, Poblete E, Strohmeier S, Salinas E, Haslwanter D, Dieterle ME, Jangra RK, Chandran K, González C, Riquelme A, Krammer F, Tischler ND, Medina RA. Long-lasting neutralizing antibody responses in SARS-CoV-2 seropositive individuals are robustly boosted by immunization with the CoronaVac and BNT162b2 vaccines. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.05.17.21257197. [PMID: 34031662 PMCID: PMC8142662 DOI: 10.1101/2021.05.17.21257197] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The durability of circulating neutralizing antibody (nAb) responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and their boosting by vaccination remains to be defined. We show that outpatient and hospitalized SARS-CoV-2 seropositive individuals mount a robust neutralizing antibody (nAb) response that peaks at days 23 and 27 post-symptom onset, respectively. Although nAb titers remained higher in hospitalized patients, both study groups showed long-lasting nAb responses that can persist for up to 12 months after natural infection. These nAb responses in previously seropositive individuals can be significantly boosted through immunization with two doses of the CoronaVac (Sinovac) or one dose of the BNT162b2 (BioNTech/Pfizer) vaccines, suggesting a substantial induction of B cell memory responses. Noteworthy, three obese previously seropositive individuals failed to mount a booster response upon vaccination, warranting further studies in this population. Immunization of naïve individuals with two doses of the CoronaVac vaccine or one dose of the BNT162b2 vaccine elicited similar levels of nAbs compared to seropositive individuals 4.2 to 13.3 months post-infection with SARS-CoV-2. Thus, this preliminary evidence suggests that both, seropositive and naïve individuals, require two doses of CoronaVac to ensure the induction of robust nAb titers.
Collapse
Affiliation(s)
- Nicolás A. Muena
- Laboratorio de Virología Molecular, Fundación Ciencia & Vida, Av. Zañartu 1482, Santiago, Chile
| | - Tamara García-Salum
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Pardo-Roa
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eileen F. Serrano
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- PhD Program in Biological Sciences, Mention in Molecular Genetics and Microbiology, Faculty of Biological Science, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge Levican
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María José Avendaño
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- PhD Program in Biological Sciences, Mention in Molecular Genetics and Microbiology, Faculty of Biological Science, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Leonardo I. Almonacid
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo Valenzuela
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Estefany Poblete
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Shirin Strohmeier
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Erick Salinas
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Denise Haslwanter
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Maria Eugenia Dieterle
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Rohit K. Jangra
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY, USA
| | - Claudia González
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Otorhinolaryngology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Arnoldo Riquelme
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Health Sciences, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Florian Krammer
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole D. Tischler
- Laboratorio de Virología Molecular, Fundación Ciencia & Vida, Av. Zañartu 1482, Santiago, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Rafael A. Medina
- Department of Pediatric Infectious Diseases and Immunology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Advanced Interdisciplinary Rehabilitation Register (AIRR) – COVID-19 Working Group, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
1779
|
Hazlewood GS, Pardo JP, Barnabe C, Schieir O, Barber CEH, Bernatsky S, Colmegna I, Hitchon C, Loeb M, Mertz D, Proulx L, Richards DP, Scuccimarri R, Tugwell P, Schünemann HJ, Mirza RD, Zhou AL, Nikolic RPA, Thomas M, Chase H, Ejaredar M, Nieuwlaat R. Canadian Rheumatology Association Recommendation for the Use of COVID-19 Vaccination for Patients With Autoimmune Rheumatic Diseases. J Rheumatol 2021; 48:1330-1339. [PMID: 33993119 DOI: 10.3899/jrheum.210288] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVE To develop guidance on the use of coronavirus disease 2019 (COVID-19) vaccines in patients with autoimmune rheumatic diseases (ARD). METHODS The Canadian Rheumatology Association (CRA) formed a multidisciplinary panel including rheumatologists, researchers, methodologists, vaccine experts, and patients. The panel used the GRADE (Grading of Recommendations Assessment, Development, and Evaluation) approach. Outcomes were prioritized according to their importance for patients and clinicians. Evidence from the COVID-19 clinical trials was summarized. Indirect evidence for non-COVID-19 vaccines in ARD was also considered. The GRADE evidence-to-decision (EtD) framework was used to develop a recommendation for the use of the 4 COVID-19 vaccines approved in Canada as of March 25, 2021 (BNT162b2, mRNA-1273, ChAdOx1, and Ad26.COV2.S), over 4 virtual panel meetings. RESULTS The CRA guideline panel suggests using COVID-19 vaccination in persons with ARD. The panel unanimously agreed that for the majority of patients, the potential health benefits of vaccination outweigh the potential harms in people with ARDs. The recommendation was graded as conditional because of low or very low certainty of the evidence on the effects in the population of interest, primarily due to indirectness and imprecise effect estimates. The panel felt strongly that persons with autoimmune rheumatic diseases who meet local eligibility should not be required to take additional steps compared to people without ARDs to obtain their vaccination. Guidance on medications, implementation, monitoring of vaccine uptake, and research priorities are also provided. CONCLUSION This recommendation will be updated over time as new evidence emerges, with the latest recommendation, evidence summaries, and EtD available on the CRA website.
Collapse
Affiliation(s)
- Glen S Hazlewood
- G.S. Hazlewood, MD, PhD, Associate Professor of Medicine, C. Barnabe, MD, MSc, Associate Professor of Medicine, C.E. Barber, MD, PhD, Assistant Professor of Medicine, Departments of Medicine and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, and Arthritis Research Canada, Richmond, British Columbia;
| | - Jordi Pardo Pardo
- J.P. Pardo, LDO, Managing Editor, Centre for Global Health, University of Ottawa, Ottawa, Ontario
| | - Cheryl Barnabe
- G.S. Hazlewood, MD, PhD, Associate Professor of Medicine, C. Barnabe, MD, MSc, Associate Professor of Medicine, C.E. Barber, MD, PhD, Assistant Professor of Medicine, Departments of Medicine and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, and Arthritis Research Canada, Richmond, British Columbia
| | - Orit Schieir
- O. Schieir, PhD, Canadian Early Arthritis Cohort Study, Toronto, Ontario
| | - Claire E H Barber
- G.S. Hazlewood, MD, PhD, Associate Professor of Medicine, C. Barnabe, MD, MSc, Associate Professor of Medicine, C.E. Barber, MD, PhD, Assistant Professor of Medicine, Departments of Medicine and Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, and Arthritis Research Canada, Richmond, British Columbia
| | - Sasha Bernatsky
- S. Bernatsky, MD, PhD, Professor of Medicine, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec
| | - Ines Colmegna
- I. Colmegna, MD, Associate Professor of Medicine, Division of Rheumatology, Department of Medicine, McGill University, Montreal, Quebec
| | - Carol Hitchon
- C. Hitchon, MD, MSc, Associate Professor of Medicine, Department of Internal Medicine, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba
| | - Mark Loeb
- M. Loeb, MD, MSc, Professor, D. Mertz, MD, MSc, Associate Professor of Medicine, H.J. Schünemann, MD, MSc, PhD, Professor of Medicine and Clinical Epidemiology, Departments of Medicine and Health Research Methods, Evidence, and Impact, McMaster GRADE Centers, Hamilton
| | - Dominik Mertz
- M. Loeb, MD, MSc, Professor, D. Mertz, MD, MSc, Associate Professor of Medicine, H.J. Schünemann, MD, MSc, PhD, Professor of Medicine and Clinical Epidemiology, Departments of Medicine and Health Research Methods, Evidence, and Impact, McMaster GRADE Centers, Hamilton
| | - Laurie Proulx
- L. Proulx, B.Com, D.P. Richards, PhD, Canadian Arthritis Patient Alliance, Toronto, Ontario
| | - Dawn P Richards
- L. Proulx, B.Com, D.P. Richards, PhD, Canadian Arthritis Patient Alliance, Toronto, Ontario
| | - Rosie Scuccimarri
- R. Scuccimarri, MD, Associate Professor of Pediatrics, Division of Pediatric Rheumatology, Department of Pediatrics, McGill University, Montreal, Quebec
| | - Peter Tugwell
- P. Tugwell, MD, Professor of Medicine, A.L. Zhou, MD, Department of Medicine, University of Ottawa, Ottawa, Ontario
| | - Holger J Schünemann
- M. Loeb, MD, MSc, Professor, D. Mertz, MD, MSc, Associate Professor of Medicine, H.J. Schünemann, MD, MSc, PhD, Professor of Medicine and Clinical Epidemiology, Departments of Medicine and Health Research Methods, Evidence, and Impact, McMaster GRADE Centers, Hamilton
| | - Reza D Mirza
- R.D. Mirza, MD, Department of Medicine, University of Toronto, Toronto, Ontario
| | - Alan L Zhou
- P. Tugwell, MD, Professor of Medicine, A.L. Zhou, MD, Department of Medicine, University of Ottawa, Ottawa, Ontario
| | - Roko P A Nikolic
- R.P. Nikolic, BSc, Cumming School of Medicine, University of Calgary, Calgary, Alberta
| | - Megan Thomas
- M. Thomas, BHSc, M. Ejaredar, PhD, Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta
| | | | - Maede Ejaredar
- M. Thomas, BHSc, M. Ejaredar, PhD, Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta
| | - Robby Nieuwlaat
- R. Nieuwlaat, MSc, PhD, Associate Professor, Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
1780
|
Abstract
Roughly 1 year after the first case of COVID-19 was identified and less than 1 year after the sequencing of SARS-CoV-2, multiple SARS-CoV-2 vaccines with demonstrated safety and efficacy in phase III clinical trials are available. The most promising vaccines have targeted the surface glycoprotein (S-protein) of SARS-CoV-2 and achieved an approximate 85%-95% reduction in the risk of symptomatic COVID-19, while retaining excellent safety profiles and modest side effects in the phase III clinical trials. The mRNA, replication-incompetent viral vector, and protein subunit vaccine technologies have all been successfully employed. Some novel SARS-CoV-2 variants evade but do not appear to fully overcome the potent immunity induced by these vaccines. Emerging real-world effectiveness data add evidence for protection from severe COVID-19. This is an impressive first demonstration of the effectiveness of the mRNA vaccine and vector vaccine platforms. The success of SARS-CoV-2 vaccine development should be credited to open science, industry partnerships, harmonization of clinical trials, and the altruism of study participants. The manufacturing and distribution of the emergency use-authorized SARS-CoV-2 vaccines are ongoing challenges. What remains now is to ensure broad and equitable global vaccination against COVID-19.
Collapse
Affiliation(s)
| | - Njira Lugogo
- Division of Pulmonary and Critical Care Medicine, and
| | | | - Anna S. Lok
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
1781
|
Shay DK, Gee J, Su JR, Myers TR, Marquez P, Liu R, Zhang B, Licata C, Clark TA, Shimabukuro TT. Safety Monitoring of the Janssen (Johnson & Johnson) COVID-19 Vaccine - United States, March-April 2021. MMWR-MORBIDITY AND MORTALITY WEEKLY REPORT 2021; 70:680-684. [PMID: 33956784 PMCID: PMC9368748 DOI: 10.15585/mmwr.mm7018e2] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
1782
|
Gleinich AS, Pepi LE, Shajahan A, Heiss C, Azadi P. Vaccines and Therapeutics for COVID-19 - How Can Understanding SARS-CoV-2 Glycosylation Lead to Pharmaceutical Advances? AMERICAN PHARMACEUTICAL REVIEW 2021; 24:14-21. [PMID: 38099300 PMCID: PMC10721230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/17/2023]
Abstract
The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected more than 160 million people worldwide. Researchers have targeted the SARS-CoV-2 structural proteins to better combat the pandemic. Of the four structural proteins, spike (S), membrane (M), envelope (E) and nucleocapsid (N), the S, M and E proteins are glycosylated whereas the N protein is phosphorylated. The glycosylation of the S protein has been reported previously by multiple research groups, and this knowledge has assisted the pharmaceutical industry in developing vaccines and treatment options. In the United States, there are currently three approved COVID-19 vaccines. All three of these vaccines use the S protein to teach host cells how to react when SARS-CoV-2 particles are present. Treatment options utilizing antivirals and immunosuppressants are being developed in addition to vaccines. Different treatment approaches are needed based on the severity of COVID-19 infection. The therapeutic options currently available are not derived through the direct knowledge on SARS-CoV-2 glycosylation. However, more research on the glycosylation of the structural proteins and how this effects SARS-CoV-2 and host cell binding could lead to new and more effective therapeutics. Herein we outline the current vaccine and therapeutic options against COVID-19 available to the public, as well as those still in development.
Collapse
Affiliation(s)
- Anne S Gleinich
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Lauren E Pepi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Asif Shajahan
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Christian Heiss
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| | - Parastoo Azadi
- Complex Carbohydrate Research Center, University of Georgia, Athens, GA
| |
Collapse
|
1783
|
Gray GE, Corey L. The path to find an HIV vaccine. J Int AIDS Soc 2021; 24:e25749. [PMID: 34002949 PMCID: PMC8130233 DOI: 10.1002/jia2.25749] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 11/11/2022] Open
Affiliation(s)
- Glenda E Gray
- South African Medical Research CouncilCape TownSouth Africa
| | - Lawrence Corey
- Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleUSA
| |
Collapse
|
1784
|
Leier HC, Bates TA, Lyski ZL, McBride SK, Lee DX, Coulter FJ, Goodman JR, Lu Z, Curlin ME, Messer WB, Tafesse FG. Previously infected vaccinees broadly neutralize SARS-CoV-2 variants. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.04.25.21256049. [PMID: 33948601 PMCID: PMC8095208 DOI: 10.1101/2021.04.25.21256049] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
We compared the serum neutralizing antibody titers before and after two doses of the BNT162b2 COVID-19 vaccine in ten individuals who recovered from SARS-CoV-2 infection prior to vaccination to 20 individuals with no history of infection, against clinical isolates of B.1.1.7, B.1.351, P.1, and the original SARS-CoV-2 virus. Vaccination boosted pre-existing levels of anti-SARS-CoV-2 spike antibodies 10-fold in previously infected individuals, but not to levels significantly higher than those of uninfected vaccinees. However, neutralizing antibody titers increased in previously infected vaccinees relative to uninfected vaccinees against every variant tested: 5.2-fold against B.1.1.7, 6.5-fold against B.1.351, 4.3-fold against P.1, and 3.4-fold against original SARS-CoV-2. Our study indicates that a first-generation COVID-19 vaccine provides broad protection from SARS-CoV-2 variants in individuals with previous infection.
Collapse
Affiliation(s)
- Hans C. Leier
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - Timothy A. Bates
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - Zoe L. Lyski
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - Savannah K. McBride
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - David X. Lee
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - Felicity J. Coulter
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - James R. Goodman
- Medical Scientist Training Program, Oregon Health & Science University; Portland, OR 97239, United States
| | - Zhengchun Lu
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| | - Marcel E. Curlin
- Division of Infectious Diseases, Oregon Health & Science University; Portland, OR 97239, United States
| | - William B. Messer
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
- Division of Infectious Diseases, Oregon Health & Science University; Portland, OR 97239, United States
| | - Fikadu G. Tafesse
- Department of Molecular Microbiology & Immunology, Oregon Health & Science University; Portland, OR 97239, United States
| |
Collapse
|
1785
|
Martinez DR, Schaefer A, Gobeil S, Li D, De la Cruz G, Parks R, Lu X, Barr M, Manne K, Mansouri K, Edwards RJ, Yount B, Anasti K, Montgomery SA, Shen S, Zhou T, Kwong PD, Graham BS, Mascola JR, Montefiori DC, Alam M, Sempowski GD, Wiehe K, Saunders KO, Acharya P, Haynes BF, Baric RS. A broadly neutralizing antibody protects against SARS-CoV, pre-emergent bat CoVs, and SARS-CoV-2 variants in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.04.27.441655. [PMID: 33948590 PMCID: PMC8095197 DOI: 10.1101/2021.04.27.441655] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SARS-CoV in 2003, SARS-CoV-2 in 2019, and SARS-CoV-2 variants of concern (VOC) can cause deadly infections, underlining the importance of developing broadly effective countermeasures against Group 2B Sarbecoviruses, which could be key in the rapid prevention and mitigation of future zoonotic events. Here, we demonstrate the neutralization of SARS-CoV, bat CoVs WIV-1 and RsSHC014, and SARS-CoV-2 variants D614G, B.1.1.7, B.1.429, B1.351 by a receptor-binding domain (RBD)-specific antibody DH1047. Prophylactic and therapeutic treatment with DH1047 demonstrated protection against SARS-CoV, WIV-1, RsSHC014, and SARS-CoV-2 B1.351infection in mice. Binding and structural analysis showed high affinity binding of DH1047 to an epitope that is highly conserved among Sarbecoviruses. We conclude that DH1047 is a broadly neutralizing and protective antibody that can prevent infection and mitigate outbreaks caused by SARS-like strains and SARS-CoV-2 variants. Our results argue that the RBD conserved epitope bound by DH1047 is a rational target for pan Group 2B coronavirus vaccines.
Collapse
Affiliation(s)
- David R. Martinez
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- These authors contributed equally
| | - Alexandra Schaefer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- These authors contributed equally
| | - Sophie Gobeil
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- These authors contributed equally
| | - Dapeng Li
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- These authors contributed equally
| | - Gabriela De la Cruz
- Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Robert Parks
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Xiaozhi Lu
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Maggie Barr
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kartik Manne
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J. Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Boyd Yount
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kara Anasti
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Stephanie A. Montgomery
- Department of Laboratory Medicine and Pathology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Shaunna Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Tongqing Zhou
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | | | - Munir Alam
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Gregory D. Sempowski
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kevin O. Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Priyamvada Acharya
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Barton F. Haynes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
1786
|
Wu K, Choi A, Koch M, Elbashir S, Ma L, Lee D, Woods A, Henry C, Palandjian C, Hill A, Quinones J, Nunna N, O'Connell S, McDermott AB, Falcone S, Narayanan E, Colpitts T, Bennett H, Corbett KS, Seder R, Graham BS, Stewart-Jones GB, Carfi A, Edwards DK. Variant SARS-CoV-2 mRNA vaccines confer broad neutralization as primary or booster series in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021. [PMID: 33880468 DOI: 10.1101/2021.04.13.439482] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of a global pandemic that has led to more than 2.8 million deaths worldwide. Safe and effective vaccines are now available, including Moderna's COVID-19 vaccine (mRNA-1273) that showed 94% efficacy in prevention of symptomatic COVID-19 disease in a phase 3 clinical study. mRNA-1273 encodes for a prefusion stabilized full length spike (S) protein of the Wuhan-Hu-1 isolate. However, the emergence of SARS-CoV-2 variants has led to concerns of viral escape from vaccine-induced immunity. Several emerging variants have shown decreased susceptibility to neutralization by vaccine induced immunity, most notably the B.1.351 variant, although the overall impact on vaccine efficacy remains to be determined. Here, we present the initial evaluation in mice of two updated COVID-19 mRNA vaccines designed to target emerging SARS-CoV-2 variants: (1) monovalent mRNA-1273.351 encodes for the S protein found in the B.1.351 lineage and (2) mRNA-1273.211 comprising a 1:1 mix of mRNA-1273 and mRNA-1273.351. Both vaccines were evaluated as a 2-dose primary series in mice; mRNA-1273.351 was also evaluated as a booster dose in animals previously vaccinated with 2-doses of mRNA-1273. The results demonstrated that a primary vaccination series of mRNA-1273.351 was effective at increasing neutralizing antibody titers against the B.1.351 lineage, while mRNA-1273.211 was most effective at providing broad cross-variant neutralization in mice. In addition, these results demonstrated a third dose of mRNA-1273.351 significantly increased both wild-type and B.1.351-specific neutralization titers. Both mRNA-1273.351 and mRNA-1273.211 are currently being evaluated in additional pre-clinical challenge models and in phase 1/2 clinical studies.
Collapse
|
1787
|
Johansen K, Nohynek H. No country or continent is on its own in the ongoing COVID-19 pandemic. Euro Surveill 2021; 26:2100430. [PMID: 33928901 PMCID: PMC8086248 DOI: 10.2807/1560-7917.es.2021.26.17.2100430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 04/29/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Hanna Nohynek
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| |
Collapse
|
1788
|
Goldschmidt-Clermont PJ, Goldschmidt AJP, Weiss RE. Rivalry between human ideation and virus mutation: two competing means of sustainability. WOMEN HEALTH CARE AND ISSUES 2021; 4:058. [PMID: 34527953 PMCID: PMC8439168 DOI: 10.31579/2642-9756/058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
For the first time in human history, obtaining a COVID-19 vaccine has become essential for the sustainability of our species. As an amazing product of collective ideation, remarkably safe and efficient vaccines have been invented, tested, distributed, and administered to the population on a voluntary basis. The fast-mutating individual behavior of the virus is probably guided by a similar goal of the sustainability of the species. With this commentary, we analyze and compare two means of sustainability through adaptability: collective ideation in the case of humans and individual mutations in the case of viruses - two very different species whose behaviors are driven by sustainability.
Collapse
Affiliation(s)
- Pascal J Goldschmidt-Clermont
- Alzady International LLC, Dean Emeritus, Professor of Medicine Emeritus, Miller School of Medicine University of Miami, Miami, Florida 33136
| | | | - Roy E Weiss
- University of Miami Leonard M. Miller School of Medicine, Department of Medicine Chairman, Miami, USA
| |
Collapse
|
1789
|
Abstract
The remarkable development of a large number of vaccines against COVID-19 in a very short period of time represents one of the greatest successes of medicine and science in history, and mass vaccination at the global level will be crucial for prevention and mitigation of COVID-19. However, there are still numerous open questions about the vaccines, and the protection they provide, and answers to those questions will not only help to control this pandemic, but they will also prepare us to react better in case of future outbreaks. This review will present the latest findings on the immune response to SARS-CoV-2 and give an update on COVID-19 immunity. It will also provide an overview of the most important vaccines against COVID-19, especially those available in Serbia, with an emphasis on their immunogenicity, efficacy and safety, as well as the platforms used for their development. In addition, a special attention will be given to open issues related to immunization against COVID-19, such as the duration of post-vaccination immunity, the degree of protection against new virus variants and the need for booster doses and mixing and matching of different COVID-19 vaccines.
Collapse
|
1790
|
Hasanpourghadi M, Novikov M, Ambrose R, Chekaoui A, Newman D, Zhou XY, Ertl HCJ. T cell responses to adenoviral vectors expressing the SARS-CoV-2 nucleoprotein. CURRENT TRENDS IN MICROBIOLOGY 2021; 15:1-28. [PMID: 35903088 PMCID: PMC9328080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
SARS-CoV-2 vaccines aim to protect against COVID-19 through neutralizing antibodies against the viral spike protein. Mutations within the spike's receptor-binding domain may eventually reduce vaccine efficacy, necessitating periodic updates. Vaccine-induced immunity could be broadened by adding T cell-inducing antigens such as SARS-CoV-2's nucleoprotein (N). Here we describe two replication-defective chimpanzee adenovirus (AdC) vectors from different serotypes expressing SARS-CoV-2 N either in its wild-type form or fused into herpes simplex virus glycoprotein D (gD), an inhibitor of an early T cell checkpoint. The vaccines induce potent and sustained CD8+ T cell responses that are broadened upon inclusion of gD. Depending on the vaccine regimen booster immunizations increase magnitude and breadth of T cell responses. Epitopes that are recognized by the vaccine-induced T cells are highly conserved among global SARS-CoV-2 isolates indicating that addition of N to COVID-19 vaccines may lessen the risk of loss of vaccine-induced protection due to variants.
Collapse
Affiliation(s)
| | - Mikhail Novikov
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Robert Ambrose
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Arezki Chekaoui
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Dakota Newman
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Xiang Yang Zhou
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| | - Hildegund C. J. Ertl
- The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104, USA, 1-215-898-3863
| |
Collapse
|
1791
|
Peng F, Yuan H, Wu S, Zhou Y. Recent Advances on Drugs and Vaccines for COVID-19. INQUIRY : A JOURNAL OF MEDICAL CARE ORGANIZATION, PROVISION AND FINANCING 2021; 58:469580211055630. [PMID: 34818922 PMCID: PMC8673875 DOI: 10.1177/00469580211055630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The current situation of Coronavirus Disease 2019 (COVID-19) worldwide is still very severe. Presently, many breakthroughs have been accomplished in the research and development of drugs for the treatment of COVID-19, especially vaccines; however, some of the so-called COVID-19-specific drugs highlighted in the early stage failed to achieve the expected curative effect. There is no antiviral therapy available, by stimulating protective immunity vaccine is the best choice for the future management of infection. Therefore, we aimed to identify the latest developments in the research and development of these drugs and vaccines and provide a reference for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Fang Peng
- 87803The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Hao Yuan
- 87803The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Sixian Wu
- 87803The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| | - Yifeng Zhou
- 87803The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan Province, China
| |
Collapse
|
1792
|
Gaunt ER, Mabbott NA. The clinical correlates of vaccine-induced immune thrombotic thrombocytopenia after immunisation with adenovirus vector-based SARS-CoV-2 vaccines. IMMUNOTHERAPY ADVANCES 2021; 1:ltab019. [PMID: 34557868 PMCID: PMC8385946 DOI: 10.1093/immadv/ltab019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/04/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022] Open
Abstract
We are at a critical stage in the COVID-19 pandemic where vaccinations are being rolled out globally, in a race against time to get ahead of the SARS-CoV-2 coronavirus and the emergence of more highly transmissible variants. A range of vaccines have been created and received either emergency approval or full licensure. To attain the upper hand, maximum vaccine synthesis, deployment, and uptake as rapidly as possible is essential. However, vaccine uptake, particularly in younger adults is dropping, at least in part fuelled by reports of rare complications associated with specific vaccines. This review considers how vaccination with adenovirus vector-based vaccines against the SARS-CoV-2 coronavirus might cause rare cases of thrombosis and thrombocytopenia in some recipients. A thorough understanding of the underlying cellular and molecular mechanisms that mediate this syndrome may help to identify methods to prevent these very rare, but serious side effects. This will also help facilitate the identification of those at highest risk from these outcomes, so that we can work towards a stratified approach to vaccine deployment to mitigate these risks.
Collapse
Affiliation(s)
- Eleanor R Gaunt
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Neil A Mabbott
- The Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| |
Collapse
|
1793
|
Kantarcioglu B, Iqbal O, Walenga JM, Lewis B, Lewis J, Carter CA, Singh M, Lievano F, Tafur A, Ramacciotti E, Gerotziafas GT, Jeske W, Fareed J. An Update on the Pathogenesis of COVID-19 and the Reportedly Rare Thrombotic Events Following Vaccination. Clin Appl Thromb Hemost 2021; 27:10760296211021498. [PMID: 34060379 PMCID: PMC8173993 DOI: 10.1177/10760296211021498] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Today the coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has become a global health problem. After more than a year with the pandemic, although our knowledge has progressed on COVID-19, there are still many unknowns in virological, pathophysiological and immunological aspects. It is obvious that the most efficient solution to end this pandemic are safe and efficient vaccines. This manuscript summarizes the pathophysiological and thrombotic features of COVID-19 and the safety and efficacy of currently approved COVID-19 vaccines with an aim to clarify the recent concerns of thromboembolic events after COVID-19 vaccination. The influx of newer information is rapid, requiring periodic updates and objective assessment of the data on the pathogenesis of COVID-19 variants and the safety and efficacy of currently available vaccines.
Collapse
Affiliation(s)
- Bulent Kantarcioglu
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Omer Iqbal
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Jeanine M. Walenga
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Bruce Lewis
- Department of Medicine, Cardiology, Loyola University Medical Center, Maywood, IL, USA
| | - Joseph Lewis
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Charles A. Carter
- Department of Clinical Research, Campbell University College of Pharmacy and Health Sciences, Campbell University, Buies Creek, NC, USA
| | - Meharvan Singh
- Department of Cellular and Molecular Physiology, Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Fabio Lievano
- Department of Medical Safety Evaluation, AbbVie Inc., North Chicago, IL, USA
| | - Alfonso Tafur
- Section of Interventional Cardiology and Vascular Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Eduardo Ramacciotti
- Hemostasis & Thrombosis Research Laboratories at Loyola University Medical Center, Maywood, IL, USA
| | - Grigoris T. Gerotziafas
- 5-Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Thrombosis Center, Service D’Hématologie Biologique Hôpital Tenon, Paris, France
| | - Walter Jeske
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| | - Jawed Fareed
- Department of Pathology and Laboratory Medicine, Cardiovascular Research Institute, Loyola University Chicago, Health Sciences Division, Maywood, IL, USA
| |
Collapse
|
1794
|
Perera PY, Perera LP. Development of leading first-generation vaccines against SARS-CoV-2. Microbes Infect 2021; 23:104841. [PMID: 34022375 PMCID: PMC8133830 DOI: 10.1016/j.micinf.2021.104841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 01/29/2023]
Abstract
SARS-CoV-2 has infected more than 167 million individuals globally. Highly effective and safe vaccines are required to accelerate the development of herd immunity to end the pandemic. This review focuses on vaccines that are being developed at unprecedented speed globally and are completing late phase clinical trials to meet this urgent need.
Collapse
Affiliation(s)
- Pin-Yu Perera
- Pathology and Laboratory Medicine, Veterans Affairs Medical Center, Washington, DC, USA.
| | - Liyanage P Perera
- Lymphoid Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
1795
|
[Development, rollout, and challenges of COVID-19 vaccines in Japan and globally]. Uirusu 2021; 71:41-44. [PMID: 35526993 DOI: 10.2222/jsv.71.41] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
1796
|
Tramontana F, Battisti S, Napoli N, Strollo R. Immuno-Endocrinology of COVID-19: The Key Role of Sex Hormones. Front Endocrinol (Lausanne) 2021; 12:726696. [PMID: 34925228 PMCID: PMC8675353 DOI: 10.3389/fendo.2021.726696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 11/12/2021] [Indexed: 01/08/2023] Open
Abstract
Epidemiological evidence shows clear gender disparities in the Coronavirus 2019 Disease (COVID-19) severity and fatality. This may reflect the contribution of gender-related factors, such as sex hormones, to COVID-19 pathogenesis. However, the mechanism linking gender disparities to COVID-19 severity is still poorly understood. In this review, we will pinpoint several elements involved in COVID-19 pathogenesis that are regulated by the two main sex hormones, estrogen and androgen. These include tissue specific gene regulation of SARS-CoV2 entry factors, innate and adaptive immune responses to infection, immunometabolism, and susceptibility to tissue injury by cytopathic effect or hyper-inflammatory response. We will discuss the mechanistic link between sex hormone regulation of COVID-19 pathogenetic factors and disease severity. Finally, we will summarize current evidence from clinical studies and trials targeting sex hormones and their signalling in COVID-19. A better understanding of the role of sex hormones in COVID-19 may identify targets for therapeutic intervention and allow optimization of treatment outcomes towards gender-based personalised medicine.
Collapse
Affiliation(s)
- Flavia Tramontana
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Sofia Battisti
- Radiology Department, Azienda Unità Sanitaria Locale (AUSL) Romagna M. Bufalini Hospital, Cesena, Italy
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Meldola, Italy
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale (DIMES), Alma Mater Studiorum-Universita di Bologna, Bologna, Italy
| | - Nicola Napoli
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Rocky Strollo
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
- *Correspondence: Rocky Strollo,
| |
Collapse
|
1797
|
Vaccines. SIDE EFFECTS OF DRUGS ANNUAL 2021. [PMCID: PMC8488686 DOI: 10.1016/bs.seda.2021.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In this volume of the Side Effects of Drugs Annual, although other vaccines will be covered, the safety of COVID vaccines is the focus as COVID-19 has led to heightened attention on vaccine safety in general. As such, this chapter will be more relevant than ever before. As noted in past SEDA issues, clinicians should be mindful of the risks of AEs and SAEs associated with each vaccine.
Collapse
|